Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 68
Filter
1.
Int J Mol Sci ; 23(3)2022 Jan 25.
Article in English | MEDLINE | ID: mdl-35163276

ABSTRACT

Hemolysis is known to cause acute kidney injury (AKI). The iron regulatory hormone hepcidin, produced by renal distal tubules, is suggested to exert a renoprotective role during this pathology. We aimed to elucidate the molecular mechanisms of renal hepcidin synthesis and its protection against hemoglobin-induced AKI. In contrast to known hepatic hepcidin induction, incubation of mouse cortical collecting duct (mCCDcl1) cells with IL-6 or LPS did not induce Hamp1 mRNA expression, whereas iron (FeS) and hemin significantly induced hepcidin synthesis (p < 0.05). Moreover, iron/heme-mediated hepcidin induction in mCCDcl1 cells was caused by the nuclear factor erythroid 2-related factor 2 (Nrf2) pathway, as indicated by increased nuclear Nrf2 translocation and induced expression of Nrf2 downstream targets GCLM (p < 0.001), NQO1 (p < 0.001), and TXNRD1 (p < 0.005), which could be prevented by the known Nrf2 inhibitor trigonelline. Newly created inducible kidney-specific hepcidin KO mice demonstrated a significant reduction in renal Hamp1 mRNA expression. Phenylhydrazine (PHZ)-induced hemolysis caused renal iron loading and oxidative stress in both wildtype (Wt) and KO mice. PHZ treatment in Wt induced inflammatory markers (IL-6, TNFα) but not Hamp1. However, since PHZ treatment also significantly reduced systemic hepcidin levels in both Wt and KO mice (both p < 0.001), a dissection between the roles of systemic and renal hepcidin could not be made. Combined, the results of our study indicate that there are kidney-specific mechanisms in hepcidin regulation, as indicated by the dominant role of iron and not inflammation as an inducer of renal hepcidin, but also emphasize the complex interplay of various iron regulatory mechanisms during AKI on a local and systemic level.


Subject(s)
Acute Kidney Injury/metabolism , Hepcidins/metabolism , Acute Kidney Injury/chemically induced , Acute Kidney Injury/physiopathology , Animals , Hemin/metabolism , Hemoglobins/metabolism , Hemolysis/physiology , Hepcidins/physiology , Iron/metabolism , Kidney/metabolism , Kidney/pathology , Kidney Tubules, Distal/metabolism , Mice , Mice, Knockout , Oxidative Stress
2.
Int Urol Nephrol ; 53(5): 935-944, 2021 May.
Article in English | MEDLINE | ID: mdl-33025407

ABSTRACT

Iron is the most abundant transition metal in the human body and an essential element required for growth and survival. Our understanding of the molecular control of iron metabolism has increased dramatically over the past 20 years due to the discovery of hepcidin, which regulates the uptake of dietary iron and its mobilization from macrophages and hepatic stores. Anemia and iron deficiency are common in chronic kidney disease. The pathogenesis of anemia of chronic kidney disease is multifactorial. Correction of anemia requires two main treatment strategies: increased stimulation of erythropoiesis, and maintenance of an adequate iron supply to the bone marrow. However, there are still many uncertainties in regard to iron metabolism in patients with chronic kidney disease and in renal replacement therapy. The aim of this review was to summarize the current knowledge on iron metabolism in this population, including new biomarkers of iron status. There is an area of uncertainty regarding diagnostic utility of both erythroferrone (ERFE) and hepcidin in end-stage renal disease (ESRD) patients. Higher concentration of hepcidin in oligoanuric patients may reflect decreased renal clearance. Furthermore, the hepcidin-lowering effect of ERFE in ESRD patients treated with erythropoiesis-stimulating agents (ESAs) may be blunted by underlying inflammation and concomitant iron treatment. Thus, future studies should validate the use of ERFE as a biomarker of erythropoiesis and predictor of response to iron and ESA therapy in dialysis-dependent patients.


