scholarly journals Lack of IL6 Improves Recovery from Anemia of Inflammation Which Gets Hampered in Presence of Excess Iron

Blood ◽  
2021 ◽  
Vol 138 (Supplement 1) ◽  
pp. 3072-3072
Author(s):  
Sayantani Sinha ◽  
Ritama Gupta ◽  
Amaliris Guerra ◽  
Paige Mc Veigh ◽  
Sara Gardenghi ◽  
...  

Abstract Anemia of inflammation (AI) is the second most common anemia after iron deficiency anemia. The predominant regulators of AI are the cytokine interleukin 6 (IL6) and the hormone hepcidin (HAMP). IL6 is an inflammatory cytokine which also limits iron absorption by inducing HAMP, which promotes the degradation of the iron exporter ferroportin. We hypothesized that knocking down both HAMP and IL6 simultaneously will help us to understand if their mode of action in AI is uniquely limited to iron absorption and erythroid iron intake or if they also have independent roles. Henceforth, we generated IL6/HampKO (DKO) mice and, unexpectedly, observed that IL6KO mice showed the best recovery in bone marrow (BM) erythropoiesis (using flow cytometry analysis and looking at the absolute number of erythroid progenitors) after BA administration when compared to wild type (WT), HampKO and DKO mice. The best differences were observed at 14 days post BA administration. In contrast, the extramedullary erythropoiesis in the spleen was more pronounced in HampKO and DKO mice compared to WT and IL6KO animals, indicating that the mechanism impairing erythropoiesis in the BM did not affect erythroid progenitors in the spleen. These observations suggest that HAMP and IL6 proteins contribute independently to AI, with IL6 having some effect on the erythropoiesis in the BM independent from the IL6-HAMP axis leading to iron restriction. Furthermore, these observations raised the question why both HampKO and DKO mice showed reduced BM erythropoiesis compared to IL6KO animals. We investigated inflammatory cytokines and altered iron parameters as potential mediators of impaired erythropoiesis. We compared several inflammatory cytokines, including IL6, TNFa and INFg following BA administration: cytokine levels were elevated 6 hrs, reduced 48hrs after BA administration and moderately increased again two weeks later. Interestingly, among all the cytokines the levels of IL1b were significantly attenuated in IL6KO mice at day 14 compared to WT and HampKO animals. Moreover, transferrin saturation and NTBI levels were higher in HampKO and DKO animals compared to IL6KO mice. These observations strongly suggested that BM erythropoiesis is more sensitive to inflammatory insult in presence of an excess of iron, while extramedullary erythropoiesis is mildly affected and can eventually thrive under supra-physiological transferrin saturation levels. To test if increased iron affects BM erythropoiesis in presence of inflammation, we treated both WT and IL6KO mice with combination of iron dextran and BA. Both WT and IL6KO mice were treated with a combination of BA and iron at day 0 followed by alternate day of iron injections showed the poorest erythropoiesis in the BM and became rapidly sick, although the effect was significantly more pronounced in WT animals, as suggested by their survival curve. Since mycobacterium infections lead to NLPR3 inflammasome activation and Caspase1 upregulation (Marim et al. Semin Immunopathology 2017), we investigated how erythroid progenitors were affected. By flow cytometry analyses, we observed a significantly higher upregulation of the Caspase1 protein in WT and DKO mice compared to IL6KO animals. This was also reproduced by culturing WT or IL6KO BM progenitor erythroid cells in presence of mouse serum derived from WT or IL6KO mice treated with BA. Most importantly, IL6KO mice treated with BA and iron showed the highest levels of Caspase1 compared to only BA treated IL6KO mice, indicating that excess of iron abrogates the beneficial effect of IL6 deficiency on erythropoiesis under conditions of AI. Furthermore, using flow cytometry, we observed in WT mice treated with BA or BA and iron a significant increase in mitochondrial mass, which is an indicator of mitochondrial stress. The mitochondrial mass was reduced in IL6KO mice treated with BA, but again increased in IL6KO mice treated with BA and iron. We have also observed an increase of mitochondrial superoxide by confocal microscopy in WT mice compared to IL6KO mice treated with BA. Altogether, these data support a model where inflammation in presence of an excess of iron impairs BM erythropoiesis through mechanisms at least in part mediated by Caspase1 and mitochondrial dysfunction, while iron excess itself is sufficient to boost extramedullary erythropoiesis to compensate and sustain RBC production. Disclosures Vinchi: PharmaNutra: Research Funding; Vifor Pharma: Research Funding; Silence Therapeutics: Membership on an entity's Board of Directors or advisory committees, Research Funding; Novartis: Research Funding. Rivella: Ionis Pharmaceuticals: Consultancy; Meira GTx: Consultancy.

