You are on page 1of 9

GASTROENTEROLOGY 2009;137:1489 –1497

Bone Morphogenetic Protein Signaling Is Impaired in an Hfe Knockout


Mouse Model of Hemochromatosis

ELENA CORRADINI,*,‡ CINZIA GARUTI,‡ GIULIANA MONTOSI,‡ PAOLO VENTURA,‡ BILLY ANDRIOPOULOS Jr,*
HERBERT Y. LIN,* ANTONELLO PIETRANGELO,‡ and JODIE L. BABITT*
*Program in Membrane Biology, Division of Nephrology, Center for Systems Biology, Massachusetts General Hospital, Harvard Medical School, Boston,
Massachusetts; ‡Center for Hemochromatosis, University Hospital of Modena and Reggio Emilia, Modena, Italy

BACKGROUND AND AIMS: Mutations in HFE are the still is not understood fully. HFE encodes an atypical
most common cause of the iron-overload disorder hered- class I major histocompatibility complex molecule that
itary hemochromatosis. Levels of the main iron regula- requires ␤2 microglobulin for appropriate localization to
tory hormone, hepcidin, are inappropriately low in he- the cell surface.6 – 8 HFE binds to transferrin receptor 1
reditary hemochromatosis mouse models and patients (TfR1)9 –11 at a site that overlaps with the transferrin
with HFE mutations, indicating that HFE regulates hep- binding site.12–15 HFE also binds to the liver-specific
cidin. The bone morphogenetic protein 6 (BMP6)–SMAD homologue TfR2.16,17 Selective disruption of the Hfe gene
signaling pathway is an important endogenous regulator in the liver recapitulates the phenotype of Hfe knockout
of hepcidin expression. We investigated whether HFE is (KO) mice, suggesting that the liver is the predominant
involved in BMP6 –SMAD regulation of hepcidin expres- organ for HFE action in the regulation of iron metabo-
sion. METHODS: The BMP6 –SMAD pathway was ex- lism.18 Notably, both mouse models and human patients
amined in Hfe knockout (KO) mice and in wild-type (WT) with HFE mutations have inappropriately low expression
mice as controls. Mice were placed on diets of varying
of the key iron regulatory hormone hepcidin,19 –23 which
iron content. Hepcidin induction by BMP6 was examined
is produced in the liver.24 –26 Hepcidin down-regulates the
in primary hepatocytes from Hfe KO mice; data were
iron exporter ferroportin on the surface of duodenal
compared with those of WT mice. RESULTS: Liver levels
enterocytes, macrophages, and hepatocytes, thereby in-
of Bmp6 messenger RNA (mRNA) were higher in Hfe KO
hibiting iron release into the bloodstream.27 It is now
mice; these were appropriate for the increased hepatic
levels of iron in these mice, compared with WT mice. widely accepted that impaired regulation of hepcidin
However, levels of hepatic phosphorylated Smad 1/5/8 expression by HFE plays a central role in the pathogen-
protein (an intracellular mediator of Bmp6 signaling) esis of HFE–HH. It recently has been postulated that
TfR1 in the liver serves to sequester HFE, and that HFE

PANCREAS, AND
and Id1 mRNA (a target gene of Bmp6) were inappropri-

BILIARY TRACT
BASIC–LIVER,
ately low for the body iron burden and Bmp6 mRNA is displaced from TfR1 by holotransferrin.28 This dis-
levels in Hfe KO, compared with WT mice. BMP6 induc- placed HFE then is able to generate a signal to up-
tion of hepcidin expression was reduced in Hfe KO hepa- regulate hepcidin via an interaction with TfR2 and trans-
tocytes compared with WT hepatocytes. CONCLU- ferrin.16,28,29 However, the precise molecular mechanism
SIONS: HFE is not involved in regulation of BMP6 by by which HFE affects hepcidin expression still is un-
iron, but does regulate the downstream signals of known.
BMP6 that are triggered by iron. A severe juvenile form of HH is caused by mutations in
the gene encoding hemojuvelin (HFE2).30 We recently
discovered that hemojuvelin is a co-receptor for the bone

H ereditary hemochromatosis (HH) is a genetically


heterogeneous hereditary disorder of iron metabo-
lism caused by a failure to prevent unneeded iron from
morphogenetic protein (BMP) signaling pathway31 and
that the BMP signaling function of hemojuvelin is im-
portant for its role in iron metabolism.31,32 Members of
entering the circulatory pool and characterized by pro- the transforming growth factor-␤ family, BMPs exert
gressive iron overload with potential for multi-organ
damage and disease.1 A common polymorphism in HFE
is associated with the most frequent and first character- Abbreviations used in this paper: ANOVA, analysis of variance; BMP,
ized form of HH, affecting approximately 5 per 1000 bone morphogenetic protein; CTR, control; HH, hereditary hemochro-
individuals of northern European descent.2 Mouse mod- matosis; ID, iron-deficient; IR, iron-enriched; KO, knockout; LIC, liver
els with disruptions of the Hfe gene have a similar iron iron concentration; P-Smad1/5/8, phosphorylated Smad1/5/8; TfR,
transferrin receptor; WT, wild-type.
overload phenotype.3–5 © 2009 by the AGA Institute
Although the discovery of the HFE gene6 dates back 0016-5085/09/$36.00
more than 10 years, the pathophysiology of HFE–HH doi:10.1053/j.gastro.2009.06.057
1490 CORRADINI ET AL GASTROENTEROLOGY Vol. 137, No. 4