Subject(s)
Iron/metabolism , Renal Insufficiency, Chronic/metabolism , Biomarkers/analysis , Hepcidins/physiology , Humans
3.
Sports Med ; 50(12): 2111-2123, 2020 Dec.
Article in English | MEDLINE | ID: mdl-33057935

ABSTRACT

Iron deficiency (ID) is a prevailing nutritional concern amongst the athletic population due to the increased iron demands of this group. Athletes' ability to replenish taxed iron stores is challenging due to the low bioavailability of dietary sources, and the interaction between exercise and hepcidin, the primary iron-regulatory hormone. To date, copious research has explored the link between exercise and iron regulation, with a more recent focus on optimising iron treatment applications. Currently, oral iron supplementation is typically the first avenue of iron replacement therapy beyond nutritional intervention, for treatment of ID athletes. However, many athletes encounter associated gastrointestinal side-effects which can deter them from fulfilling a full-term oral iron treatment plan, generally resulting in sub-optimal treatment efficacy. Consequently, various strategies (e.g. dosage, composition, timing) of oral iron supplementation have been investigated with the goal of increasing fractional iron absorption, reducing gastric irritation, and ultimately improving the efficacy of oral iron therapy. This review explores the various treatment strategies pertinent to athletes and concludes a contemporary strategy of oral iron therapy entailing morning supplementation, ideally within the 30 min following morning exercise, and in athletes experiencing gut sensitivity, consumed on alternate days or at lower doses.


Subject(s)
Dietary Supplements , Exercise , Iron Deficiencies , Iron/administration & dosage , Athletes , Hepcidins/physiology , Humans
4.
Int J Mol Sci ; 21(11)2020 May 28.
Article in English | MEDLINE | ID: mdl-32481481

ABSTRACT

Iron deficiency (ID) is the most frequent nutritional deficiency in the whole population worldwide, and the second most common cause of anemia in the elderly. The prevalence of anemia is expecting to rise shortly, because of an ageing population. Even though WHO criteria define anemia as a hemoglobin serum concentration <12 g/dL in women and <13 g/dL in men, several authors propose different and specific cut-off values for the elderly. Anemia in aged subjects impacts health and quality of life, and it is associated with several negative outcomes, such as longer time of hospitalization and a higher risk of disability. Furthermore, it is an independent risk factor of increased morbidity and mortality. Even though iron deficiency anemia is a common disorder in older adults, it should be not considered as a normal ageing consequence, but a sign of underlying dysfunction. Relating to the molecular mechanism in Iron Deficiency Anemia (IDA), hepcidin has a key role in iron homeostasis. It downregulates the iron exporter ferroportin, inhibiting both iron absorption and release. IDA is frequently dependent on blood loss, especially caused by gastrointestinal lesions. Thus, a diagnostic algorithm for IDA should include invasive investigation such as endoscopic procedures. The treatment choice is influenced by the severity of anemia, underlying conditions, comorbidities, and the clinical state of the patient. Correction of anemia and iron supplementation should be associated with the treatment of the causal disease.


Subject(s)
Anemia, Iron-Deficiency/etiology , Anemia, Iron-Deficiency/therapy , Iron/administration & dosage , Administration, Oral , Aged , Aged, 80 and over , Aging , Algorithms , Disabled Persons , Female , Hemoglobins/analysis , Hepcidins/physiology , Humans , Infusions, Parenteral , Iron/pharmacokinetics , Iron Deficiencies , Male , Nutritional Sciences , Prevalence , Quality of Life , Risk Factors
5.
Article in German | MEDLINE | ID: mdl-32557515

ABSTRACT

Iron (Fe) is an essential trace element. In daily veterinary practice, it plays a pivotal role e. g. due to its role in Fe deficiency anaemia. The bioavailability of Fe, for example for heme and hemoglobin synthesis, sets high demands on Fe homeostasis. The discovery of hepcidin as being an important regulative protein made a hormone-like regulation of the Fe metabolism evident. Hepcidin is synthesized by the liver and regulates the trans-membranous Fe-transporter ferroportin. An increase of hepcidin leads to a decrease of Fe export from the cell into the extracellular space, the consequence being an internalisation of Fe in the reticuloendothelial system as well as in mononuclear cells. Additionally, enteral Fe uptake decreases. The induction of hepatic hepcidin synthesis seems to be caused by high Fe- and transferrin concentrations in plasma. In addition to this, an increase of cytokines during inflammation similarly triggers hepatic hepcidin synthesis. This finding offers an explanation for the frequently observed decrease of Fe in serum/plasma during acute inflammation, the mechanism thus being termed as cytokine-hepcidin-link. Based on the fact that numerous pathogens require Fe for their own metabolism, internalisation of Fe into the intracellular compartment during inflammation has hence been categorised as being a part of the innate immunity. Iron supplementation, initiated by the veterinarian or the farmer, interferes with this regulation. Currently however, there is a lack of knowledge regarding the clinical and metabolic impacts of parenteral or oral Fe supplementation to farm animals. Therefore, the acquisition of added scientific data via prospective studies is warranted. In consequence, novel findings may lead to a reassessment of Fe supplementation strategies for ruminants, pigs and/or horses.