2002 ◽  
Vol 282 (4) ◽  
pp. G598-G607 ◽  
Author(s):  
Andreas Rolfs ◽  
Herbert L. Bonkovsky ◽  
James G. Kohlroser ◽  
Kristina McNeal ◽  
Ashish Sharma ◽  
...  

Hereditary hemochromatosis (HHC) is one of the most frequent genetic disorders in humans. In healthy individuals, absorption of iron in the intestine is tightly regulated by cells with the highest iron demand, in particular erythroid precursors. Cloning of intestinal iron transporter proteins provided new insight into mechanisms and regulation of intestinal iron absorption. The aim of this study was to assess whether, in humans, the two transporters are regulated in an iron-dependent manner and whether this regulation is disturbed in HHC. Using quantitative PCR, we measured mRNA expression of divalent cation transporter 1 (DCT1), iron-regulated gene 1 (IREG1), and hephaestin in duodenal biopsy samples of individuals with normal iron levels, iron-deficiency anemia, or iron overload. In controls, we found inverse relationships between the DCT1 splice form containing an iron-responsive element (IRE) and blood hemoglobin, serum transferrin saturation, or ferritin. Subjects with iron-deficiency anemia showed a significant increase in expression of the spliced form, DCT1(IRE) mRNA. Similarly, in subjects homozygous for the C282Y HFE mutation, DCT1(IRE) expression levels remained high despite high serum iron saturation. Furthermore, a significantly increased IREG1 expression was observed. Hephaestin did not exhibit a similar iron-dependent regulation. Our data show that expression levels of human DCT1 mRNA, and to a lesser extent IREG1 mRNA, are regulated in an iron-dependent manner, whereas mRNA of hephaestin is not affected. The lack of appropriate downregulation of apical and basolateral iron transporters in duodenum likely leads to excessive iron absorption in persons with HHC.


Blood ◽  
2008 ◽  
Vol 112 (11) ◽  
pp. 5387-5387
Author(s):  
Eun Sil Park ◽  
In-Suk Kim ◽  
Jae-Young Lim

Abstract Backgroud . The aims of this study were to measure the serum levels of prohepcidin in anemic and non anemic infants and to evaluate the correlation between prohepcidin and iron parameters and inflammatory cytokines. Procedure. One hundred and sixty nine patients aged from 6 months to 24 months were enrolled and divided into 4 groups according to hemoglobin and serum ferritin (SF). Routine hematologic labs, soluble transferrin receptor (sTfR), serum prohepcidin, C reactive protein (CRP), and interleukin-6 (IL-6) were assessed. The subgroup of anemia of inflammation (AI) was defined as Hb <11 g/dL and SF >50 μg/L, the subgroup of iron deficiency anemia (IDA) as Hb <11 g/dL and SF <12 μg/L, the normal group as Hb ≥11 g/dL and SF ≥12 μg/L, and the unclassified anemia group (UCA) as <11 g/dL and SF 12–50 μg/L. Results. The prohepcidin levels ranged from 38.86 to 874.25 ng/mL throughout all the infants (258.60±130.03 ng/mL) but were significantly lower in the AI group than in the normal group (201.93±71.74 vs 299.97±120.68 ng/mL, P=0.007). However there was no significant difference between anemic subgroups. Prohepcidin levels were positively correlated with CRP (r=0.400, P=0.048) in the AI group and were negatively correlated with sTfR in the UCA group (r=−0.376, P=0.022). However in both groups there were no independent predictors in multiple regression analysis. Conclusions. This study examines the prohepcidin levels in infants with anemia and without anemia. The prohepcidin levels of anemic infants were significantly lower than those of infants without anemia. However there was no correlation between prohepcidin and inflammatory cytokines or iron parameters in the groups of AI and IDA, thus we cannot differentiate between them using prohepcidin levels.