their effects by binding to type I and type II serine cient diet, ID). All mice were killed at 12 weeks of age, and
threonine kinase receptors, which phosphorylate specific blood and livers were harvested for analysis.
intracellular SMAD proteins (SMAD1/5/8). Phosphory-
lated SMAD1/5/8 (P-SMAD1/5/8) binds to the com- Hematologic and Iron Parameters
mon mediator SMAD4, and the SMAD complex trans- Blood was obtained by retroorbital puncture. Se-
locates to the nucleus to affect transcription of target rum iron and total iron binding capacity were deter-
genes such as ID1.33–35 We and others have shown that mined using a kit (Pointe Scientific Inc) according to the
HAMP (encoding hepcidin) is also a target gene that is manufacturer’s instructions. Transferrin saturation was
up-regulated by BMP signals at the transcriptional level calculated as follows: (serum iron ⫼ total iron binding
in vitro.31,32,36 –39 In mice, BMP ligands increase hepatic capacity) ⫻ 100%. Red blood cells, hemoglobin, and he-
Hamp1 messenger RNA (mRNA) expression and reduce matocrit levels were determined using an automated cell
serum iron, whereas BMP inhibitors reduce hepatic counter at the clinical-chemical laboratory of the Univer-
Hamp1 mRNA expression, mobilize reticuloendothelial sity Hospital of Modena.
cell iron stores, and increase serum iron.32,40,41 Impaired
hepatic BMP signaling, through mutations in genes en- Tissue Iron Content
coding the BMP co-receptor hemojuvelin,42,43 the com- Tissue specimens were analyzed for non– heme
mon BMP/transforming growth factor-␤ intracellular iron content by the method of Torrance and Bothwell.46
mediator Smad4,36 or the ligand Bmp6,41,44 leads to low The liver iron concentration (LIC) for each animal was
hepcidin levels and iron overload in mouse models. Re- the mean of measurements performed on 2 different
cent evidence also suggests that BMP signaling in the tissue specimens from each liver, Germantown, MD.
liver is modulated by iron.40,45 Parenteral iron increases
Quantitative Real-Time Reverse-
hepatic Smad1/5/8 phosphorylation within 1 hour,40
Transcription Polymerase Chain Reaction
and long-term changes in dietary iron modulate hepatic
Bmp6 mRNA expression in mice concordantly with Id1 Total RNA was isolated from mouse liver tissue
and Hamp1 mRNA.45 Collectively, these data indicate using the RNeasy Mini Kit (QIAgen, Germantown, MD),
that BMP–SMAD signaling is a main regulatory pathway with DNAse digestion with the RNase-Free DNase Set
acting in iron metabolism. (QIAgen) or TRIzol reagent (Invitrogen, Carlsbad, CA)
Here, we dissect the BMP–SMAD signaling pathway in according to the manufacturers’ instructions as previ-
ously described.41,47 Real-time quantification of Bmp6,
an Hfe KO mouse model to determine whether HFE
Bmp4, Bmp2, Id1, Hamp1, Rpl19, and glyceraldehyde-3-phos-
regulation of hepcidin expression involves the BMP–
phate dehydrogenase (Gapdh) mRNA transcripts was per-
SMAD signaling pathway. Our data show that hepatic
formed using 2-step quantitative real-time reverse-tran-
Bmp6 mRNA expression is regulated appropriately by
scription polymerase chain reaction as previously
dietary iron content and tissue iron burden in Hfe KO
described.41,47 Samples were analyzed in duplicate or trip-
PANCREAS, AND
BILIARY TRACT

mice. However, HFE appears to be involved in the down-


BASIC–LIVER,

licate, and expression levels were normalized to the


stream signals of BMP6 that are triggered by iron.
housekeeping genes Rpl19 or Gapdh.

Materials and Methods Western Blot


Animals Livers were disrupted with a manual homogenizer
in lysis buffer (1 ⫻ Tris-buffered saline, 0.1% sodium
Wild-type (WT) 129S6/SvEvTac mice and Hfe KO dodecyl sulfate, 10 ␮L/mL Triton X-100, 1 g/100 mL Na
mice bred on a 129S6/SvEvTac background,4 kindly pro- deoxycholate, 2 ␮L/mL ethylenediaminetetraacetic acid)
vided by Dr Nancy C. Andrews (Duke University Medical containing protease inhibitors (Complete Mini; Roche
Center), received humane care under approved experi- Diagnostic, Indianapolis, IN) and phosphatase inhibitors
mental protocols according to the University Hospital of (Halt Phosphatase Inhibitor Cocktail; Thermo Scientific,
Modena guidelines. Six Hfe KO (3 males, 3 females) and Rockford, IL). Western blot of liver lysates for P-Smad1/
6 WT (2 males, 4 females) mice were maintained on a 5/8 relative to total Smad1 protein and chemilumines-
standard rodent diet containing 330 ppm iron (2018 cence quantitation was performed as previously de-
Teklad; Harlan, Indianapolis, IN) until 12 weeks of age scribed.32
(control diet, CTR). Dietary iron loading was achieved by
feeding six 8-week-old Hfe KO and six WT mice (3 males, Primary Hepatocyte Cultures and BMP6
3 females each) the same standard diet supplemented Treatment
with 2% carbonyl iron for 4 weeks (iron-enriched diet, IR). Primary hepatocytes from 8- to 12-week-old male
Six 7-week-old Hfe KO and 6 WT mice (4 males, 2 females 129S6/SvEvTac WT and Hfe KO mice maintained on a
each) were fed with a diet with negligible iron content standard rodent diet (300 ppm iron, Teklad; Harlan) were
(customized diet, Teklad; Harlan) for 5 weeks (iron-defi- isolated as previously described47 with the following
October 2009 HFE AND BMP SIGNALING 1491

modifications. Hepatocytes were plated on plastic multi-


well plates at a density of 300,000 – 400,000 cells/mL in
Dulbecco’s modified Eagle medium (Invitrogen) supple-
mented with 100 IU/mL penicillin, 30 mg/mL streptomy-
cin, and 10% fetal calf serum. Twenty-four hours after
isolation, hepatocytes were serum-starved for 6 hours in
Dulbecco’s modified Eagle medium with 1% fetal calf
serum, followed by stimulation with 0.1–2.0 ng/mL of
BMP6 (R&D Systems, Minneapolis, MN) in Dulbecco’s
modified Eagle medium with 1% fetal calf serum for 16
hours.

