scholarly journals NCOA4 is Regulated by HIF and Mediates Mobilization of Murine Hepatic Iron Stores After Blood Loss

Blood ◽  
2020 ◽  
Author(s):  
Xiuqi Li ◽  
Larisa Lozovatsky ◽  
Abitha Sukumaran ◽  
Luis Gonzalez ◽  
Anisha Jain ◽  
...  

The mechanisms by which phlebotomy promotes the mobilization of hepatic iron stores are not well understood. NCOA4 (nuclear receptor coactivator 4) is a widely-expressed intracellular protein previously shown to mediate the autophagic degradation of ferritin. Here, we investigate a local requirement for NCOA4 in the regulation of hepatic iron stores and examine mechanisms of NCOA4 regulation. Hepatocyte-targeted Ncoa4 knockdown in non-phlebotomized mice had only modest effects on hepatic ferritin subunit levels and non-heme iron concentration. After phlebotomy, mice with hepatocyte-targeted Ncoa4 knockdown exhibited anemia and hypoferremia similar to control mice with intact Ncoa4 regulation, but showed a markedly impaired ability to lower hepatic ferritin subunit levels and hepatic non-heme iron concentration. This impaired hepatic response was observed even when dietary iron was limited. In both human and murine hepatoma cell lines, treatment with chemicals that stabilize hypoxia inducible factor (HIF), including desferrioxamine, cobalt chloride, and dimethyloxalylglycine, raised NCOA4 mRNA. This NCOA4 mRNA induction occurred within 3 hours, preceded a rise in NCOA4 protein, and was attenuated in the setting of dual HIF-1a and HIF-2a knockdown. In summary, we show for the first time that NCOA4 plays a local role in facilitating iron mobilization from the liver after blood loss and that HIF regulates NCOA4 expression in cells of hepatic origin. Because the prolyl hydroxylases that regulate HIF stability are oxygen and iron-dependent enzymes, our findings suggest a novel mechanism by which hypoxia and iron deficiency may modulate NCOA4 expression to impact iron homeostasis.

Blood ◽  
2019 ◽  
Vol 134 (Supplement_1) ◽  
pp. 431-431
Author(s):  
Xiuqi Li ◽  
Larisa Lozovatsky ◽  
Karin E. Finberg