Subject(s)
Hepcidins , Inflammation , Iron , Livestock , Animals , Dietary Supplements , Hepcidins/metabolism , Hepcidins/physiology , Inflammation/metabolism , Inflammation/physiopathology , Inflammation/veterinary , Iron/metabolism , Iron/pharmacokinetics , Iron/physiology , Livestock/metabolism , Livestock/physiology
6.
Int J Infect Dis ; 97: 303-305, 2020 Aug.
Article in English | MEDLINE | ID: mdl-32497811

ABSTRACT

The coronavirus 2 (SARS-CoV-2) pandemic is viciously spreading through the continents with rapidly increasing mortality rates. Current management of COVID-19 is based on the premise that respiratory failure is the leading cause of mortality. However, mounting evidence links accelerated pathogenesis in gravely ill COVID-19 patients to a hyper-inflammatory state involving a cytokine storm. Several components of the heightened inflammatory state were addressed as therapeutic targets. Another key component of the heightened inflammatory state is hyper-ferritinemia which reportedly identifies patients with increased mortality risk. In spite of its strong association with mortality, it is not yet clear if hyper-ferritinemia in COVID-19 patients is merely a systemic marker of disease progression, or a key modulator in disease pathogenesis. Here we address implications of a possible role for hyper-ferritinemia, and altered iron homeostasis in COVID-19 pathogenesis, and potential therapeutic targets in this regard.


Subject(s)
Coronavirus Infections/pathology , Iron Overload/virology , Pneumonia, Viral/pathology , Betacoronavirus , COVID-19 , Coronavirus Infections/mortality , Cytokine Release Syndrome/virology , Ferroptosis , Hepcidins/physiology , Humans , Inflammation , Iron/blood , Mitochondria/pathology , Mitochondria/physiology , Oxidative Stress , Pandemics , Pneumonia, Viral/mortality , SARS-CoV-2
7.
Nutrients ; 12(5)2020 May 08.
Article in English | MEDLINE | ID: mdl-32397086

ABSTRACT

Despite the crucial role of the liver as the central regulator of iron homeostasis, no studies have directly tested the modulation of liver gene and protein expression patterns during iron deficiency instauration and recovery with fermented milks. Fermented goat milk consumption improves the key proteins of intestinal iron metabolism during iron deficiency recovery, enhancing the digestive and metabolic utilization of iron. The aim of this study was to assess the influence of fermented goat or cow milk consumption on liver iron homeostasis during iron-deficiency anemia recovery with normal or iron-overload diets. Analysis included iron status biomarkers, gene and protein expression in hepatocytes. In general, fermented goat milk consumption either with normal or high iron content up-regulated liver DMT1, FPN1 and FTL1 gene expression and DMT1 and FPN1 protein expression. However, HAMP mRNA expression was lower in all groups of animals fed fermented goat milk. Additionally, hepcidin protein expression decreased in control and anemic animals fed fermented goat milk with normal iron content. In conclusion, fermented goat milk potentiates the up-regulation of key genes coding for proteins involved in iron metabolism, such as DMT1, and FPN1, FTL1 and down-regulation of HAMP, playing a key role in enhanced iron repletion during anemia recovery, inducing a physiological adaptation of the liver key genes and proteins coordinated with the fluctuation of the cellular iron levels, favoring whole-body iron homeostasis.