Statistics
All data were controlled for normal distribution
(Kolgomorov–Smirnov and Shapiro–Wilk tests). In cases
of skewed distribution, log-transformed data were used.
A 2-tailed independent Student t test was used to com- Figure 1. Liver iron concentration and Bmp6 mRNA levels are in-
pare normalized mean values of considered variables be- creased, phosphorylated Smad1/5/8 protein and Id1 mRNA are un-
tween strains under the same diet regimen. Two-way changed, and Hamp1 mRNA expression is decreased in Hfe KO vs WT
analysis of variance (ANOVA), with the Holm–Sidak post mice on a standard diet. Twelve-week-old Hfe KO mice (N ⫽ 6) and WT
control mice (N ⫽ 6) were analyzed for hepatic Bmp6, Id1, and Hamp1
hoc test for pair-wise multiple comparisons, was used to relative to Rpl19 mRNA expression by quantitative real-time reverse-
compare, between and within considered groups, the transcription polymerase chain reaction, and for P-Smad1/5/8 relative
mean levels of the considered variables as a function of to total Smad1 protein expression by Western blot quantified using
diet, strain, and the interaction between these 2 factors. IPLab Spectrum software (Rockville, MD). Results are reported as the
One-way ANOVA, with the Fisher’s least significant dif- mean ⫾ SD for the fold change from WT mice. Exact P values deter-
mined by 2-tailed independent Student’s t-tests are shown for the
ference post hoc test for pair-wise multiple comparisons, comparisons between Hfe KO mice and WT mice. Serum iron, serum
was used to compare mean values for the BMP6 dose- transferrin saturation (Tf Sat), and LIC also were determined. Results
response curves in primary hepatocyte cultures. The Pear- from pooled samples are shown for serum iron and Tf Sat. Mean values
son r coefficient was used to assess the correlation be- are shown for LIC.
tween continuous variables, and the z-statistic was used
to compare r coefficients. For small sample sizes, the
Spearman r coefficient also was calculated to confirm the with a recent article showing that hepatic Bmp6 mRNA
results. In all statistical analyses, a P value of less than .05 expression correlates with dietary iron content and

PANCREAS, AND
was considered significant. All analyses were conducted tissue iron burden in mice with C57BL/6 and DBA/2

BILIARY TRACT
BASIC–LIVER,
using SPSS v.17.0 (SPSS Inc, Chicago, IL) and SigmaStat backgrounds.45 Similar to the previous study,45 we did
v.3.5 (Systat Software Inc, Richmond, CA) statistical soft- not find changes in hepatic Bmp2 and Bmp4 mRNA
ware. levels in Hfe KO mice despite significant iron overload
(data not shown). In contrast to the findings for Bmp6
mRNA, hepatic P-Smad1/5/8 (an intracellular media-
Results tor of BMP6 signaling) and Id1 mRNA (a target gene
Hepatic Bmp6 mRNA Is Increased, Whereas up-regulated by BMP6) were not increased in Hfe KO
P-Smad1/5/8 Protein and Id1 mRNA Are compared with WT mice (Figure 1, dark gray and light gray
Not Increased, in Hfe KO Mice Compared bars). Consistent with previous studies,19 –22 Hamp1
With WT Mice on a Standard Diet mRNA levels were lower in Hfe KO compared with WT
To examine the role of HFE in the BMP–SMAD mice, and inappropriately low for LIC (Figure 1, white
signaling pathway, we analyzed 12-week-old Hfe KO mice bars).
on a 129/SvEvTac background compared with matched
WT mice maintained on a standard diet for blood and Hfe KO Mice Have Equivalent Hepatic
tissue iron parameters, hepatic Bmp6 mRNA, P-Smad1/ Bmp6 mRNA Expression, but Lower
5/8 protein, Id1 mRNA, and hepcidin (Hamp1) mRNA P-Smad1/5/8 Protein and Id1 mRNA
expression (Figure 1). Hfe KO animals manifested a well- Expression, Compared With WT Mice With
known iron overload phenotype3–5 with increased LIC, Equivalent Iron Burden
serum iron, and transferrin saturation compared with To further examine the modulation of the BMP6 –
WT controls (Figure 1, bottom). Hepatic Bmp6 relative to SMAD pathway by iron and to compare animals with
Rpl19 mRNA levels were significantly higher in Hfe KO similar body iron burden, we fed Hfe KO mice and
compared with WT mice (black bars). This is consistent matched WT mice an ID diet, a CTR diet, or an IR diet
1492 CORRADINI ET AL GASTROENTEROLOGY Vol. 137, No. 4