The ubiquitously expressed intracellular protein NCOA4 mediates the degradation of ferritin in vitro (Mancias et al., Nature 2014; Dowdle et al., Nat Cell Biol 2014) and its loss disrupts systemic iron balance in mice (Bellelli et al., Cell Rep 2016). While a mechanism for increased NCOA4 protein turnover under high iron conditions has been reported in transformed cell lines of non-hepatic origin (Mancias et al., Elife, 2015), factors that regulate NCOA4 mRNA levels have not yet been described. Here we investigate stimuli that induce NCOA4 mRNA expression in hepatoma cells as well as in liver, the major iron depot of the body. We found that treating the human hepatoma cell line, Hep3B, for 18 hrs with increasing concentrations of the iron chelator desferrioxamine (DFO; 25-100μM) resulted in significantly higher NCOA4 mRNA (ANOVA P<.0001; 1.8 fold increase at the highest dose). Additionally, the murine hepatoma cell line, Hepa1-6, responded to 18 hrs of DFO treatment (25-100μM) with a dose-dependent rise in Ncoa4 mRNA levels (ANOVA P<.0001; 2.7 fold increase at the highest dose). DFO treatment of Hep3B and Hepa1-6 cells also increased mRNA levels of the transferrin receptor, confirming induction of iron deficiency. Because DFO has also been shown to stabilize hypoxia inducible factor (HIF), we examined the effects of other chemical hypoxia mimetics on hepatoma cell lines. Hep3B cells treated for 18 hrs with CoCl2 (25-75 μM), which is known to enhance the stability of HIF1α, showed a 2-fold increase in NCOA4 mRNA (ANOVA; P<.0001). Similarly, treatment of Hep3B cells with dimethyloxalylglycine (DMOG; 1mM), a competitive inhibitor of HIF prolyl-hydroxylases that promote HIF-α degradation, resulted in a 3-fold increase in NCOA4 mRNA at 12 hrs, which was further enhanced at 18 hrs. These results suggest that hepatoma cells in culture may respond to decreased iron availability and/or hypoxia by upregulating NCOA4 mRNA levels. We also investigated if Ncoa4 mRNA expression is upregulated in mouse livers under conditions associated with depletion of hepatic iron stores and/or hypoxia. C57BL/6N mice raised on a 45 p.p.m. iron diet were subjected to a large-volume phlebotomy (500μl; accompanied by intraperitoneal saline volume replacement) and fed an iron-deficient diet (2-6 p.p.m.) after bleeding. Seven days after phlebotomy, mice showed significantly lower blood hemoglobin levels (Student's T test P<.0001) and hepatic non-heme iron concentrations (P<.0001). However, the phlebotomized mice exhibited 1.8-fold higher hepatic Ncoa4 mRNA levels (P<.01) compared to non-phlebotomized controls. Additionally, we examined if hepatic Ncoa4 expression changed during normal pregnancy, a state in which extra-hepatic iron demands are increased by both the expanding maternal blood volume and the growing fetus. Compared to non-pregnant females, C57BL/6N mice at day 18.5 of pregnancy showed significantly lower liver non-heme iron concentrations (P<.01), while hepatic Ncoa4 mRNA levels were 3.5-fold higher (P<.01) in pregnant females compared to non-pregnant controls. By mining a published murine gene expression dataset (GEO accession GDS5818), we found that hepatic Ncoa4 mRNA levels are relatively high in early life (embryonic day 18 and postnatal day 5) and decline by adulthood (postnatal day 56), suggesting a relatively greater hepatic requirement for NCOA4 function during periods of high growth. Collectively, these in vivo data show that Ncoa4 upregulation correlates with conditions in which mobilizing iron for systemic use takes precedence over building hepatic iron stores. In summary, we show for the first time that Ncoa4 mRNA rises significantly in physiological and pathophysiological states in which increased extra-hepatic iron demands induce a reduction in hepatic iron stores. Additionally, we show that Ncoa4 mRNA levels in cultured cells of hepatic origin rise in response to hypoxia mimetics. Because NCOA4 functions as a cargo receptor that traffics ferritin to the autolysosome, NCOA4 protein is expected to be consumed during the process of ferritinophagy. We suggest that upregulation of hepatic Ncoa4 mRNA in response to hypoxia may represent a physiological adaptation to replenish NCOA4 protein that is needed to support continued mobilization of iron from the liver during periods of increased systemic iron demands. Disclosures No relevant conflicts of interest to declare.


Blood ◽  
2018 ◽  
Vol 132 (Supplement 1) ◽  
pp. 1046-1046
Author(s):  
Xiuqi Li ◽  
Larisa Lozovatsky ◽  
Dong Liu ◽  
Nadia Ayala-Lopez ◽  
Karin E. Finberg