Blood ◽  
2021 ◽  
Vol 138 (Supplement 1) ◽  
pp. 4155-4155
Author(s):  
Keren Sam ◽  
Atif Irfan Khan ◽  
Anam Khan ◽  
Sobia Aamir ◽  
Fatima Sajid ◽  
...  

Abstract Background Anemia in inflammatory bowel disease (IBD) presents as a common extraintestinal manifestation resulting in many complications. This condition is often missed or underrated, anemia is secondary to blood loss or defective absorption of iron which can result in a combination of iron deficiency anemia (IDA) or anemia of chronic disease (ACD). The management is iron replacement therapy which improves the quality of life in these patients. Due to the constraints in the use of oral iron, parenteral preparations are more used in IBD patients. Commonly used iron sucrose and ferric carboxymaltose are often associated with side effects leading to poor compliance. Our study explores data about ferric derisomaltose also known as iron isomaltoside (IIM), a recently approved IV iron preparation. The FDA approved this drug in 2020 for patients with poor compliance with other iron preparations. We explored the efficacy and safety data of ferric derisomaltose in adult patients with IBD. Material/Methods A literature search was performed using the following databases: PubMed, Cochrane, Embase, Clinical trials.gov, and Web of Science. The search was completed without using any filter and we used the MeSH Terms for "anemia", "iron deficiency anemia", "inflammatory bowel disease", and "ferric compounds". A total of 1590 articles were screened, and we finally selected 2 trials and 2 observational studies. We followed the PRISMA guidelines for literature search and selection of studies. Case reports, preclinical trials, meta-analyses, and review articles were excluded. Trial and observational studies related to IBD were included. Results In total, 294 patients with anemia in inflammatory bowel disease received ferric derisomaltose intravenously as a single dose between 500 mg to 2000 mg. All patients were >18 years of age. The mean pre-treatment hemoglobin (Hb) level varied from 10.0 g/dL to 12.3 g/dL. The mean serum ferritin ranged from 19.6 µg/L to 57 µg/L and the mean transferrin saturation (TSAT) ranged from 8.8% to 18.5%. J. Stein et al investigated the effectiveness and safety of IIM in routine practical care of IDA in IBD patients. Dahlerup et al (NCT01599702) compared single dose IIM with multiple dosages of IIM in IBD patients. The remaining two studies investigated the effectiveness and safety of high dose IIM in patients with IBD. Results are summarized in Table 1. Efficacy: The increase in hemoglobin ranged from 0.6 g/dl to 2.9 g/dl from baseline while the increase in serum ferritin ranged between 102 µg /L to 250 µg /L and transferrin saturation increased in the range of 15.0%-23.7% in 10 to 52 weeks post iron isomaltoside (IIM) therapy. All markers of iron deficiency anemia showed significant improvement. In the observational study by J. Stein et al, the hemoglobin increased from a mean of 10.7 g/dl to 13.1 g/dl, serum ferritin increased from 57 µg /L to 146 µg /L, and TSAT from 8.8% to 23.7% at 16 weeks. Dahlerup et al demonstrated a Hb increase of >2 g/dl in 75% of patients at 10 weeks. According to W. Reinisch et al (NCT 01410435), there was a mean increase in serum ferritin from 32 µg /L to 102 µg /L and a decrease in TSAT from the baseline, but this decrease was not statistically significant. Safety: Adverse events were noted in 8 (3.6%) patients. Out of the 8 patients, 4 (<2%) were serious adverse events such as perianal abscess, miliary tuberculosis, nephrolithiasis, and worsening of ulcerative colitis, which may not be related to the study drug. The other 4 (1.8%) had life-threatening events, anaphylaxis being the most common. All four patients recovered. The most common mild adverse events reported were hypersensitivity reaction (HSR) seen in 3 patients. 2 patients discontinued due to mild HSR. There were no reports of death due to side effects of the drug. Conclusion Ferric derisomaltose has demonstrated a substantial increment in iron parameters in anemia in patients with IBD. This was measured in terms of hemoglobin response, serum ferritin, and transferrin saturation. Greater Hb response was achieved irrespective of concomitant treatment with steroids and anti-TGF-ß.The adverse events were mild, and patients recovered showing high compliance. Since the average duration of follow-up in our study was 21 weeks, long-term follow-up is limited for this drug, we need further studies to assess the need for maintenance therapy. Figure 1 Figure 1. Disclosures Anwer: GlaxoSmithKline: Research Funding; BMS / Celgene: Honoraria, Research Funding; Janssen pharmaceutical: Honoraria, Research Funding; Allogene Therapeutics: Research Funding.


