scholarly journals An Inflammatory Pathway Mediating Rapid Hepcidin-Independent Hypoferremia

Blood ◽  
2014 ◽  
Vol 124 (21) ◽  
pp. 214-214
Author(s):  
Martina U. Muckenthaler ◽  
Claudia Guida ◽  
Sandro Altamura ◽  
Felix A. Klein ◽  
Bruno Galy ◽  
...  

Abstract Hypoferremia represents an innate immune response to infection and inflammation sequestering iron from pathogens. The iron-hormone hepcidin is induced by such stimuli, causing degradation of the iron exporter ferroportin (Fpn) and reduced iron release from macrophages, suggesting that hepcidin is the crucial effector of inflammatory hypoferremia. Here we report the discovery of a fast, hepcidin-independent hypoferremia pathway. Stimulation of the toll-like receptor (TLR) 2 and TLR6 triggers profound decreases in Fpn mRNA and protein expression in bone marrow-derived macrophages, liver and spleen of mice without changing hepcidin expression. Furthermore, C326S Fpn knock-in mice with a disrupted hepcidin/Fpn regulatory circuitry respond to injection of the TLR2/6 ligand FSL1 by Fpn down regulation and a reduction of serum iron levels. Our findings challenge the prevailing role of hepcidin in hypoferremia, uncovering a rapid and potent inflammatory response pathway. Disclosures No relevant conflicts of interest to declare.

2016 ◽  
Vol 8 (5) ◽  
pp. 517-528 ◽  
Author(s):  
Andrew E. Armitage ◽  
Pei Jin Lim ◽  
Joe N. Frost ◽  
Sant-Rayn Pasricha ◽  
Elizabeth J. Soilleux ◽  
...  

Withdrawal of iron from serum (hypoferraemia) is a conserved innate immune antimicrobial strategy that can withhold this critical nutrient from invading pathogens, impairing their growth. Hepcidin (Hamp1) is the master regulator of iron and its expression is induced by inflammation. Mice lacking Hamp1 from birth rapidly accumulate iron and are susceptible to infection by blood-dwelling siderophilic bacteria such as Vibrio vulnificus. In order to study the innate immune role of hepcidin against a background of normal iron status, we developed a transgenic mouse model of tamoxifen-sensitive conditional Hamp1 deletion (termed iHamp1-KO mice). These mice attain adulthood with an iron status indistinguishable from littermate controls. Hamp1 disruption and the consequent decline of serum hepcidin concentrations occurred within hours of a single tamoxifen dose. We found that the TLR ligands LPS and Pam3CSK4 and heat-killed Brucella abortus caused an equivalent induction of inflammation in control and iHamp1-KO mice. Pam3CSK4 and B. abortus only caused a drop in serum iron in control mice, while hypoferraemia due to LPS was evident but substantially blunted in iHamp1-KO mice. Our results characterise a powerful new model of rapidly inducible hepcidin disruption, and demonstrate the critical contribution of hepcidin to the hypoferraemia of inflammation.


2020 ◽  
Author(s):  
fali zhang ◽  
peng zhao ◽  
zhongming Qian ◽  
mingkang zhong

Abstract BackgroundLPS triggers inflammation response in periphery, whether the infection in CNS induced by LPS ICV injection affected the peripheral iron metabolism was unknown , The current study was to find out whether LPS injected to the brain could regulate hepcidin expression in liver and peripheral iron metabolism. MethodsWide type mice (IL-6+/+) and IL-6-/- mice of 8-week-old were performed on ICV injection with LPS. After 6h, hepcidin expression in liver, as well as serum iron and transferrin saturation was detected and calculated, we also tested the IL-6/JAK2/STAT3 pathway in hepcidin regulation in liver of IL-6 knockout (IL-6-/- mice) and IL-6+/+ mice, AG490 as an inhibitor of JAK2 was used to confirm the effect of IL-6/JAK2/STAT3 pathway on hepcidin expression in liver. ResultsHepcidin mRNA, IL-6 mRNA and protein expression in the liver of IL-6-/- mice was significantly lower than IL-6+/+ mice after LPS administration. IL-6 deficiency abolished the decrease of serum iron, transferrin saturation induced by LPS injection. IL-6 deficiency also abolished the decrease of Fpn1, increase of pSTAT3 and Ft-L protein in liver. AG490 significantly reduced the pSTAT3 protein and abolished the changes of Fpn1 and Ft-L expression induced by LPS in liver. ConclusionThese finding provided further evidence that the effect of central inflammation on the hepatic hepcidin expression and peripheral iron metabolism.