Abstract The intracellular protein NCOA4 mediates the autophagic degradation of ferritin in vitro (Mancias et al., Nature 2014; Dowdle et al., Nat Cell Biol 2014); mice with global Ncoa4 disruption show hyperferremia, microcytic anemia, and ferritin accumulation in multiple organs, including liver (Bellelli et al., Cell Rep 2016). Here, we dissect the requirement for NCOA4 in hepatic iron mobilization after acute blood loss, using Ncoa4-targeting siRNA that was conjugated to triantennary N-acetylgalactosamine (GalNAc-Ncoa4 siRNA) to promote uptake by hepatocytes. On experimental day 0, 8-week-old female C57BL/6N mice underwent a single 500 μl phlebotomy, which was followed immediately by intraperitoneal volume replacement (saline) and concomitant subcutaneous injection of either GalNAc-Ncoa4 siRNA (3 mg/kg) or saline vehicle. The phlebotomized mice then underwent a second, terminal blood collection and organ harvest at either 3 days later (when mice subjected to this phlebotomy protocol are known to reach their hematocrit nadir) or 7 days later (when mice subjected to this phlebotomy protocol are known to exhibit substantial hematocrit recovery). To provide an experimental baseline, a group of 8-week-old mice that were not phlebotomized and did not receive a subcutaneous injection of either saline or GalNAc-Ncoa4-siRNA were analyzed on experimental day 0. Injection of GalNAc-Ncoa4 siRNA immediately after phlebotomy resulted in marked hepatic Ncoa4 knockdown at both 3 and 7 days after phlebotomy. Compared to non-phlebotomized mice (NPM), phlebotomized mice (PM) treated with either saline or GalNAc-Ncoa4 siRNA showed similar reductions in red blood cell count, hemoglobin concentration, and hematocrit at day 3, confirming that similar blood volumes were removed. Compared to NPM, PM treated with saline showed significantly lower levels of ferritin subunits by immunoblotting of liver lysates, consistent with a model in which phlebotomy induces the degradation of ferritin complexes in hepatocytes. By contrast, in PM treated with GalNAc-Ncoa4 siRNA, hepatic ferritin subunit levels did not decrease after blood loss, and at post-phlebotomy day 7, hepatic ferritin subunit levels were significantly higher in PM treated with GalNAc-Ncoa4 siRNA than in PM treated with saline. By post-phlebotomy day 7, mean liver non-heme iron concentration (LIC) was also significantly lower in PM injected with saline compared to NPM, suggesting that non-heme iron had been mobilized from their livers in the setting of increased iron demand for erythropoiesis. By contrast, in PM treated with GalNAc-Ncoa4 siRNA, mean LIC failed to decrease after phlebotomy. Hepatic Tfrc mRNA and protein were upregulated in PM treated GalNAc-Ncoa4 siRNA compared to saline, suggesting that an acute reduction in NCOA4 activity reduces cytosolic iron levels in hepatocytes. Interestingly, when compared to NPM, saline-treated PM showed a trend towards higher hepatic Ncoa4 mRNA when assessed at either day 3 (P=0.09) or day 7 (P=0.06) after blood loss, raising the possibility that hepatic NCOA4 requirements increase in response to phlebotomy. Although phlebotomy failed to lower hepatic iron stores in PM with hepatocellular Ncoa4 knockdown, hematological recovery at day 7 was similar in both treatment groups. At day 7 after phlebotomy, splenic non-heme iron stores were similarly depleted in both treatment groups. Interestingly, compared to PM treated with saline, PM treated with GalNAc-Ncoa4 siRNA showed lower hepatic hepcidin expression at day 7, raising the possibility that greater dietary iron absorption may compensate for the lack of hepatic iron mobilization in this group to allow for normal hematological recovery. Collectively, these findings suggest that NCOA4 activity is required to mobilize iron from the liver when extrahepatic iron demands increase following acute blood loss. However, in our model with hepatocellular Ncoa4 knockdown, dietary iron uptake and/or iron release from extrahepatic storage sites may compensate for the lack of hepatic iron mobilization to support stress erythropoiesis. While ferritin degradation has been shown to occur through both lysosomal and proteasomal pathways in vitro, our findings suggest that if NCOA4 expression is reduced following acute blood loss, other cellular pathways in hepatocytes cannot compensate to mobilize iron from the liver. Disclosures No relevant conflicts of interest to declare.


Blood ◽  
2010 ◽  
Vol 116 (21) ◽  
pp. 4258-4258
Author(s):  
Ramsey M. Wehbe ◽  
Rebecca L. Whittlesey ◽  
Nancy C. Andrews ◽  
Karin E. Finberg