Hepatic Bmp6 mRNA Expression Is


Appropriately Up-Regulated by Iron in Hfe
KO Mice
To further understand the regulation of the
BMP6 –SMAD pathway by iron in Hfe KO mice, we com-
pared hepatic Bmp6 mRNA, P-Smad1/5/8 protein, Id1
mRNA, and Hamp1 mRNA expression in Hfe KO mice
maintained on ID, CTR, and IR diets. The same param-
eters were examined in WT mice as a control. In Hfe KO
mice, hepatic Bmp6 mRNA levels were modulated signif-
icantly by dietary iron, and were regulated concordantly
with LIC in a manner similar to WT mice (compare
Figure 4A with Figure 2). To examine whether the in-
crease in Bmp6 mRNA levels in Hfe KO mice was appro-
priate for iron burden, we compared the ratio of Bmp6
mRNA levels with LIC. Bmp6 mRNA levels relative to LIC
in Hfe KO mice were not significantly different from
those in WT mice across all diets (Figure 4B). The Pear-
Figure 2. Dietary iron modulates the iron burden of WT and Hfe KO son chi-square test showed a significant correlation be-
mice. Seven- to 8-week-old Hfe KO mice and WT control mice were tween Bmp6 mRNA levels and LIC in the overall animal
maintained on an ID, CTR, or IR diet for 4 –5 weeks (N ⫽ 6 per group). population (r ⫽ 0.726, P ⫽ .0001). This correlation was
Mice were analyzed for LIC, serum transferrin saturation (Tf Sat), and
serum iron levels. Results for serum Tf Sat and serum iron are from
maintained after separate analysis in WT (r ⫽ 0.726, P ⫽
pooled samples. LIC is expressed as the mean ⫾ SD. Exact P values .001) and Hfe KO mice (r ⫽ 0.730, P ⫽ .001), and was not
determined by 2-way ANOVA (or NS if not significant) are shown. significantly different between the 2 genotypes (z ⫽
⫺0.0233, P ⫽ .9814). Because of the small sample size,
nonparametric statistics also were used, with similar re-
sults. These data suggest that hepatic Bmp6 mRNA cor-
for 4 or 5 weeks (N ⫽ 6 per group). WT mice on an ID
diet had a lower mean cellular volume (54.6 ⫾ 0.63 vs
59.8 ⫾ 1.78; P ⫽ .0004) and a trend toward a lower
hematocrit level (47.75 ⫾ 4.3 vs 52.43 ⫾ 2.4; P ⫽ .05)
compared with WT mice on a CTR diet, but no other
significant differences in erythroid parameters were
PANCREAS, AND

found among animals receiving different diets (data not


BILIARY TRACT

BASIC–LIVER,

shown). Dietary iron caused a dose-dependent increase in


serum iron in WT mice, but was less effective at modu-
lating serum iron parameters in Hfe KO mice, in which
the iron burden already was high (Figure 2, bottom).
Dietary iron also significantly modulated LIC in both WT
and Hfe KO mice (Figure 2). Notably, WT and Hfe KO
mice on an IR diet had equivalent LIC and serum iron
parameters (Figure 2, black bars). We therefore examined
the BMP6 –SMAD signaling pathway in WT and Hfe KO
mice on an IR diet to compare mice with a similar body
iron burden.
Hepatic Bmp6 mRNA showed a trend toward higher Figure 3. Hfe KO mice have adequate hepatic Bmp6 mRNA expres-
levels in Hfe KO compared with WT mice maintained on sion, but lower P-Smad1/5/8 protein, Id1 mRNA, and Hamp1 mRNA
IR diet (Figure 3, black bars). This is consistent with the expression compared with WT mice with equivalent iron burden. Eight-
week-old Hfe KO mice and WT control mice were maintained on an IR
nonsignificant trend toward increased LIC in the Hfe KO diet for 4 weeks (N ⫽ 6 mice per group). Mice were analyzed for Bmp6,
mice. In contrast, hepatic P-Smad1/5/8 protein and Id1 Id1, and Hamp1 relative to Rpl19 mRNA expression, and for P-Smad1/
mRNA levels (Figure 3, dark gray and light gray bars) were 5/8 relative to total Smad1 protein expression as in Figure 1. Results are
significantly lower in Hfe KO compared with WT mice on reported as the mean ⫾ SD for the fold change from WT mice. Exact P
values determined by 2-tailed independent Student’s t-tests are shown
an IR diet. As expected, Hamp1 mRNA expression was for the comparisons between Hfe KO mice and WT mice. Serum iron,
significantly lower in Hfe KO compared with WT mice on serum transferrin saturation (Tf Sat), and LIC were also determined as
an IR diet (Figure 3, white bars). described in Figure 1.
October 2009 HFE AND BMP SIGNALING 1493

Figure 4. Dietary iron appropri-


ately up-regulates hepatic Bmp6
mRNA expression in Hfe KO
mice. (A) WT and Hfe KO mice
on an ID, CTR, or IR diet from
Figure 2 (N ⫽ 6 per group) were
analyzed for Bmp6 relative to
Rpl19 mRNA expression as in
Figure 1. (B) Ratio of Bmp6
mRNA from panel A relative to
LIC. Results are expressed as the
mean ⫾ SD for the fold change
from WT mice on a CTR diet. Ex-
act P values are shown. (B) P val-
ues determined by 2-way ANOVA
were not significant for compari-
sons among all groups.

relates with LIC and is increased appropriately relative to mRNA expression was approximately 40% lower in Hfe
body iron burden in Hfe KO mice. KO compared with WT hepatocytes, consistent with
prior studies.37 Low doses of BMP6 ranging from 0.5 to
Intracellular Mediators and Targets of BMP6 2 ng/mL significantly increased Hamp1 mRNA expression
Signaling in the Liver Are Not Increased up to 2-fold in WT primary hepatocyte cultures (Figure 8,
Appropriately Relative to Iron Burden and
Bmp6 mRNA Expression in Hfe KO Mice closed rhombus). However, Hamp1 mRNA induction by
BMP6 was significantly lower in Hfe KO hepatocytes
Dietary iron also significantly modulated hepatic (Figure 8, open squares) compared with WT hepatocytes
P-Smad 1/5/8 protein (Figure 5A and B), Id1 mRNA treated with identical BMP6 concentrations.
(Figure 6A), and Hamp1 mRNA expression (Figure 7A) in
Hfe KO mice. The latter results are consistent with a prior
study showing that hepcidin levels can be modulated by Discussion
dietary iron in Hfe KO mice.48 However, the levels of Mutations in the gene encoding HFE are the most
P-Smad 1/5/8 protein relative to body iron burden (LIC) common cause of HH.2 Recent data suggest that one
were reduced significantly by approximately 30%– 40% in mechanism by which HFE mutations cause HH is by