Subject(s)
Anemia, Iron-Deficiency/metabolism , Cation Transport Proteins/genetics , Cation Transport Proteins/metabolism , Eating/physiology , Fermentation , Gene Expression , Hepcidins/genetics , Hepcidins/metabolism , Homeostasis/genetics , Iron/metabolism , Liver/metabolism , Milk , Animals , Apoferritins/genetics , Apoferritins/metabolism , Apoferritins/physiology , Cation Transport Proteins/physiology , Cattle , Goats , Hepcidins/physiology , Humans , Intestinal Mucosa/metabolism , Rats, Wistar
9.
J Immunol ; 203(9): 2485-2496, 2019 11 01.
Article in English | MEDLINE | ID: mdl-31562210

ABSTRACT

Anemia is a frequent and challenging complication of mycobacterial infections. We used a model of disseminated Mycobacterium avium infection in mice to investigate the mechanisms of mycobacteria-induced anemia. We found increased formation of RBC in the bone marrow and spleen of infected mice. Infection induced reticulocytosis and the premature egress of immature progenitors to the systemic circulation in an IFN-γ (IFNG)-dependent way. The newly formed RBC had reduced CD47 surface expression and a reduced life span and were phagocytosed in the liver of infected mice, increasing iron recycling in this organ. The increased engulfment and degradation of RBC was independent of IFNG sensing by macrophages. Together, our findings demonstrate that mycobacterial infection alters the formation of erythrocytes, leading to their accelerated removal from circulation and hemolytic anemia. This comprehensive elucidation of the mechanisms underlying mycobacteria-induced anemia has important implications for its efficient clinical management.


Subject(s)
Anemia/etiology , Erythrocytes/physiology , Interferon-gamma/physiology , Mycobacterium Infections/complications , Animals , Bone Marrow Cells/cytology , CD47 Antigen/analysis , Cell Differentiation , Erythropoiesis , Hepcidins/physiology , Mice , Mice, Inbred C57BL , Mycobacterium Infections/blood , Phagocytosis
10.
Nat Metab ; 1(5): 519-531, 2019 05.
Article in English | MEDLINE | ID: mdl-31276102

ABSTRACT

Iron is critical for life but toxic in excess because of iron-catalysed formation of pro-oxidants that cause tissue damage in a range of disorders. The Nrf2 transcription factor orchestrates cell-intrinsic protective antioxidant responses, and the peptide hormone hepcidin maintains systemic iron homeostasis, but is pathophysiologically decreased in haemochromatosis and beta-thalassaemia. Here, we show that Nrf2 is activated by iron-induced, mitochondria-derived pro-oxidants and drives Bmp6 expression in liver sinusoid endothelial cells, which in turn increases hepcidin synthesis by neighbouring hepatocytes. In Nrf2 knockout mice, the Bmp6-hepcidin response to oral and parenteral iron is impaired and iron accumulation and hepatic damage are increased. Pharmacological activation of Nrf2 stimulates the Bmp6-hepcidin axis, improving iron homeostasis in haemochromatosis and counteracting the inhibition of Bmp6 by erythroferrone in beta-thalassaemia. We propose that Nrf2 links cellular sensing of excess toxic iron to control of systemic iron homeostasis and antioxidant responses, and may be a therapeutic target for iron-associated disorders.


Subject(s)
Bone Morphogenetic Protein 6/physiology , Hepcidins/physiology , Homeostasis/physiology , Iron/metabolism , NF-E2-Related Factor 2/physiology , beta-Thalassemia/physiopathology , Humans
11.
Am J Pathol ; 189(9): 1814-1830, 2019 09.
Article in English | MEDLINE | ID: mdl-31287995

ABSTRACT

The liver secretes hepcidin (Hepc) into the bloodstream to reduce blood iron levels. Hepc accomplishes this by triggering degradation of the only known cellular iron exporter ferroportin in the gut, macrophages, and liver. We previously demonstrated that systemic Hepc knockout (HepcKO) mice, which have high serum iron, develop retinal iron overload and degeneration. However, it was unclear whether this is caused by high blood iron levels or, alternatively, retinal iron influx that would normally be regulated by retina-produced Hepc. To address this question, retinas of liver-specific and retina-specific HepcKO mice were studied. Liver-specific HepcKO mice had elevated blood and retinal pigment epithelium (RPE) iron levels and increased free (labile) iron levels in the retina, despite an intact blood-retinal barrier. This led to RPE hypertrophy associated with lipofuscin-laden lysosome accumulation. Photoreceptors also degenerated focally. In contrast, there was no change in retinal or RPE iron levels or degeneration in the retina-specific HepcKO mice. These data indicate that high blood iron levels can lead to retinal iron accumulation and degeneration. High blood iron levels can occur in patients with hereditary hemochromatosis or result from use of iron supplements or multiple blood transfusions. Our results suggest that high blood iron levels may cause or exacerbate retinal disease.