Abstract Abstract 4258 Mutations in TMPRSS6 (matriptase-2), a transmembrane serine protease expressed by the liver, result in the clinical phenotype of iron refractory iron deficiency anemia (IRIDA). Additionally, common polymorphisms in TMPRSS6 have been associated with variation in laboratory parameters of iron homeostasis in healthy populations. TMPRSS6 increases iron absorption by reducing expression of the hepatic hormone, hepcidin, via down-regulation of a BMP/SMAD signaling cascade. Hepcidin promotes the internalization and degradation of the duodenal iron transporter, ferroportin, thereby inhibiting iron absorption. Previous studies have demonstrated that adult mice with Tmprss6 deficiency exhibit elevated hepatic hepcidin mRNA levels that are associated with decreased hepatic iron stores. In one study, genetic loss of Tmprss6 was shown to result in significant elevation of hepatic hepcidin expression in mice at birth; however, whether this hepcidin elevation was associated with abnormalities in iron homeostasis was not reported. We therefore asked if the elevated hepcidin levels present in newborn Tmprss6-/- pups correlate with abnormal parameters of iron homeostasis in the fetal or neonatal periods. To answer this question, we intercrossed Tmprss6+/− mice to generate Tmprss6+/+, Tmprss6+/−, and Tmprss6-/- progeny for phenotypic characterization at either gestational day 17.5 (E17.5) or postnatal day 0 (P0). Consistent with prior observations, Tmprss6-/- pups at P0 showed a 4.6-fold increase in hepatic hepcidin mRNA compared to Tmprss6+/+ littermates (p=.006). However, despite this elevation in hepcidin expression, Tmprss6-/- pups were not pale, and they showed no significant differences in body mass or hepatic non-heme iron concentration compared to Tmprss6+/+ and Tmprss6+/− littermates. At E17.5, Tmprss6-/- fetuses showed a 50-fold increase in hepatic hepcidin mRNA compared to Tmprss6+/+ littermates (p=.005). However, Tmprss6-/- fetuses also were not pale, and they showed no significant difference in body mass compared to Tmprss6+/+ and Tmprss6+/− littermates. Surprisingly, hepatic non-heme iron concentration at E17.5 was significantly higher in Tmprss6-/- fetuses than in Tmprss6+/+ fetuses (p=.003). To determine if the increased hepcidin expression of Tmprss6-/- fetuses might affect iron homeostasis in their pregnant mothers, we measured iron parameters in Tmprss6+/− females gestating E17.5 litters that were enriched for either Tmprss6+/+ or Tmprss6-/- fetuses. No significant effects of fetal genotype on maternal iron parameters were observed. In summary, our results demonstrate that Tmprss6 regulates hepcidin expression in the fetal and neonatal periods in mice. However, Tmprss6 deficiency does not appear to be associated with systemic iron deficiency at these stages of development, and fetal Tmprss6 expression does not have a significant effect on maternal iron homeostasis in late gestation. These results may have implications for understanding the maintenance of iron homeostasis in early development, and may provide insight into the evolution of IRIDA as well as other disorders of iron homeostasis. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2013 ◽  
Vol 122 (21) ◽  
pp. 179-179
Author(s):  
Christine E. McLaren ◽  
Mary J. Emond ◽  
Pradyumna D. Phatak ◽  
Paul C. Adams ◽  
V. Nathan Subramaniam ◽  
...  