PANCREAS, AND
Hfe KO compared with WT mice across all diets (Figure causing inappropriately low expression of the key iron

BILIARY TRACT
BASIC–LIVER,
5C), and there was a similar trend toward reduced regulatory hormone hepcidin.18 –23 However, the mecha-
P-Smad1/5/8 protein relative to Bmp6 mRNA in Hfe KO nisms by which HFE affects hepcidin expression still are
vs WT mice that reached significance for the IR diet unknown. We and others recently showed that BMP–
group (Figure 5D). Id1 mRNA expression relative to LIC SMAD signaling is a main regulatory pathway acting in
or to Bmp6 mRNA expression was reduced even more hepcidin regulation and iron metabolism.31,32,36 – 41,44 – 45
significantly in Hfe KO compared with WT mice by ap- We therefore investigated whether HFE was involved in
proximately 65% across all diets (Figure 6B and C). the BMP–SMAD signaling pathway.
Hamp1 mRNA expression relative to LIC and Bmp6 In the present work we show that Hfe KO mice have
mRNA levels reinforced the well-known inappropriately higher hepatic Bmp6 mRNA expression than WT mice.
low hepcidin levels in Hfe KO mice (Figure 7B and C). Furthermore, we show that dietary iron modulates he-
These results suggest that although hepatic P-Smad1/5/8 patic Bmp6 mRNA levels in both WT and Hfe KO mice in
protein, Id1 mRNA, and Hamp1 mRNA can be regulated an equivalent manner. Similar modulation of hepatic
by dietary iron, they are all inappropriately low relative to Bmp6 mRNA by dietary iron content, and a similar cor-
iron burden and hepatic Bmp6 mRNA levels in Hfe KO relation between hepatic Bmp6 mRNA levels and liver
mice. iron content, was seen previously in 2 other mouse
strains.45 These data suggest that the increased hepatic
BMP6 Induction of Hepcidin Expression Is Bmp6 mRNA in Hfe KO mice is appropriate for the
Impaired in Hfe KO Mouse Hepatocytes increased iron burden, and that HFE is not necessary for
To further confirm whether HFE plays a role in the regulation of Bmp6 mRNA levels in response to iron.
hepcidin induction by BMP6, we tested the ability of We did not find any changes in hepatic Bmp2 and Bmp4
BMP6 to induce hepcidin expression in primary hepato- mRNA levels in Hfe KO compared with WT mice, despite
cyte cultures from Hfe KO vs WT mice. Basal Hamp1 significant iron overload. We also did not find any
1494 CORRADINI ET AL GASTROENTEROLOGY Vol. 137, No. 4

Figure 5. In Hfe KO mice, di-


etary iron modulates hepatic
P-Smad1/5/8 protein expression,
but P-Smad1/5/8 is not increased
appropriately relative to iron bur-
den compared with WT mice. (A
and B) WT and Hfe KO mice on an
ID, CTR, or IR diet from Figure 2
(N ⫽ 6 per group) were analyzed
for hepatic P-Smad1/5/8 relative
to total Smad1 expression by (A,
representative blot) Western blot
(B) quantitated by IPLab Spec-
trum software. (C and D) Ratio of
P-Smad1/5/8 from panel B rela-
tive to (C) LIC or (D) hepatic Bmp6
mRNA. Results are expressed as
the mean ⫾ SD for the fold
change from WT mice on a CTR
diet. Exact P values determined
PANCREAS, AND
BILIARY TRACT

by 2-way ANOVA are shown.


BASIC–LIVER,

changes in hepatic Bmp2 and Bmp4 mRNA levels in Hfe rum iron, transferrin saturation, and LIC, that is, WT
KO or WT mice maintained on an IR diet compared with mice on an IR diet vs Hfe KO on an IR diet, we found that
their own control diet groups. These data are in agree- Hfe KO mice had significantly lower hepatic P-Smad1/
ment with a prior study showing that hepatic Bmp2 5/8 protein and Id1 mRNA levels compared with WT
mRNA was increased only under extreme iron overload mice. Furthermore, we found that across all dietary iron
conditions and Bmp4 mRNA was not altered by dietary groups, Hfe KO mice had significantly lower hepatic
iron in 2 other mouse strains.45 Together with the recent P-Smad1/5/8 protein and Id1 mRNA levels relative to
finding that BMP6 is a ligand for the BMP co-receptor LIC, significantly lower Id1 relative to Bmp6 mRNA, and
hemojuvelin,41 and that Bmp6 null mice have an iron- a trend toward lower P-Smad1/5/8 protein relative to
overload phenotype that resembles juvenile hemochro- Bmp6 mRNA that reached statistical significance for the
matosis owing to hemojuvelin mutations,41,44 these data IR diet, compared with WT mice. These data suggest that
reinforce the idea of the preeminent role of endogenous the downstream signals of BMP6 triggered by iron are
BMP6 in iron metabolism in vivo. impaired in Hfe KO mice. The impairment in Hfe KO
In contrast to the findings for Bmp6 mRNA expression, mice was more pronounced for Id1 mRNA compared with
hepatic P-Smad 1/5/8 protein and Id1 mRNA levels are P-Smad1/5/8 expression, and even more pronounced for
the same in Hfe KO animals and WT animals on a Hamp1 mRNA expression. These findings can be ex-
standard iron diet, and therefore inappropriately low plained in several ways. First, the kinetics of SMAD
relative to the higher body iron and Bmp6 mRNA levels in phosphorylation in response to BMP ligand is much
the Hfe KO mice. Indeed, when we examined WT vs Hfe faster and more transient compared with the regulation
KO mice with comparable Bmp6 mRNA expression, se- of downstream mRNAs. It therefore may be more diffi-
October 2009 HFE AND BMP SIGNALING 1495

