Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 20
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Nat Med ; 22(8): 945-51, 2016 08.
Artigo em Inglês | MEDLINE | ID: mdl-27428900

RESUMO

Iron is an essential component of the erythrocyte protein hemoglobin and is crucial to oxygen transport in vertebrates. In the steady state, erythrocyte production is in equilibrium with erythrocyte removal. In various pathophysiological conditions, however, erythrocyte life span is compromised severely, which threatens the organism with anemia and iron toxicity. Here we identify an on-demand mechanism that clears erythrocytes and recycles iron. We show that monocytes that express high levels of lymphocyte antigen 6 complex, locus C1 (LY6C1, also known as Ly-6C) ingest stressed and senescent erythrocytes, accumulate in the liver via coordinated chemotactic cues, and differentiate into ferroportin 1 (FPN1, encoded by SLC40A1)-expressing macrophages that can deliver iron to hepatocytes. Monocyte-derived FPN1(+)Tim-4(neg) macrophages are transient, reside alongside embryonically derived T cell immunoglobulin and mucin domain containing 4 (Timd4, also known as Tim-4)(high) Kupffer cells (KCs), and depend on the growth factor Csf1 and the transcription factor Nrf2 (encoded by Nfe2l2). The spleen, likewise, recruits iron-loaded Ly-6C(high) monocytes, but these do not differentiate into iron-recycling macrophages, owing to the suppressive action of Csf2. The accumulation of a transient macrophage population in the liver also occurs in mouse models of hemolytic anemia, anemia of inflammation, and sickle cell disease. Inhibition of monocyte recruitment to the liver during stressed erythrocyte delivery leads to kidney and liver damage. These observations identify the liver as the primary organ that supports rapid erythrocyte removal and iron recycling, and uncover a mechanism by which the body adapts to fluctuations in erythrocyte integrity.


Assuntos
Eritrócitos/metabolismo , Hepatócitos/metabolismo , Ferro/metabolismo , Células de Kupffer/metabolismo , Fígado/metabolismo , Anemia , Anemia Hemolítica , Anemia Falciforme , Animais , Antígenos Ly/metabolismo , Proteínas de Transporte de Cátions/metabolismo , Diferenciação Celular , Modelos Animais de Doenças , Eritrócitos/citologia , Fator Estimulador de Colônias de Granulócitos e Macrófagos/metabolismo , Inflamação , Células de Kupffer/citologia , Fator Estimulador de Colônias de Macrófagos/metabolismo , Macrófagos/citologia , Macrófagos/metabolismo , Proteínas de Membrana/metabolismo , Camundongos , Monócitos/citologia , Monócitos/metabolismo , Fator 2 Relacionado a NF-E2/metabolismo , Baço
2.
FASEB J ; 30(2): 813-23, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26506980

RESUMO

Because ferroportin (Fpn) is the only known mammalian cellular iron exporter, understanding its localization and regulation within the retina would shed light on the direction of retinal iron flux. The hormone hepcidin may regulate retinal Fpn, as it triggers Fpn degradation in the gut. Immunofluorescence was used to label Fpn in retinas of mice with 4 different genotypes (wild type; Fpn C326S, a hepcidin-resistant Fpn; hepcidin knockout; and ceruloplasmin/hephaestin double knockout). No significant difference in Fpn levels was observed in these retinas. Fpn localized to the abluminal side of the outer plexiform vascular endothelial cells, Müller glia cells, and the basolateral side of the retinal pigment epithelium. Adeno-associated virus (AAV)-hepcidin was injected into the eyes of hepcidin knockout mice, while AAV-lacZ was injected into the contralateral eyes as a control. AAV-hepcidin injected eyes had increased ferritin immunolabeling in retinal vascular endothelial cells. Fpn C326S mice had systemic iron overload compared to wild type and had the fastest retinal iron accumulation of any hereditary model studied to date. The results suggest that physiologic hepcidin levels are insufficient to alter Fpn levels within the retinal pigment epithelium and Müller cells, but may limit iron transport into the retina from vascular endothelial cells.