Abstract Variability in the severity of iron overload among homozygotes for the HFE C282Y polymorphism is one of the major problems extant in our understanding of hereditary hemochromatosis (HH). We conducted exome sequencing of DNA from C282Y homozygotes with markedly increased iron stores (cases) and C282Y homozygotes with normal or mildly increased iron stores (controls) to identify rare and common causal variants associated with variability of disease expression in HH. Criteria for cases included serum ferritin >1000 µg/L at diagnosis, and (a) mobilized body iron >10 g by quantitative phlebotomy, and/or (b) hepatic iron concentration >236 µmol/g dry weight. Criteria for controls included (a) serum ferritin <300 µg/L, or (b) age ≥50 y with ≤3.0 g iron removed by phlebotomy or age ≥40 y with ≤2.5 g iron removed by phlebotomy to achieve serum ferritin <50 µg/L. Deep sequencing of the full exome was performed in 33 cases and 14 controls. After quality control filtering, the dataset included 82,068 SNPs and 1,403 insertions/deletions (indels). Our initial analysis tested for differences in the distribution of variants between groups for each gene separately using the Sequence Kernel Association Test (SKAT) that includes rare and common variants but downweights the contribution of common variants to the test statistic. Only non-synonymous variants were included in the by-gene tests. Principal components were constructed from the exome variants to adjust for possible confounding by ancestry and to confirm no ancestral outliers. All study participants were male, and all clustered closely together within a larger group of Europeans in a principal components analysis of ancestry. Mean (SD) ages at presentation were 54 (11.0) y and 56 (9.4) y for cases and controls, respectively. Median serum ferritin was 2788 µg/L in those with increased iron stores and 309 μg/L in those with normal or mildly increased iron stores. The median transferrin saturation (94%) was greater in cases than in the comparison group (70%). In a preliminary analysis, we found 9 genes associated with case-control status. To separate effects of alcohol use and/or alcohol addiction variants, an analysis was conducted to compare the 13 controls and 22 cases who reported never using alcohol or only very light use. The two most significant genes identified in this comparison were GNPAT (p=7.4x10-6) and CDHR2 (p=2.8x10-4). A quantile-quantile (QQ) plot is shown in the Figure, comparing the observed distribution of –(log10p-values) from 10,337 genes to the expected uniform distribution if there were no variants modifying severity of expression, and gives evidence of the effect of the GNPAT gene.Figure 1Figure 1. Inspection of the two variants contributing to the GNPAT by-gene p-value revealed one missense variant (rs11558492) for which 0/13 controls had a polymorphism, while 16/22 cases had at least one missense variant, and one case was homozygous for this missense variant. The latter case presented at the early age of 26 with a serum ferritin of 1762 µg/L, 4+ hepatocellular iron and hepatic iron concentration of 284.4 µmol/g dry weight. GNPAT (aka DHAPAT) mutations/deletions have been found in peroxisomal disease, a class of diseases in which increased hepatic iron is observed (Biochim Biophys Acta 1801:272-280, 2010). GNPAT rs11558492 is common among people of European descent but might interact with aberrant HFE to increase risk of hepatic iron overload. Three rare variants in CDHR2 accounted for its low p-value, having a cumulative frequency of 4/13 among controls and 0/22 among cases: rs115050587, rs752138, rs143224505 with minor allele frequencies, MAF = 1.4%, 4.7% and 0.06%, respectively. The first two polymorphisms are predicted to be highly damaging by PolyPhen2 and the third probably damaging. Expression levels of CDHR2 recently have been associated with increased hepatocyte iron and elevated serum ferritin in liver allograft patients (J Clin Invest 122:368-382, 2012). These data indicate associations between iron status in HFE C282Y homozygotes and genes with previous links to iron overload that may modify severity of disease expression. Of note, the data suggest that more than one modifier gene may be involved in determining severity of disease in HFE C282Y homozygotes. Our results identify candidate genes for expanded studies that would examine their functional significance for iron absorption and metabolism. Disclosures: No relevant conflicts of interest to declare.


2019 ◽  
Vol 59 ◽  
pp. 272-280 ◽  
Author(s):  
Lingyu Wu ◽  
Yaqun Zou ◽  
Yu Miao ◽  
Jiayou Zhang ◽  
Suqin Zhu ◽  
...  

Circulation ◽  
2014 ◽  
Vol 130 (suppl_2) ◽  
Author(s):  
Xiaoyan Yang ◽  
Athanassios Vassilopoulos ◽  
Seong-Hoon Park ◽  
David Gius ◽  
Hossein Ardehali