Figure 6. In Hfe KO mice, dietary iron modulates hepatic Id1 mRNA expression, but Id1 mRNA is not increased appropriately relative to iron burden
and Bmp6 mRNA expression compared with WT mice. (A) WT and Hfe KO mice on an ID, CTR, or IR diet from Figure 2 (N ⫽ 6 per group) were
analyzed for Id1 relative to Rpl19 mRNA as in Figure 1. (B and C) Ratio of Id1 mRNA from panel A relative to (B) LIC or (C) hepatic Bmp6 mRNA.
Results are expressed as the mean ⫾ SD for the fold change from WT mice on a CTR diet. Exact P values determined by 2-way ANOVA are shown.

cult to capture the transient SMAD phosphorylation that HFE also affects hepcidin expression through an
state when using a model looking at the effects of additional non–BMP–SMAD pathway.
chronic changes in dietary iron levels and iron burden. The involvement of HFE in BMP6-mediated induction
Second, there may be a cascade effect through the BMP– of hepcidin expression is supported further by the rela-
SMAD signaling pathway that is able to amplify the tive resistance of Hfe KO primary hepatocyte cultures to
impairment step by step. In support of one or both of Hamp1 mRNA induction by BMP6 ligand. It is unlikely
these hypotheses, the modulation of P-Smad1/5/8 by that the iron-loaded state of these Hfe KO hepatocytes
iron in WT mice was less pronounced than the modula- per se impairs BMP–SMAD signaling because WT mice
tion of Id1 mRNA and Hamp1 mRNA. Another possibility with higher LIC owing to dietary iron loading had in-
is that HFE may affect the BMP signaling pathway down- creased hepatic P-Smad1/5/8 protein and Id1 mRNA lev-
stream of SMAD phosphorylation. Finally, it is possible els appropriate for their increased Bmp6 mRNA levels.

PANCREAS, AND
BILIARY TRACT
BASIC–LIVER,

Figure 7. In Hfe KO mice, dietary iron modulates hepatic Hamp1 mRNA expression, but Hamp1 mRNA is not increased appropriately relative to iron
burden and Bmp6 mRNA expression compared with WT mice. (A) WT and Hfe KO mice on an ID, CTR, or IR diet from Figure 2 (N ⫽ 6 per group)
were analyzed for Hamp1 relative to Rpl19 mRNA as in Figure 1. (B and C) Ratio of Hamp1 mRNA from panel A relative to (B) LIC or (C) hepatic Bmp6
mRNA. Results are expressed as the mean ⫾ SD for the fold change from WT mice on a CTR diet. Exact P values determined by 2-way ANOVA are
shown.
1496 CORRADINI ET AL GASTROENTEROLOGY Vol. 137, No. 4

expression.28,29 We previously showed that BMP6 signal-


ing mediated by the co-receptor hemojuvelin in complex
with BMP type I and type II receptors causes phosphor-
ylation of SMAD proteins, which in turn increases
transcription of hepcidin mRNA.31,32,39,49 Based on our
data here, we propose that HFE increases hepcidin ex-
pression through an interaction with the BMP6/hemoju-
velin/SMAD signaling pathway. Future studies will be
needed to determine the precise molecular mechanisms
by which HFE affects downstream BMP6 signals in hepa-
tocytes and whether TfR2 plays a role in this process.

References
1. Pietrangelo A. Hereditary hemochromatosis—a new look at an
old disease. N Engl J Med 2004;350:2383–2397.
2. Merryweather-Clarke AT, Pointon JJ, Shearman JD, et al. Global
prevalence of putative haemochromatosis mutations. J Med
Figure 8. Hepcidin induction by BMP6 is impaired in Hfe KO hepato- Genet 1997;34:275–278.
cytes. Primary hepatocyte cultures from Hfe KO vs WT control mice were 3. Zhou XY, Tomatsu S, Fleming RE, et al. HFE gene knockout
treated with low doses of exogenous BMP6 ligand ranging from 0.1 to 2.0 produces mouse model of hereditary hemochromatosis. Proc
ng/mL. Hamp1 relative to Gapdh mRNA was analyzed by quantitative Natl Acad Sci U S A 1998;95:2492–2497.
real-time reverse-transcription polymerase chain reaction. Results are ex- 4. Levy JE, Montross LK, Cohen DE, et al. The C282Y mutation
pressed as the mean ⫾ SD for the fold increase over untreated cells from causing hereditary hemochromatosis does not produce a null
5 independent experiments each conducted in triplicate. *P ⬍ .05, **P ⬍ allele. Blood 1999;94:9 –11.
.01, ***P ⬍ .001 compared with untreated cells for a given genotype; #P ⬍ 5. Bahram S, Gilfillan S, Kühn LC, et al. Experimental hemochroma-
.05, ##P ⬍ .01 for Hfe KO compared with WT cells treated with identical tosis due to MHC class I HFE deficiency: immune status and iron
concentrations of BMP6 as determined by ANOVA. metabolism. Proc Natl Acad Sci U S A 1999;96:13312–13317.
6. Feder JN, Gnirke A, Thomas W, et al. A novel MHC class I-like
gene is mutated in patients with hereditary haemochromatosis.
Interestingly, a prior study showed that Hfe KO primary Nat Genet 1996;13:399 – 408.
mouse hepatocytes showed equivalent increases in Hamp 7. Feder JN, Tsuchihashi Z, Irrinki A, et al. The hemochromatosis
founder mutation in HLA-H disrupts beta2-microglobulin interaction
mRNA expression in response to BMP2, BMP4, and and cell surface expression. J Biol Chem 1997;272:14025–14028.
BMP9 ligands compared with WT hepatocytes.37 One 8. Waheed A, Parkkila S, Zhou XY, et al. Hereditary hemochromato-
explanation for these findings is that we used relatively sis: effects of C282Y and H63D mutations on association with
low doses of BMP6 ligand, whereas Truksa et al37 used beta2-microglobulin, intracellular processing, and cell surface
relatively high doses of BMP ligands. We hypothesize expression of the HFE protein in COS-7 cells. Proc Natl Acad Sci
U S A 1997;94:12384 –12389.
PANCREAS, AND