Subject(s)
Hepcidins/physiology , Iron Overload/etiology , Iron/metabolism , Liver/metabolism , Retina/metabolism , Retinal Degeneration/etiology , Animals , Blood-Retinal Barrier , Female , Iron Overload/metabolism , Iron Overload/pathology , Liver/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Retina/pathology , Retinal Degeneration/metabolism , Retinal Degeneration/pathology
12.
Dev Period Med ; 23(2): 137-141, 2019.
Article in English | MEDLINE | ID: mdl-31280251

ABSTRACT

Iron is an element whose content in the human organism remains under strict control not only due to its involvement in many life processes but also because of its potential toxicity. The latest studies in iron metabolism, especially the involvement of hepcidin, which is the main regulator of iron homeostasis, broadened our knowledge in many medical fields (immunology, nephrology, hematology, gastrology). The present paper is a review of the literature devoted to the importance of hepcidin under selected conditions.


Subject(s)
Communicable Diseases/metabolism , Hepcidins/metabolism , Homeostasis , Iron/metabolism , Metabolic Diseases/metabolism , Chronic Disease , Communicable Diseases/physiopathology , Hepcidins/physiology , Humans , Metabolic Diseases/physiopathology
13.
Hepatology ; 70(6): 1986-2002, 2019 12.
Article in English | MEDLINE | ID: mdl-31127639

ABSTRACT

A failure of iron to appropriately regulate liver hepcidin production is central to the pathogenesis of hereditary hemochromatosis. SMAD1/5 transcription factors, activated by bone morphogenetic protein (BMP) signaling, are major regulators of hepcidin production in response to iron; however, the role of SMAD8 and the contribution of SMADs to hepcidin production by other systemic cues remain uncertain. Here, we generated hepatocyte Smad8 single (Smad8fl/fl ;Alb-Cre+ ), Smad1/5/8 triple (Smad158;Alb-Cre+ ), and littermate Smad1/5 double (Smad15;Alb-Cre+ ) knockout mice to investigate the role of SMAD8 in hepcidin and iron homeostasis regulation and liver injury. We found that Smad8;Alb-Cre+ mice exhibited no iron phenotype, whereas Smad158;Alb-Cre+ mice had greater iron overload than Smad15;Alb-Cre+ mice. In contrast to the sexual dimorphism reported for wild-type mice and other hemochromatosis models, hepcidin deficiency and extrahepatic iron loading were similarly severe in Smad15;Alb-Cre+ and Smad158;Alb-Cre+ female compared with male mice. Moreover, epidermal growth factor (EGF) failed to suppress hepcidin in Smad15;Alb-Cre+ hepatocytes. Conversely, hepcidin was still increased by lipopolysaccharide in Smad158;Alb-Cre+ mice, although lower basal hepcidin resulted in lower maximal hepcidin. Finally, unlike most mouse hemochromatosis models, Smad158;Alb-Cre+ developed liver injury and fibrosis at 8 weeks. Liver injury and fibrosis were prevented in Smad158;Alb-Cre+ mice by a low-iron diet and were minimal in iron-loaded Cre- mice. Conclusion: Hepatocyte Smad1/5/8 knockout mice are a model of hemochromatosis that encompasses liver injury and fibrosis seen in human disease. These mice reveal the redundant but critical role of SMAD8 in hepcidin and iron homeostasis regulation, establish a requirement for SMAD1/5/8 in hepcidin regulation by testosterone and EGF but not inflammation, and suggest a pathogenic role for both iron loading and SMAD1/5/8 deficiency in liver injury and fibrosis.


Subject(s)
Hepatocytes/metabolism , Iron Overload/etiology , Iron/metabolism , Liver Cirrhosis, Experimental/etiology , Smad1 Protein/physiology , Smad5 Protein/physiology , Smad8 Protein/physiology , Animals , Cells, Cultured , Epidermal Growth Factor/pharmacology , Female , Hepcidins/physiology , Male , Mice , Mice, Inbred C57BL
14.
Contrib Nephrol ; 198: 124-134, 2019.
Article in English | MEDLINE | ID: mdl-30991414