Blood ◽  
2009 ◽  
Vol 114 (9) ◽  
pp. 1913-1918 ◽  
Author(s):  
Curry L. Koening ◽  
Jennifer C. Miller ◽  
Jenifer M. Nelson ◽  
Diane M. Ward ◽  
James P. Kushner ◽  
...  

Hepcidin is the major regulator of systemic iron homeostasis in mammals. Hepcidin is produced mainly by the liver and is increased by inflammation, leading to hypoferremia. We measured serum levels of bioactive hepcidin and its effects on serum iron levels in mice infected with Borrelia burgdorferi. Bioactive hepcidin was elevated in the serum of mice resulting in hypoferremia. Infected mice produced hepcidin in both liver and spleen. Both intact and sonicated B burgdorferi induced hepcidin expression in cultured mouse bone marrrow macrophages. Hepcidin production by cultured macrophages represents a primary transcriptional response stimulated by B burgdorferi and not a secondary consequence of cytokine elaboration. Hepcidin expression induced by B burgdorferi was mediated primarily by activation of Toll-like receptor 2.


Blood ◽  
2011 ◽  
Vol 118 (21) ◽  
pp. 3182-3182
Author(s):  
Srila Gopal ◽  
Rao Madhumathi ◽  
Damien R Ashby

Abstract Abstract 3182 Methods: We carried out a prospective study of 167 patients with CKD stages 1 to 4 followed over a two year period. Using a competitive radioimmunoassay, we measured plasma hepcidin at baseline and studied its association to baseline clinical parameters, as well as the development of anemia and need for ESA over a two year follow up period. Exclusion criteria included any erythropoietin stimulating agent (ESA) therapy at baseline. Variables were log transformed to satisfy normality assumptions. Results: Median (P5-P95) Glomerular filtration rate (GFR) was 42 (20–109) ml/min and 45% were anemic at the time of enrollment by KDOQI criteria, 11 % (18/167) having severe anemia. In patients with CKD, Hepcidin was positively correlated with serum iron (Spearmans rho,p:serum iron,0.22,0.005) and negatively correlated to EPO levels (Spearmans rho,p:-0.23,0.003 vss.-0.55,0.002). Hepcidin levels at baseline were not significantly associated with GFR, hemoglobin or CRP, and did not differ according to diabetes or ethnicity. Though Hepcidin levels did not differ by the presence of anemia at baseline, they were significantly higher among patients who developed severe anemia (n=23) at the end of the first year (40 ng/mL vs. 25 ng/mL, p<0.01). A total of 13 patients initiated ESA during follow up; a 2-fold higher hepcidin was associated with a hazard ratio of 2.25 (95% CI 0.89 to 5.62) for the need to initiate ESA therapy (Fig 1), adjusted for GFR. Conclusion: High hepcidin, unrelated to inflammation, is a predictor for the development of anemia and the need for initiation of ESA therapy in CKD patients. The role of Hepcidin as a therapeutic target in the anemia of CKD needs further study. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2012 ◽  
Vol 120 (21) ◽  
pp. 4420-4420
Author(s):  
Liu Xiaoli ◽  
Bintao Huang ◽  
Qingfeng Du ◽  
Jinfang Zhang ◽  
Na Xu ◽  
...  

Abstract Abstract 4420 Objective: The purpose was to investigate the role of the EphB4 in imatinib (IM) resistance and the mechanism why the homoharringtonine (HHT)+IM regimen gained more treatment profits than simple HHT or IM treating myeloid leukemia. Method: The stable under-expressing EphB4 cells (K562-R-EphB4-sh) were obtained. The cell viability and IC50 under the incubation of IM or HHT+IM was tested by MTT. PE Annexin V apoptosis detected the apoptosis rate of K562-R cells. Subcutaneous K562 xenograft models were established. The activated signal proteins in cells and tissues such as RhoA, MEK and ERK were tested by Western blot. Result: K562-R-EphB4-sh cell and xenograft were sensitivity to IM. Activated RhoA was not involved in K562-R-EphB4-sh cell and xenograft tissue. But phosphorylation of MEK/ERK was overexpression in K562-R-EphB4-sh cell and tissue. The apoptosis rate reached 58.71 ± 2.39% with K562-R cell incubated with HHT+IM, which was higher to K562-R cell incubated with IM (P=0.002). IC50 of K562-R cell incubated by IM was 5.45 mg/L. But under the stimulation of HHT+IM, IC50 of K562-R decreased from 5.45 to 1.17 mg/L (P<0.001). K562-R xenograft volumes significantly decreased with IM+HHT treatment comparing with before treatment (1692.82 ± 317.14 mm3 versus 975.56 ± 132.42 mm3, P<0.001). HHT blocked the expressions of EphB4/RhoA in K562-R cell and xenograft, but HHT cannot down-regulate the expression of P- MEK/ERK. Conclusions: A new marker of IM resistance mediated by the activation of EphB4/RhoA pathway. HHT+IM regimen gained more treatment profits than simple HHT or IM treating myeloid leukemia by blocking EphB4/RhoA pathway in Ph+ myeloid leukemia. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2012 ◽  
Vol 120 (21) ◽  
pp. 611-611
Author(s):  
Josephine Volovetz ◽  
Aileen W. Zhen ◽  
Vera Gaun ◽  
Bonnie Patchen ◽  
Paula G. Fraenkel