that low doses of endogenous BMP6 ligand are expressed


BILIARY TRACT

9. Parkkila S, Waheed A, Britton RS, et al. Association of the trans-


BASIC–LIVER,

under normal physiologic conditions, and that HFE is ferrin receptor in human placenta with HFE, the protein defective
important for the downstream signals to BMP6 under in hereditary hemochromatosis. Proc Natl Acad Sci U S A 1997;
these physiologic conditions. Under nonphysiologic con- 94:13198 –13202.
ditions of high levels of exogenous BMP ligand, the large 10. Feder JN, Penny DM, Irrinki A, et al. The hemochromatosis gene
product complexes with the transferrin receptor and lowers its
excess of BMP ligand can bypass the role of HFE in affinity for ligand binding. Proc Natl Acad Sci U S A 1998;95:
optimizing the BMP6 response via BMP type I and type II 1472–1477.
receptors and the co-receptor hemojuvelin. In addition, 11. Waheed A, Parkkila S, Saarnio J, et al. Association of HFE protein
the role of HFE in regulating downstream BMP signals with transferrin receptor in crypt enterocytes of human duode-
might be specific to BMP6 ligand. Further experiments num. Proc Natl Acad Sci U S A 1999;96:1579 –1584.
12. Lebrón JA, West AP Jr, Bjorkman PJ. The hemochromatosis pro-
will be needed to determine the ligand specificity of HFE tein HFE competes with transferrin for binding to the transferrin
involvement in the BMP pathway. receptor. J Mol Biol 1999;294:239 –245.
In summary, we propose the following model for the 13. Bennett MJ, Lebrón JA, Bjorkman PJ. Crystal structure of the
role of HFE in iron metabolism. Our data, in conjunction hereditary haemochromatosis protein HFE complexed with trans-
with the data of others,28,45 suggest that iron sensing ferrin receptor. Nature 2000;403:46 –53.
14. West AP Jr, Giannetti AM, Herr AB, et al. Mutational analysis of
occurs at multiple levels. First, chronic iron loading in- the transferrin receptor reveals overlapping HFE and transferrin
creases hepatic Bmp6 mRNA. The mechanism for this binding sites. J Mol Biol 2001;313:385–397.
up-regulation remains to be determined; however, it does 15. Giannetti AM, Björkman PJ. HFE and transferrin directly compete
not appear to require HFE. Second, recent work suggests for transferrin receptor in solution and at the cell surface. J Biol
that HFE may participate in the sensing of increased Chem 2004;279:25866 –25875.
16. Goswami T, Andrews NC. Hereditary hemochromatosis protein,
serum iron levels by becoming displaced from TfR1 by HFE, interaction with transferrin receptor 2 suggests a molecular
holotransferrin and by interacting with a TfR2/trans- mechanism for mammalian iron sensing. J Biol Chem 2006;281:
ferrin complex, which then acts to increase hepcidin 28494 –28498.
October 2009 HFE AND BMP SIGNALING 1497