ABSTRACT

BACKGROUND: The pathogenesis of anemia in chronic kidney disease (CKD) could be multifactorial. In recent animal studies, hepcidin knockout (KO) mice with adenine-induced CKD did not exhibit anemia and iron deficiency. Hepcidin has emerged as a major player in the development of anemia in CKD. We suspected that erythropoietin (EPO) deficiency may not be the mainstay of anemia in CKD, although relative EPO deficiency could contribute to the failure to increase hemoglobin (Hb) levels. Some factors may interfere with the differentiation of erythroids. SUMMARY: Based on previous flow cytometric analysis, the differentiation and maturation of bone marrow erythroid precursors were compared between 2 mouse models of anemia, namely, EPO-KO mice and adenine-induced CKD mice. EPO-KO mice exhibited greater than 50% reduction in the CD71-low/Ter119-high population, which represents a mature erythroid stage in the bone marrow. In contrast, these mice exhibited no reduction in the CD71-high/Ter119-low and CD71-high/Ter119-high cell populations, which represent an early erythroid stage. However, in CKD mice, the percentages of CD71-high/Ter119-low and CD71-high/Ter119-high erythroid cells, which correspond to proerythroblasts and basophilic erythroblasts, respectively, were decreased in bone marrow. Thus, the CKD mice exhibited a decrease in the number of cells expressing transferrin receptor 1 (TfR1) or early stage erythroblasts, which was completely different from the results obtained for EPO-KO mice. Thus, in CKD, decreased expression of TfR1 in erythroblasts as well as increased hepcidin levels in circulation may hamper erythroblast differentiation by decreasing the iron supply, as iron is an indispensable component of erythroblast differentiation. We conclude that deregulated iron metabolism could be the principal cause of anemia in CKD, impeding the differentiation of erythroblasts. We propose that the "hepcidin-anemia axis" is involved in the pathogenesis of CKD-associated anemia. For the treatment of anemia in CKD, declining hepcidin levels are essential for efficient erythropoiesis. Key Messages: These findings have led us to target the hepcidin-anemia axis as a new treatment strategy for anemia in CKD, including via newly developed erythropoiesis-stimulating agent and hypoxia inducible factor stabilizers.


Subject(s)
Anemia/pathology , Hepcidins/drug effects , Renal Insufficiency, Chronic/complications , Anemia/etiology , Animals , Bone Marrow Cells/cytology , Cell Differentiation , Erythroblasts/cytology , Erythroid Cells/cytology , Erythropoiesis , Hepcidins/physiology , Humans , Iron/metabolism , Mice , Renal Insufficiency, Chronic/pathology
15.
Nephrology (Carlton) ; 24(7): 751-757, 2019 Jul.
Article in English | MEDLINE | ID: mdl-30175513

ABSTRACT

AIM: Relatively few haemodialysis (HD) patients remain independent of recombinant human erythropoietin ('rHU-EPO free patients'). We investigated the role of EPO and hepcidin, two key hormones involved in anaemia. METHODS: We report a monocentric case-control series. Iron status, EPO and hepcidin levels were analysed in 15 Adult HD (Age > 18 years) with a stable haemoglobin (Hb) level that have not received rHU-EPO for at least 6 months (=rHU-EPO free patients); and in 60 controls with a stable rHU-EPO dose and Hb level. RESULTS: The rHU-EPO free patients had a higher Hb level compared to controls (12.1 ± 0.99 g/dL vs 11.1 ± 0.73, P = 0.0014), and a lower ferritin level (183 ± 102 vs 312 ± 166 ng/mL, P = 0.001). Hepcidin levels were lower in the rHU-EPO free patients (12.53 ± 10.46 ng/mL) compared to the controls (37.95 ± 34.33 ng/mL), P = 0.0033. Hepcidin levels correlated significantly with ferritin levels; but neither with transferrin saturation, C-reactive protein nor EPO levels. Unsupervised analysis revealed that rHU-EPO free patients had a specific clinical/biological profile (presence of renal cyst, longer dialysis vintage, lower ferritin, and EPO and hepcidin levels compared to the control group). Finally, we showed that a lower ferritin level might be a surrogate marker of a lower hepcidin status in this population. CONCLUSION: Recombinant human erythropoietin free patients seem to restore the EPO-hepcidin axis that is critical for erythropoiesis. A specific combination of clinical and biological parameters may help to detect future rHU-EPO free patients.