Abstract Abstract 611 Hepcidin, a peptide hormone produced in the liver, decreases intestinal iron absorption and macrophage iron release via effects on ferroportin. Hepcidin is a potential drug target for patients with iron overload syndromes because its levels are inappropriately low in these individuals. To generate a tool for identifying small molecules that modulate Hepcidin expression, we stably transfected human hepatocytes (HepG2) cells with a reporter construct containing 3 kilobases of the human Hepcidin promoter upstream of a firefly reporter gene. We then used high throughput methods to screen 10,360 chemicals in duplicate from the Harvard Institute of Chemistry and Cell Biology library for their effect on Hepcidin expression and cell viability. Regulators were identified as chemicals that caused a change >3 standard deviations above or >1.5 standard deviations below the mean of the other chemicals (z-score >3 or <-1.5), while not adversely affecting cell viability, quantified by a nonlytic fluorescence assay. Using these criteria, we identified 32 small molecules that upregulated and 3 that downregulated Hepcidin expression. Functional classification of the positive regulators indicated: 4 anti-inflammatory agents, 4 antimicrobials, 6 antineoplastic drugs, 6 kinase inhibitors, and 12 with other or unknown function. Of the positive modulators, two were flavones, consistent with our prior discovery that the isoflavone genistein upregulates Hepcidin expression. Of the negative regulators, one was a kinase inhibitor and two were of unknown function. Experiments are underway to characterize the mechanism of action of these regulators. The best candidates will subsequently be tested in mouse models of iron overload syndromes with the intention of developing new therapies for diseases in which Hepcidin is inappropriately regulated. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2009 ◽  
Vol 114 (22) ◽  
pp. SCI-25-SCI-25
Author(s):  
Jodie L. Babitt

Abstract Abstract SCI-25 Systemic iron balance is regulated by the key iron regulatory hormone hepcidin. Secreted by the liver, hepcidin inhibits iron absorption from the diet and iron mobilization from body stores by decreasing cell surface expression of the iron export protein ferroportin. Iron administration increases hepcidin expression, thereby providing a feedback mechanism to limit further iron absorption, while anemia and hypoxia inhibit hepcidin expression, thereby increasing iron availability for erythropoiesis. Hepcidin excess is thought to have a role in the anemia of inflammation, while hepcidin deficiency is thought to be the common pathogenic mechanism underlying the iron overload disorder hereditary hemochromatosis, due to mutations in the genes encoding hepcidin itself (HAMP), HFE, transferrin receptor 2 (TFR2), or hemojuvelin (HFE2). Notably the precise molecular mechanisms by which iron levels are “sensed” and how this iron “signal” is transduced to modulate hepcidin expression have remained elusive. We have recently discovered that hemojuvelin is a co-receptor for the bone morphogenetic protein (BMP) signaling pathway, and that hemojuvelin-mediated BMP signals increase hepcidin expression at the transcriptional level. In addition to patients with HFE2 mutations and Hfe2 knockout mice, other genetic mouse models associated with impaired hepatic BMP signaling through a global knockout of the ligand Bmp6, or selective hepatic knockout of an intracellular mediator of BMP signaling, Smad4, also cause inappropriately low hepcidin expression and iron overload. Exogenous BMP6 administration in mice increases hepatic hepcidin expression and reduces serum iron, while BMP6 antagonists inhibit hepatic hepcidin expression, mobilize reticuloendothelial cell iron stores and increase serum iron. Not only does the BMP6-hemojuvelin-SMAD pathway regulate hepcidin expression and thereby systemic iron homeostasis, but also the BMP6-SMAD pathway itself is regulated by iron. Acute iron administration in mice increases phosphorylation of Smad proteins in the liver, and chronic changes in dietary iron modulate hepatic Bmp6 mRNA expression and phosphorylation of Smad proteins concordantly with Hamp mRNA expression. Together, these data support the paramount role of the BMP6-hemojuvelin-SMAD signaling pathway in the iron-mediated regulation of hepcidin expression and systemic iron homeostasis, and suggest that modulators of this pathway may be an alternative therapeutic strategy for treating disorders of iron homeostasis. Recent work elucidating the role of the BMP signaling pathway in hepcidin regulation and systemic iron homeostasis will be presented. Disclosures Babitt: Ferrumax Pharmaceuticals, Inc.: Equity Ownership.