Background: Sirtuins (SIRTs) are NAD+-dependent deacetylases and critical regulators of energy metabolism and response to oxidative stress in the heart. Iron is essential for these processes but is toxic when present in excess. Thus, SIRTs may regulate iron levels to ensure adequate supply of this element for their biological functions. SIRT2 is among the least characterized SIRTs and is mainly present in the cytoplasm. We hypothesized that SIRT2 might be required for cellular iron homeostasis. Methods and Results: Iron content was significantly lower in SIRT2-/- mouse embryonic fibroblasts (MEFs) compared to SIRT2+/+ MEFs (non-heme iron: 0.073 vs. 0.060 nmol/μg protein, p=0.02). Gene expression of ferroportin-1 (FPN1), the major cellular iron exporter, was significantly increased in SIRT2-/- MEFs. Similarly, silencing SIRT2 in HepG2 cells decreased cellular iron levels and increased FPN1 expression, indicating that enhanced FPN1 in SIRT2 knockout or knockdown condition increases iron export and reduces cellular iron. To investigate the underlying mechanism, we focused our studies on nuclear factor (erythroid-derived 2)-like 2 (Nrf2), a known regulator of FPN1. Our results demonstrated that Nrf2 is upregulated and translocates into the nucleus in SIRT2-/- MEFs and knocking down Nrf2 in SIRT2-/- MEFs reverses iron deficiency and FPN1 expression. Furthermore, Nrf2 is acetylated by P300/CBP and can be deacetylated by SIRT2. Finally, to confirm the role of SIRT2 in iron regulation, cellular heme and non-heme iron in the heart (major iron-consuming organ) and liver (major iron-storage organ) were measured in wild type (WT) and SIRT2-/- mice. Heme and non-heme iron content were significantly decreased in SIRT2-/- mouse livers compared to WT livers (heme: 2.25 vs. 1.65 nmol/mg protein, p=0.002; non-heme iron: 0.073 vs. 0.064 nmol/μg protein, p=0.03). Furthermore, heme levels were also significant decreased in the heart, while non-heme iron was not significantly altered. Conclusions: Our results suggest that SIRT2 regulates cellular iron homeostasis by deacetylating NRF2 and altering iron export through FPN1.


2014 ◽  
Vol 115 (suppl_1) ◽  
Author(s):  
Xiaoyan Yang ◽  
Athanassios Vassilopoulos ◽  
Seong-Hoon Park ◽  
David Gius ◽  
Hossein Ardehali

Background: Sirtuins (SIRTs) are NAD+-dependent deacetylases, which regulate energy metabolism and response to oxidative stress in the heart. Iron is essential for these processes but is toxic when present in excess. However, whether SIRTs are involved in maintaining cellular iron homeostasis is not known. SIRT2 is among the least characterized SIRTs and is mainly present in the cytoplasm. We hypothesized that SIRT2 is required for cellular iron homeostasis. Methods and Results: Iron content was significantly lower in SIRT2-/- mouse embryonic fibroblasts (MEFs) compared to SIRT2+/+ MEFs (non-heme iron: 0.073 vs. 0.060 nmol/μg protein, p=0.02), andlevels of ferroportin-1 (FPN1), the major cellular iron exporter, was significantly increased in SIRT2-/- MEFs. Similarly, silencing SIRT2 in HepG2 cells decreased cellular iron levels and increased FPN1 expression, indicating that enhanced FPN1 with SIRT2 downregulation drove iron export and caused a reduction in cellular iron levels. Furthermore, iron export assays showed that iron export was increased in HepG2 cells with SIRT2 knockdown. To investigate the underlying mechanism, we focused our studies on nuclear factor (erythroid-derived 2)-like 2 (Nrf2), a known regulator of FPN1. Our results demonstrated that Nrf2 is upregulated and translocates into the nucleus in SIRT2-/- MEFs and knocking down Nrf2 in SIRT2-/- MEFs reverses iron deficiency. Furthermore, Nrf2 is acetylated by P300/CBP and can be deacetylated by SIRT2. Finally, to confirm the role of SIRT2 in iron regulation, cellular heme and non-heme iron in the heart (major iron-consuming organ) and liver (major iron-storage organ) were measured in wild type (WT) and SIRT2-/- mice. Heme and non-heme iron content were significantly decreased in SIRT2-/- mouse livers compared to WT livers (heme: 2.25 vs. 1.65 nmol/mg protein, p=0.002; non-heme iron: 0.073 vs. 0.064 nmol/μg protein, p=0.03). Furthermore, heme levels were also significant decreased in the heart, while non-heme iron was not significantly altered. Conclusions: Our results suggest that SIRT2 regulates cellular iron homeostasis by deacetylating NRF2 and altering iron export through FPN1.