17. Chen J, Chloupkova M, Gao J, et al. HFE modulates transferrin Hfe, transferrin receptor 2 (Tfr2), and IL-6. Proc Natl Acad Sci
receptor 2 levels in hepatoma cells via interactions that differ U S A 2006;103:10289 –10293.
from transferrin receptor 1-HFE interactions. J Biol Chem 2007; 38. Truksa J, Peng H, Lee P, et al. Different regulatory elements are
282:36862–36870. required for response of hepcidin to interleukin-6 and bone mor-
18. Vujic Spasic M, Kiss J, Herrmann T, et al. Hfe acts in hepatocytes phogenetic proteins 4 and 9. Br J Haematol 2007;139:138 –
to prevent hemochromatosis. Cell Metab 2008;7:173–178. 147.
19. Ahmad KA, Ahmann JR, Migas MC, et al. Decreased liver hepcidin 39. Verga Falzacappa MV, Casanovas G, Hentze MW, et al. A bone
expression in the Hfe knockout mouse. Blood Cells Mol Dis morphogenetic protein (BMP)-responsive element in the hepcidin
2002;29:361–366. promoter controls HFE2-mediated hepatic hepcidin expression
20. Nicolas G, Viatte L, Lou DQ, et al. Constitutive hepcidin expres- and its response to IL-6 in cultured cells. J Mol Med 2008;86:
sion prevents iron overload in a mouse model of hemochroma- 531–540.
tosis. Nat Genet 2003;34:97–101. 40. Yu PB, Hong CC, Sachidanandan C, et al. Dorsomorphin inhibits
21. Muckenthaler M, Roy CN, Custodio AO, et al. Regulatory defects BMP signals required for embryogenesis and iron metabolism.
in liver and intestine implicate abnormal hepcidin and Cybrd1 Nat Chem Biol 2008;4:33– 41.
expression in mouse hemochromatosis. Nat Genet 2003;34: 41. Andriopoulos B Jr, Corradini E, Xia Y, et al. BMP6 is a key
102–107. endogenous regulator of hepcidin expression and iron metabo-
22. Bridle KR, Frazer DM, Wilkins SJ, et al. Disrupted hepcidin regu- lism. Nat Genet 2009;41:482– 487.
lation in HFE-associated haemochromatosis and the liver as a 42. Huang FW, Pinkus JL, Pinkus GS, et al. A mouse model of juvenile
regulator of body iron homoeostasis. Lancet 2003;361:669 – hemochromatosis. J Clin Invest 2005;115:2187–2191.
673. 43. Niederkofler V, Salie R, Arber S. Hemojuvelin is essential for
23. Piperno A, Girelli D, Nemeth E, et al. Blunted hepcidin response dietary iron sensing, and its mutation leads to severe iron over-
to oral iron challenge in HFE-related hemochromatosis. Blood load. J Clin Invest 2005;115:2180 –2186.
2007;110:4096 – 4100. 44. Meynard D, Kautz L, Darnaud V, et al. Lack of the bone morpho-
24. Pigeon C, Ilyin G, Courselaud B, et al. A new mouse liver-specific genetic protein BMP6 induces massive iron overload. Nat Genet
gene, encoding a protein homologous to human antimicrobial 2009;41:478 – 481.
peptide hepcidin, is overexpressed during iron overload. J Biol 45. Kautz L, Meynard D, Monnier A, et al. Iron regulates phosphory-
Chem 2001;276:7811–7819. lation of Smad1/5/8 and gene expression of Bmp6, Smad7, Id1,
25. Krause A, Neitz S, Magert HJ, et al. LEAP-1, a novel highly and Atoh8 in the mouse liver. Blood 2008;112:1503–1509.
46. Torrance JD, Bothwell TH. Tissue iron stores. In: Cook JD, ed.
disulfide-bonded human peptide, exhibits antimicrobial activity.
Methods in hematology. Volume 1. New York: Churchill Living-
FEBS Lett 2001;480:147–150.
stone, 1980:90 –115.
26. Park CH, Valore EV, Waring AJ, et al. Hepcidin, a urinary antimi-
47. Montosi G, Corradini E, Garuti C, et al. Kupffer cells and macro-
crobial peptide synthesized in the liver. J Biol Chem 2001;276:
phages are not required for hepatic hepcidin activation during
7806 –7810.
iron overload. Hepatology 2005;41:545–552.
27. Nemeth E, Tuttle MS, Powelson J, et al. Hepcidin regulates
48. Gehrke SG, Herrmann T, Kulaksiz H, et al. Iron stores modulate
cellular iron efflux by binding to ferroportin and inducing its inter-
hepatic hepcidin expression by an HFE-independent pathway.
nalization. Science 2004;306:2090 –2093.
Digestion 2005;72:25–32.
28. Schmidt PJ, Toran PT, Giannetti AM, et al. The transferrin recep-
49. Xia Y, Babitt JL, Sidis Y, et al. Hemojuvelin regulates hepcidin
tor modulates Hfe-dependent regulation of hepcidin expression.
expression via a selective subset of BMP ligands and receptors
Cell Metab 2008;7:205–214.
independently of neogenin. Blood 2008;111:5195–5204.
29. Gao J, Chen J, Kramer M, et al. Interaction of the hereditary

PANCREAS, AND
BILIARY TRACT
BASIC–LIVER,
hemochromatosis protein HFE with transferrin receptor 2 is re-
quired for transferrin-induced hepcidin expression. Cell Metab Received March 14, 2009. Accepted June 24, 2009.
2009;9:217–227.
30. Papanikolaou G, Samuels ME, Ludwig EH, et al. Mutations in Reprint requests
HFE2 cause iron overload in chromosome 1q-linked juvenile Address requests for reprints to: Dr Jodie L. Babitt, MD, Program
hemochromatosis. Nat Genet 2004;36:77– 82. in Membrane Biology, Division of Nephrology, Center for Systems
31. Babitt JL, Huang FW, Wrighting DM, et al. Bone morphogenetic Biology, Massachusetts General Hospital, 185 Cambridge Street,
protein signaling by hemojuvelin regulates hepcidin expression. CPZN-8216, Boston, Massachusetts 02114. e-mail: babitt.
Nat Genet 2006;38:531–539. jodie@mgh.harvard.edu; fax: (617) 643-3182.
32. Babitt JL, Huang FW, Xia Y, et al. Modulation of bone morphoge-
netic protein signaling in vivo regulates systemic iron balance. Acknowledgments
J Clin Invest 2007;117:1933–1939. A.P. and J.L.B. contributed equally to this article.
33. Shi Y, Massague J. Mechanisms of TGF-beta signaling from cell
membrane to the nucleus. Cell 2003;113:685–700. Conflict of interest
34. Miyazono K, Miyazawa K. Id: a target of BMP signaling. Sci STKE The authors disclose no conflicts.
2002;2002:PE40.
35. Korchynskyi O, Ten Dijke P. Identification and functional charac- Funding
terization of distinct critically important bone morphogenetic pro- Antonello Pietrangelo and Elena Corradini were supported in part
tein-specific response elements in the Id1 promoter. J Biol Chem by the European Union (EUROIRON Ct. 2006-037296) and the
2002;277:4883– 4891. Italian Ministry of University and Research (MIUR-2006) grants;
36. Wang RH, Li C, Xu X, et al. A role of SMAD4 in iron metabolism Herbert Y. Lin was supported in part by National Institutes of Health
through the positive regulation of hepcidin expression. Cell grants RO1 DK-069533 and RO1 DK-071837; and Jodie L. Babitt
Metab 2005;2:399 – 409. was supported in part by National Institutes of Health grant K08
37. Truksa J, Peng H, Lee P, et al. Bone morphogenetic proteins 2, 4, DK-075846 and by a Claflin Distinguished Scholar Award from the
and 9 stimulate murine hepcidin 1 expression independently of Massachusetts General Hospital.

You might also like