Subject(s)
Anemia/drug therapy , Erythropoietin/physiology , Hepcidins/physiology , Renal Dialysis , Renal Insufficiency, Chronic/complications , Adult , Aged , Aged, 80 and over , Anemia/etiology , Erythropoietin/therapeutic use , Female , Ferritins/blood , Humans , Male , Middle Aged , Recombinant Proteins/therapeutic use , Renal Insufficiency, Chronic/blood
16.
Arterioscler Thromb Vasc Biol ; 39(2): 178-187, 2019 02.
Article in English | MEDLINE | ID: mdl-30587002

ABSTRACT

Objective- Inflammatory stimuli enhance the progression of atherosclerotic disease. Inflammation also increases the expression of hepcidin, a hormonal regulator of iron homeostasis, which decreases intestinal iron absorption, reduces serum iron levels and traps iron within macrophages. The role of macrophage iron in the development of atherosclerosis remains incompletely understood. The objective of this study was to investigate the effects of hepcidin deficiency and decreased macrophage iron on the development of atherosclerosis. Approach and Results- Hepcidin- and LDL (low-density lipoprotein) receptor-deficient ( Hamp-/-/ Ldlr-/-) mice and Hamp+/+/ Ldlr-/- control mice were fed a high-fat diet for 21 weeks. Compared with control mice, Hamp-/-/ Ldlr-/- mice had decreased aortic macrophage activity and atherosclerosis. Because hepcidin deficiency is associated with both increased serum iron and decreased macrophage iron, the possibility that increased serum iron was responsible for decreased atherosclerosis in Hamp-/-/ Ldlr-/- mice was considered. Hamp+/+/ Ldlr-/- mice were treated with iron dextran so as to produce a 2-fold increase in serum iron. Increased serum iron did not decrease atherosclerosis in Hamp+/+/ Ldlr-/- mice. Aortic macrophages from Hamp-/-/ Ldlr-/- mice had less labile free iron and exhibited a reduced proinflammatory (M1) phenotype compared with macrophages from Hamp+/+/ Ldlr-/- mice. THP1 human macrophages treated with an iron chelator were used to model hepcidin deficiency in vitro. Treatment with an iron chelator reduced LPS (lipopolysaccharide)-induced M1 phenotypic expression and decreased uptake of oxidized LDL. Conclusions- In summary, in a hyperlipidemic mouse model, hepcidin deficiency was associated with decreased macrophage iron, a reduced aortic macrophage inflammatory phenotype and protection from atherosclerosis. The results indicate that decreasing hepcidin activity, with the resulting decrease in macrophage iron, may prove to be a novel strategy for the treatment of atherosclerosis.


Subject(s)
Atherosclerosis/etiology , Hepcidins/physiology , Animals , Atherosclerosis/prevention & control , Female , Hepcidins/deficiency , Iron/blood , Macrophages/physiology , Male , Mice , Mice, Inbred C57BL , Receptors, LDL/physiology
17.
Ann Biol Clin (Paris) ; 76(6): 705-715, 2018 Dec 01.
Article in English | MEDLINE | ID: mdl-30257815

ABSTRACT

Hepcidin has progressively become essential in clinical practice for the diagnosis and follow-up of a large spectrum of diseases. Anyway, its own biochemical and structural characteristics have complicated and delayed the acquisition of a standardized quantifying tool of the peptide.


Subject(s)
Hepcidins/analysis , Age Factors , Female , Gene Expression Regulation , Hepcidins/chemistry , Hepcidins/metabolism , Hepcidins/physiology , Humans , Immunoassay/methods , Inflammation/genetics , Inflammation/metabolism , Inflammation/pathology , Male
19.
PLoS One ; 13(5): e0196684, 2018.
Article in English | MEDLINE | ID: mdl-29738538