Blood ◽  
2019 ◽  
Vol 134 (Supplement_1) ◽  
pp. 479-479
Author(s):  
Zhenghao Chen ◽  
Gaspard Cretenet ◽  
Hanneke ter Burg ◽  
Gabriela Andrejeva ◽  
Jeffrey C Rathmell ◽  
...  

Chronic lymphocytic leukemia (CLL) has become a paradigm for a cancer that depends on signals from the microenvironment. In lymph nodes (LN), CLL cells receive from surrounding cells proliferative and pro-survival signals, which can protect against chemotherapy and also the Bcl-2 inhibitor venetoclax (VEN). To avoid drug resistance, combination of VEN with the Bruton's tyrosine kinase (BTK) inhibitor ibrutinib (IBR) is currently explored. IBR has the potency to drive CLL cells out of the LN to eventually die by VEN-induced apoptosis. The activation status in the LN likely affects tumor metabolism, though it is yet unclear how, and whether such putative metabolic changes can be linked to VEN resistance. In this study,we aimed to investigate the altered metabolism of CLL cells in the tumor microenvironment (TME), what signals determine these changes, and how to counteract VEN resistance at the metabolic level. We compared the metabolic status of CLL cells in LN biopsy material and paired peripheral blood (PB) CLL cells. Both higher mitochondrial mass and glucose uptake (flow cytometry) were found in CLL cells derived from LN as compared to PB. To determine which TME signals affect metabolism, we mimicked 3 LN signals: CD40, B cell receptor (BCR) and toll-like receptor (TLR) signaling.Invitrostimulation of PB CLL was followed by mitochondrial mass and glucose uptake (flow cytometry), oxygen consumption rate (OCR) and extracellular acidification rate (ECAR) measured on Seahorse XF Analyzer. The datademonstrated that CD40 but surprisingly not BCR or TLR signaling recapitulated the metabolic changes observed in LN cells. Next, two sets of metabolomics were performed for samples both in vitro and in vivo. Firstly, PB samples of 10 patients were stimulated with/without CD40L for 48 hours and analyzed by mass-spec for metabolic intermediates. In parallel, metabolomics was performed on PB samples from 13 patients sampled before and after 3 months of ibrutinib treatment. The data show various metabolic pathways are activated in the TME, particularly citric acid (TCA) cycle, pyruvate metabolism, glycolysis, and fatty acid metabolism. Apart from these general changes, the highest-ranking shifts in metabolites point to involvement of amino acids to fuel the TCA cycle, which in turn drives oxidative phosphorylation (OXPHOS). Overall, CD40 signaling results in increased glycolysis, but more dominantly, increased OXPHOS, while IBR in fact has opposing effects (Figure 1), indicating that TME-driven metabolic alterations are mitigated by IBR treatment. In order to link the altered metabolic state to VEN resistance, PB-derived cells were treated with OXPHOS inhibitors during CD40 stimulation. Remarkably, OXPHOS inhibition by oligomycin (ATP synthase inhibitor) did not affect CLL activation, yet counteracted strongly for VEN resistance. Of several BCL-2 family members induced by CD40 ligation, both anti-apoptotic MCL-1 and BCL-XL were downregulated by oligomycin. These data suggest that OXPHOS inhibition affects CD40 signals to counteract VEN resistance. In conclusion, metabolic changes of CLL in LN are recapitulated by CD40 signal, while IBR treatment shows opposite effects, together providing indirect insight into the LN metabolism. In LN, most of the key metabolic pathways are enhanced, particularly OXPHOS. Finally, we found OXPHOS inhibition reverses CLL VEN resistance. Our findings link CLL metabolism in LN microenvironment to VEN resistance, and may provide novel clues for therapeutic targeting in the treatment of VEN resistance patients. Disclosures Forconi: Roche: Honoraria; Gilead Sciences: Research Funding; Abbvie: Consultancy, Honoraria, Other: Travel, Accommodations, Expenses, Speakers Bureau; Menarini: Consultancy; Novartis: Honoraria; Janssen-Cilag: Consultancy, Honoraria, Other: Travel, Accommodations, Expenses, Speakers Bureau. van der Windt:Genmab: Employment. Eldering:Celgene: Research Funding; Janssen Pharmaceutical Companies: Research Funding; Roche: Research Funding.