2000 ◽  
Vol 343 (5) ◽  
pp. 327-331 ◽  
Author(s):  
Emanuele Angelucci ◽  
Gary M. Brittenham ◽  
Christine E. McLaren ◽  
Marta Ripalti ◽  
Donatella Baronciani ◽  
...  

Blood ◽  
2009 ◽  
Vol 114 (22) ◽  
pp. 625-625
Author(s):  
Karin E. Finberg ◽  
Rebecca Whittlesey ◽  
Mark D. Fleming ◽  
Nancy C. Andrews

Abstract Abstract 625 HFE-associated hereditary hemochromatosis is an autosomal recessive disorder characterized by inappropriately elevated absorption of dietary iron by the gastrointestinal mucosa, resulting in excessive storage of iron in multiple organs. A significant proportion of individuals who are homozygous for HFE mutations fail to develop clinical symptoms, suggesting that environmental and/or genetic factors may influence the penetrance of this disorder. In vitro and animal studies have revealed that HFE promotes the expression of hepcidin, a circulating hormone produced by the liver that acts to inhibit iron absorption by the duodenum. In contrast, TMPRSS6, a transmembrane serine protease produced by the liver, acts to inhibit hepcidin expression; both humans and mice harboring TMPRSS6 mutations display impaired intestinal iron absorption, resulting in a phenotype of iron-refractory iron deficiency anemia (IRIDA). Here we asked if heterozygous or homozygous loss of Tmprss6 function could modify the iron overload phenotype of Hfe null (Hfe-/-) mice, a mouse model of human HFE-hemochromatosis. To test this, we bred Hfe-/- mice to Tmprss6-/- mice; the latter harbor a targeted disruption of the Tmprss6 serine protease domain and exhibit an IRIDA phenotype. We generated Hfe-/-Tmprss6+/+, Hfe-/-Tmprss6+/-, and Hfe-/-Tmprss6-/- female mice (6-10 mice per genotype), in which parameters of systemic iron homeostasis were compared at eight weeks of age by Student's t test. Consistent with previous study of Hfe-/- mice, Hfe-/- mice harboring two wild type Tmprss6 alleles (Hfe-/-Tmprss6+/+ mice) showed serum iron concentration, transferrin saturation, and hepatic non-heme iron content that were significantly elevated compared to wild type mice of similar genetic background. Heterozygosity for Tmprss6 mutation, however, markedly reduced the severity of the hemochromatosis phenotype of Hfe-/- mice. Compared to Hfe-/- mice with two wild type Tmprss6 alleles, Hfe-/- mice that were heterozygous for Tmprss6 mutation (Hfe-/-Tmprss6+/- mice) showed significant reductions in serum iron concentration (p<0.01), transferrin saturation (p<0.005), and non-heme iron content of liver (p<10-4). Furthermore, homozygosity for Tmprss6 mutation completely ameliorated the iron overload phenotype of Hfe-/- mice and in fact led to systemic iron deficiency. Compared to both Hfe-/-Tmprss6+/+ and Hfe-/-Tmprss6+/- mice, Hfe-/-Tmprss6-/- mice showed markedly reduced serum iron concentration (p<10-7), transferrin saturation (p<10-10), and non-heme iron content of liver (p<10-4). Hfe-/-Tmprss6-/- mice also displayed iron deficiency anemia and appeared phenotypically similar to previously characterized Tmprss6-/- mice harboring two wild type copies of Hfe. In summary, these results demonstrate that Tmprss6 is a genetic modifier of the Hfe-hemochromatosis phenotype in mice. These findings suggest that natural genetic variation in the human ortholog TMPRSS6 might modify the clinical penetrance of HFE-hemochromatosis and raise the possibility that pharmacological inhibition of TMPRSS6 activity might prove an effective therapy in this disorder. Disclosures: No relevant conflicts of interest to declare.


Sign in / Sign up

Export Citation Format

Share Document