ABSTRACT

OBJECTIVES: Anemia is a known driver for hypoxia inducible factor (HIF) which leads to increased renal erythropoietin (EPO) synthesis. Bone marrow (BM) EPO receptor (EPOR) signals are transduced through a JAK2-STAT5 pathway. The origins of anemia of chronic kidney disease (CKD) are multifactorial, including impairment of both renal EPO synthesis as well as intestinal iron absorption. We investigated the HIF- EPO- EPOR axis in kidney, BM and proximal tibia in anemic juvenile CKD rats. METHODS: CKD was induced by 5/6 nephrectomy in young (20 days old) male Sprague-Dawley rats while C group was sham operated. Rats were sacrificed 4 weeks after CKD induction and 5 minutes after a single bolus of IV recombinant human EPO. An additional control anemic (C-A) group was daily bled for 7 days. RESULTS: Hemoglobin levels were similarly reduced in CKD and C-A (11.4 ± 0.3 and 10.8±0.2 Vs 13.5±0.3 g/dL in C, p<0.0001). Liver hepcidin mRNA was decreased in CA but increased in CKD. Serum iron was unchanged while transferrin levels were mildly decreased in CKD. Kidney HIF2α protein was elevated in C-A but unchanged in CKD. Kidney EPO protein and mRNA levels were unchanged between groups. However, BM EPO protein (which reflects circulating EPO) was increased in C-A but remained unchanged in CKD. BM and proximal tibia EPOR were unchanged in C-A but decreased in CKD. Proximal tibial phospho-STAT5 increased after the EPO bolus in C but not in CKD. CONCLUSIONS: Compared to blood loss, anemia in young CKD rats is associated with inappropriate responses in the HIF-EPO-EPO-R axis: kidney HIF2α and renal EPO are not increased, BM and bone EPOR levels, as well as bone pSTAT5 response to EPO are reduced. Thus, anemia of CKD may be treated with additional therapeutic avenues beyond iron and EPO supplementation.


Subject(s)
Anemia/etiology , Basic Helix-Loop-Helix Transcription Factors/physiology , Erythropoietin/physiology , Receptors, Erythropoietin/physiology , Renal Insufficiency, Chronic/complications , Signal Transduction/physiology , Anemia/physiopathology , Animals , Bone Marrow/metabolism , Disease Models, Animal , Erythropoietin/biosynthesis , Erythropoietin/genetics , Erythropoietin/pharmacology , Hepcidins/physiology , Kidney/metabolism , Liver/metabolism , Male , Nephrectomy/adverse effects , RNA, Messenger/biosynthesis , Rats , Rats, Sprague-Dawley , Recombinant Proteins/pharmacology , Renal Insufficiency, Chronic/blood , Renal Insufficiency, Chronic/physiopathology , STAT5 Transcription Factor/physiology
20.
Infect Immun ; 86(7)2018 07.
Article in English | MEDLINE | ID: mdl-29735522

ABSTRACT

Iron is an essential micronutrient for most microbes and their hosts. Mammalian hosts respond to infection by inducing the iron-regulatory hormone hepcidin, which causes iron sequestration and a rapid decrease in the plasma and extracellular iron concentration (hypoferremia). Previous studies showed that hepcidin regulation of iron is essential for protection from infection-associated mortality with the siderophilic pathogens Yersinia enterocolitica and Vibrio vulnificus However, the evolutionary conservation of the hypoferremic response to infection suggests that not only rare siderophilic bacteria but also common pathogens may be targeted by this mechanism. We tested 10 clinical isolates of Escherichia coli from children with sepsis and found that both genetic iron overload (by hepcidin-1 knockout [HKO]) and iatrogenic iron overload (by intravenous iron) potentiated infection with 8 out of the 10 studied isolates: after peritoneal injection of E. coli, iron-loaded mice developed sepsis with 60% to 100% mortality within 24 h, while control wild-type mice suffered 0% mortality. Using one strain for more detailed study, we show that iron overload allows rapid bacterial multiplication and dissemination. We further found that the presence of non-transferrin-bound iron (NTBI) in the circulation is more important than total plasma or tissue iron in rendering mice susceptible to infection and mortality. Postinfection treatment of HKO mice with just two doses of the hepcidin agonist PR73 abolished NTBI and completely prevented sepsis-associated mortality. We demonstrate that the siderophilic phenotype extends to clinically common pathogens. The use of hepcidin agonists promises to be an effective early intervention in patients with infections and dysregulated iron metabolism.


Subject(s)
Bacteremia/mortality , Escherichia coli Infections/mortality , Hepcidins/physiology , Animals , Bacteremia/etiology , Bacteremia/microbiology , Child , Escherichia coli Infections/etiology , Escherichia coli Infections/microbiology , Hepcidins/agonists , Humans , Iron/metabolism , Iron Overload/complications , Mice , Mice, Inbred C57BL , Mice, Knockout , Transferrin/analysis
SELECTION OF CITATIONS
SEARCH DETAIL
...