Blood ◽  
2009 ◽  
Vol 114 (22) ◽  
pp. 159-159
Author(s):  
Chante Richardson ◽  
Grant C. Bullock ◽  
Lorrie L Delehanty ◽  
Anne-Laure Talbot ◽  
Kamaleldin E Elagib ◽  
...  

Abstract Abstract 159 The anemias of chronic disease (ACD) are a common complication of malignancy, inflammation and kidney disorders. In ACD, there is dysregulation of iron homeostasis, decreased proliferation of erythroid progenitors, diminished production of erythropoietin (EPO), and shortened lifespan of RBC. Multiple pathophysiologic mechanisms have been implicated in the development of ACD, including elevated production of hepcidin and inflammatory cytokines, IFNγ, TRAIL, Interleukins-1β, 6, 10, 15, & TNFα. These cytokines are thought to directly inhibit erythroid differentiation through unknown mechanisms. The current study addressed the hypothesis that inhibition of erythropoiesis in ACD may arise through synergistic effects of iron deprivation and specific inflammatory cytokines. To identify relevant cytokines, candidate factors were applied to primary human erythroid progenitors in iron replete and restricted cultures. Peripheral blood human CD34+ progenitors from healthy donors underwent standard prestimulation for 72 hours, followed by culture in unilineage erythroid medium (4.5 U/ml EPO + 25ng/ml SCF) for 4-5 days under iron replete (100% transferrin saturation) or iron restricted (15% transferrin saturation) conditions. Candidate cytokines were screened for effects on viability, proliferation, and differentiation using cell counting and flow cytometric analysis of the erythroid cell surface marker GPA and the megakaryocytic antigen CD41a. Contrary to previous reports, the majority of cytokines (TRAIL & Interleukins-1β, 6, 10, 15) showed no effects on erythroid proliferation or differentiation under iron replete or restricted conditions. By contrast, both IFNγ and TNFα displayed potent inhibitory effects under iron restricted conditions but only weak effects in iron replete cultures. Typically, iron restriction alone reduced the proportion of GPA+ cells by 50%, whereas IFNγ or TNFα combined with iron restriction caused a 90% reduction. While both cytokines cooperated with iron restriction in blocking upregulation of GPA and promoting cell death, each cytokine also had distinctive effects on morphology and differentiation. IFNγ enhanced megakaryocytic development, while TNFα retained cells as immature, CD34+ progenitors. The synergistic inhibition of erythroid differentiation with iron restriction and TNFα was confirmed in vivo using a murine model of dietary iron deprivation coupled with continuous infusion of low-dose TNFα. Regarding the mechanism for this synergy, we have previously shown that erythroid iron deprivation leads to inactivation of the aconitase enzymes, which normally convert citrate to isocitrate, and that provision of exogenous isocitrate abrogates the erythroid inhibition associated with iron deprivation. Accordingly, participation of this pathway was assessed in the more potent erythroid inhibition associated with IFNγ or TNFα plus iron deprivation. Strikingly, isocitrate administration not only abrogated effects due to iron deprivation but also those due to the inflammatory cytokines, leading to complete rescue of erythroid differentiation. To address the underlying basis for erythroid cross-talk of iron and cytokine signaling, we screened pathways implicated in iron metabolism and inflammation. Two relevant pathways identified were Jun kinase (JNK) and calmodulin-associated kinase II (CAMKII), important in TNFα and IFNγ signaling, respectively. In particular, TNFα and iron deprivation synergized in the activation of JNK, and IFNγ and iron deprivation synergized in activating CAMKII. In both cases, isocitrate partially restored the activation to basal levels. As an important negative control, iron deprivation did not affect IFNγ activation of STAT1 phosphorylation, indicating that its effects were not due to upregulation of receptor expression or function. Altogether, these data suggest that among the various cytokines implicated in ACD, only IFNγ and TNFα synergize with iron deprivation in the inhibition of erythropoiesis. These actions occur through cross-talk between intracellular signaling pathways, specifically pathways involving aconitase and cytokine-activated kinases. The connection of aconitase/metabolism with inflammation is novel and has implications for clinical treatment of ACD, as well as for new understanding of erythroid and inflammatory signaling. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2018 ◽  
Vol 132 (Supplement 1) ◽  
pp. 1760-1760
Author(s):  
Amy Zhou ◽  
Maggie J. Allen ◽  
Mary Fulbright ◽  
Jared S Fowles ◽  
Daniel A.C. Fisher ◽  
...  