Blood ◽  
2010 ◽  
Vol 116 (21) ◽  
pp. 4251-4251
Author(s):  
Pedro Ramos ◽  
Ella Guy ◽  
Robert W Grady ◽  
Maria de Sousa ◽  
Stefano Rivella

Abstract Abstract 4251 A deficient hepcidin response to iron is the principal mechanism responsible for increased iron uptake from the diet leading to iron overload. In hereditary hemochromatosis (HH), mutations in the HFE gene lead to iron overload through abnormally low levels of hepcidin. Interestingly, hepcidin has been shown to respond to a variety of stimuli, including iron, hypoxia, erythropoiesis and inflammation, requiring integration of the respective signals for its regulation. Further studies showed that HFE/Hfe could also modulate cellular iron uptake by associating with the transferrin receptor-1 (Tfrc), a crucial protein for iron uptake by erythroid cells. In addition, some studies have reported altered erythropoietic values in HH patients. Despite these findings, the role of Hfe in erythropoiesis was never explored. We hypothesized that Hfe influences erythropoiesis by two distinct mechanisms: 1) limiting hepcidin expression, thereby increasing iron availability, under conditions of simultaneous iron overload and stress erythropoiesis; 2) participating directly in the control of transferrin-bound iron uptake by erythroid cells. To test this hypothesis we investigated the role of Hfe in erythropoiesis, aiming to uncover the relative contribution of each of the aforementioned mechanisms. When erythropoiesis was challenged by phlebotomy, Hfe-KO animals were able to recover faster from anemia (p≤0.05) than either normal or iron overloaded wt mice. In Hfe-KO mice, despite their increased iron load, downregulation of hepcidin in response to phlebotomy or erythropoietin administration was comparable to that seen in wt mice. In contrast, iron overloaded wt mice showed increased hepcidin expression both at steady state and after erythropoietic stimulation compared to wt or Hfe-KO mice. In phlebotomized mice fed a standard diet, analysis of serum iron and transferrin saturation indicated that wt mice on the standard diet were able to increase their serum iron very rapidly. After 24 hours, both wt and Hfe-KO mice had similar serum iron and transferrin saturation levels. On the other hand, wt mice kept on an iron deficient diet over the course of phlebotomy, were unable to overcome the phlebotomy-induced anemia. In contrast, Hfe-KO mice fed the low iron diet were able to recover from anemia, although at a slower pace than either Hfe-KO or wt mice on a standard diet. These data indicate that gastrointestinal iron absorption in both wt and Hfe-KO mice is a major factor leading to recovery from anemia, although the excess iron in the liver of Hfe-KO mice contributes to restoration of the red blood cell reservoir. Phlebotomy is the main tool utilized to treat iron overload in HH patients. However, our data suggests that this treatment leads to both mobilization of iron from stores and increased gastrointestinal iron absorption. These observations suggest that patients might benefit from a controlled iron diet or from supplementation with hepcidin or an hepcidin agonist to limit iron absorption. Next, we determined that Hfe is expressed in erythroid cells and that it interacts with Tfrc in murine erythroleukemia cells. Moreover, we discovered that the level of Tfrc expression in Hfe-KO cells is 80% of that seen in wt cells, as measured by flow cytometry. This observation, together with measurement of iron uptake using 59Fe-saturated transferrin, indicated that Hfe-KO erythroid cells take up significantly more iron than wt cells. To confirm that Hfe plays a role in erythropoiesis independent from that in the liver, we transplanted Hfe-KO or wt bone marrow cells into lethally irradiated wt recipients and analyzed their recovery from phlebotomy. We observed that recovery from anemia was faster in Hfe→wt than in wt→wt and was associated with increased mean corpuscular hemoglobin levels, suggesting that lack of Hfe in the hematopoietic compartment can lead to increased hemoglobin production. In summary, our results indicate that lack of Hfe enhances iron availability for erythropoiesis by two distinct mechanisms. On the one hand, Hfe plays an important role in maintaining erythroid iron homeostasis by limiting the response of hepcidin to iron, particularly under conditions of erythropoietic stimulation. On the other hand, lack of Hfe contributes directly to increased iron intake by erythroid progenitors, even in the absence of iron overload. Disclosures: No relevant conflicts of interest to declare.


Sign in / Sign up

Export Citation Format

Share Document