Abstract Introduction: Hepcidin, a peptide-hormone produced by hepatocytes, is a key regulator of iron homeostasis in mammals. Hepcidin levels are affected by several factors including inflammation, iron concentration, and erythropoietic signaling, and elevated hepcidin levels can lead to anemia. Inflammatory cytokines such as IL-6 induce hepcidin mRNA transcription via JAK/STAT signaling, whereas high serum iron levels and high erythropoietic drive via erythroferrone (ERFE) suppress hepcidin transcription via BMP/SMAD signaling. Anemia is a common problem in patients with myelofibrosis (MF). In a previous study of primary myelofibrosis (PMF) patients, serum hepcidin levels were found to be elevated compared to normal controls and were associated with increased RBC transfusion requirement and reduced survival (Pardanani et al. Am J Hematol 2013;88(4):312-316). Ruxolitinib is a JAK1/2 inhibitor approved for treatment of patients with MF. Treatment with ruxolitinib reduces spleen volume, constitutional symptoms, and inflammatory cytokines, but does not lead to anemia improvement in MF (Verstovsek et al. NEJM 2012;366:799-807). To further assess the relationship between hepcidin and anemia in MF, we measured hepcidin levels in 99 MF patients including 49 PMF and 50 secondary MF (sMF) patients, as well as 24 patients treated with ruxolitinib. We also evaluated the relationship between hepcidin and hemoglobin levels, inflammatory cytokines, and serum iron markers. Methods: Hepcidin levels (ng/mL) were measured using a commercially available ELISA assay from Intrinsic Lifesciences (Intrinsic Hepcidin IDxTM ELISA) in 99 MF patient plasma samples and 9 normal controls. Plasma cytokine levels (IFN, IL-10, IL-2, IL-2Rα, IL-6, IL-8, MIP-1α, MIP-1β, TNFα, VEGF, TGF-β1, TGF-β2, TGF-β3) were measured via multiplex cytokine analysis (Mesoscale Discovery). CRP, transferrin saturation, and ferritin were measured using standard laboratory methods at our institution. Statistical analyses were performed using GraphPad Prism software. Results: Hepcidin was significantly elevated in MF patients compared to normals (median 81.1 vs 8.5, p <0.001). There was no difference in hepcidin expression between PMF and sMF (median 103.3 vs 80.8, p = 0.380). Hepcidin positively correlated with IL-2Rα and IL-6, but did not correlate with other cytokines evaluated. Hepcidin expression did not differ in samples obtained from patients on treatment with ruxolitinib versus those who were not (median 105.1 vs 80.2, respectively, p = 0.998). A trend between higher hepcidin levels and lower hemoglobin was observed, although it was not statistically significant (p = 0.719). No correlation was found between hepcidin levels and CRP, transferrin saturation, or ferritin. These results are consistent with the previous study confirming that hepcidin is elevated in MF. In addition, we found a correlation between hepcidin and IL-6, which was not observed previously (Pardanani et al. Am J Hematol 2013;88(4):312-316). The lack of significant correlation between hepcidin and hemoglobin may be due to our smaller sample size. Despite ruxolitinib therapy leading to decreases in inflammatory cytokine production, hepcidin levels were markedly elevated in patients treated with ruxolitinib. Conclusion: These findings indicate that abnormal hepcidin production is a hallmark of both PMF and sMF. Hepcidin expression correlated with IL-6 and IL-2Rα, but not with other inflammatory cytokines, nor with CRP. These observations suggest a complex relationship between inflammation and hepcidin in MF. Although a significant correlation between hepcidin and hemoglobin was not found, this may have been related to the relatively small sample size. In addition, RBC transfusion in a subset of patients may have impacted the lack of correlation between iron indices and hepcidin. Hepcidin was high in patients treated with ruxolitinib, suggesting that suppression of JAK-STAT signaling (such as downstream of IL-6) via ruxolitinib may not be sufficient to normalize hepcidin production. Alternatively, inhibition of hepcidin production via ACVR1/ALK2 has been proposed to be the mechanism behind anemia improvement observed with momelotinib therapy (Asshoff et al. Blood 2017;129(13): 1823 - 1830). Therefore, the role of hepcidin as a therapeutic target for treatment of anemia in patients with MF warrants further exploration. Disclosures Zhou: Incyte: Speakers Bureau. Oh:Gilead: Research Funding; Janssen: Research Funding; Incyte: Consultancy, Membership on an entity's Board of Directors or advisory committees, Research Funding; Takeda: Research Funding; CTI Biopharma: Research Funding.


2021 ◽  
Vol 80 (Suppl 1) ◽  
pp. 518.2-518
Author(s):  
E. Galushko ◽  
A. Semashko ◽  
A. Gordeev ◽  
A. Lila

Background:Anemia of inflammation (AI) and iron deficiency anemia (IDA) are the two most prevalent forms of anemia in patients with rheumatoid arthritis (RA). Diagnosis becomes challenging if AI is associated with true ID (AI/ID), as there is still a lack of a gold standard for differentiation between AI and AI/ID. However, as therapies to overcome anemia differ, proper diagnosis and understanding of underlying pathophysiological regulations are necessary.Objectives:The aim of the study was to evaluate the clinical efficiency of hepcidin, a key regulator of iron metabolism, in the diagnosis of IDA, as well as the differential diagnosis of AI/ID and AI in patients with RA.Methods:The study was undertaken 96 patients with RA, 67 of them were diagnosed anemia according to WHO criteria (104,3±21,4 g/l). Anemic patients and anemia-free patients with RA (n=29) were comparable (p>0.05) in age (44.4±14.8 and 49.8±9.3 years), disease duration (73.5±65.4 and 59.8±48.3 months) and DAS28 (6.3±1.6 and 5.9±1.9). All cases were subjected to following tests: complete blood count with peripheral smear, serum C-reactive protein, serum interleukin-6, iron studies, serum soluble transferrin receptor (sTfR), and serum hepcidin. Patients with RA and anemia were divided two groups: 25 patients with IDA and 42 - with AI. The AI cases were subdivided into pure AI and AI with coexistent ID (n=15).Results:The mean serum hepcidin concentration was significantly increased in pure AI patients (123.85±25.8 ng/mL) as compared to those in IDA patients (63.9±22.8 ng/mL, P < 0.05) and anemia-free patients with RA (88.1±39.09 ng/mL). Also, compared to pure AI patients [normal sTfR levels (<3 µg/mL)], the serum hepcidin concentration was reduced significantly in AI patients with ID [high sTfR levels (≥3 µg/mL)] with a mean of 79.0±23.97 ng/mL.Conclusion:Hepcidin measurement can provide a useful tool for differentiating AI from IDA and also help to identify an iron deficiency in AI patients. This might aid in the appropriate selection of therapy for these patients.Disclosure of Interests:None declared


Nutrients ◽  
2020 ◽  
Vol 13 (1) ◽  
pp. 104
Author(s):  
Aneta Jończy ◽  
Rafał Mazgaj ◽  
Rafał Radosław Starzyński ◽  
Piotr Poznański ◽  
Mateusz Szudzik ◽  
...  

In mammals, 2 × 1012 red blood cells (RBCs) are produced every day in the bone marrow to ensure a constant supply of iron to maintain effective erythropoiesis. Impaired iron absorption in the duodenum and inefficient iron reutilization from senescent RBCs by macrophages contribute to the development of anemia. Ferroportin (Fpn), the only known cellular iron exporter, as well as hephaestin (Heph) and ceruloplasmin, two copper-dependent ferroxidases involved in the above-mentioned processes, are key elements of the interaction between copper and iron metabolisms. Crosslinks between these metals have been known for many years, but metabolic effects of one on the other have not been elucidated to date. Neonatal iron deficiency anemia in piglets provides an interesting model for studying this interplay. In duodenal enterocytes of young anemic piglets, we identified iron deposits and demonstrated increased expression of ferritin with a concomitant decline in both Fpn and Heph expression. We postulated that the underlying mechanism involves changes in copper distribution within enterocytes as a result of decreased expression of the copper transporter—Atp7b. Obtained results strongly suggest that regulation of iron absorption within enterocytes is based on the interaction between proteins of copper and iron metabolisms and outcompetes systemic regulation.


2004 ◽  
Vol 4 (3) ◽  
pp. 241-248 ◽  
Author(s):  
Rute Cândida Pereira ◽  
Alcides da Silva Diniz ◽  
Luiz Oscar Cardoso Ferreira

The authors focus iron intake regulation in the body and the probable mechanisms related to iron absorption. They analyze the impact of iron absorption deficiency resulting in iron deficiency anemia, a public health issue of great impact in the world influencing child and maternal health risk increase. This paper aims at highlighting the problems affecting the uptake or inhibiting processes of iron absorption in an attempt to correlate information on conditioning factors and current findings. This study is a document based descriptive study comprising literature review. In food, iron has different forms, such as the heme and non-heme forms following different absorption pathways with different efficiency rates, depending on conditioning factors, such as diet profile, physiological aspects, iron chemical state, absorption regulation, transportation, storing, excretion and the presence of disease, They also discuss the current difficulties in dealing with iron nutritional deficiency in vulnerable groups, children and pregnant women, and focus data on iron consumption, adhesion to breast feeding and the frequency of prenatal care visits.


1963 ◽  
Vol 204 (1) ◽  
pp. 171-175 ◽  
Author(s):  
W. S. Ruliffson ◽  
J. M. Hopping

The effects in rats, of age, iron-deficiency anemia, and ascorbic acid, citrate, fluoride, and ethylenediaminetetraacetate (EDTA) on enteric radioiron transport were studied in vitro by an everted gut-sac technique. Sacs from young animals transported more than those from older ones. Proximal jejunal sacs from anemic animals transported more than similar sacs from nonanemic rats, but the reverse effect appeared in sacs formed from proximal duodenum. When added to media containing ascorbic acid or citrate, fluoride depressed transport as did anaerobic incubation in the presence of ascorbic acid. Anaerobic incubation in the presence of EDTA appeared to permit elevated transport. Ascorbic acid, citrate, and EDTA all enhanced the level of Fe59 appearing in serosal media. These results appear to agree with previously established in vivo phenomena and tend to validate the in vitro method as one of promise for further studies of factors affecting iron absorption and of the mechanism of iron absorption.


Sign in / Sign up

Export Citation Format

Share Document