Assuntos
Proteínas de Transporte de Cátions/metabolismo , Hepcidinas/metabolismo , Ferro/metabolismo , Epitélio Pigmentado da Retina/metabolismo , Substituição de Aminoácidos , Animais , Proteínas de Transporte de Cátions/genética , Hepcidinas/genética , Camundongos , Camundongos Knockout , Mutação de Sentido Incorreto , Epitélio Pigmentado da Retina/patologia
3.
Eur J Immunol ; 45(11): 3073-86, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-26332507

RESUMO

Lipocalin-2 (Lcn2) is an innate immune peptide with pleiotropic effects. Lcn2 binds iron-laden bacterial siderophores, chemo-attracts neutrophils and has immunomodulatory and apoptosis-regulating effects. In this study, we show that upon infection with Salmonella enterica serovar Typhimurium, Lcn2 promotes iron export from Salmonella-infected macrophages, which reduces cellular iron content and enhances the generation of pro-inflammatory cytokines. Lcn2 represses IL-10 production while augmenting Nos2, TNF-α, and IL-6 expression. Lcn2(-/-) macrophages have elevated IL-10 levels as a consequence of increased iron content. The crucial role of Lcn-2/IL-10 interactions was further demonstrated by the greater ability of Lcn2(-/-) IL-10(-/-) macrophages and mice to control intracellular Salmonella proliferation in comparison to Lcn2(-/-) counterparts. Overexpression of the iron exporter ferroportin-1 in Lcn2(-/-) macrophages represses IL-10 and restores TNF-α and IL-6 production to the levels found in wild-type macrophages, so that killing and clearance of intracellular Salmonella is promoted. Our observations suggest that Lcn2 promotes host resistance to Salmonella Typhimurium infection by binding bacterial siderophores and suppressing IL-10 production, and that both functions are linked to its ability to shuttle iron from macrophages.


Assuntos
Proteínas de Fase Aguda/imunologia , Homeostase/imunologia , Ferro/metabolismo , Lipocalinas/imunologia , Macrófagos/metabolismo , Proteínas Oncogênicas/imunologia , Salmonelose Animal/imunologia , Proteínas de Fase Aguda/metabolismo , Animais , Western Blotting , Lipocalina-2 , Lipocalinas/metabolismo , Macrófagos/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteínas Oncogênicas/metabolismo , Reação em Cadeia da Polimerase em Tempo Real , Salmonelose Animal/metabolismo , Salmonella typhimurium , Transfecção
4.
Cell Metab ; 20(5): 787-798, 2014 Nov 04.
Artigo em Inglês | MEDLINE | ID: mdl-25444678

RESUMO

Cholesterol metabolism is closely interrelated with cardiovascular disease in humans. Dietary supplementation with omega-6 polyunsaturated fatty acids including arachidonic acid (AA) was shown to favorably affect plasma LDL-C and HDL-C. However, the underlying mechanisms are poorly understood. By combining data from a GWAS screening in >100,000 individuals of European ancestry, mediator lipidomics, and functional validation studies in mice, we identify the AA metabolome as an important regulator of cholesterol homeostasis. Pharmacological modulation of AA metabolism by aspirin induced hepatic generation of leukotrienes (LTs) and lipoxins (LXs), thereby increasing hepatic expression of the bile salt export pump Abcb11. Induction of Abcb11 translated in enhanced reverse cholesterol transport, one key function of HDL. Further characterization of the bioactive AA-derivatives identified LX mimetics to lower plasma LDL-C. Our results define the AA metabolomeasconserved regulator of cholesterol metabolism, and identify AA derivatives as promising therapeutics to treat cardiovascular disease in humans.


Assuntos
Ácido Araquidônico/metabolismo , Colesterol/metabolismo , Metaboloma , Animais , Anti-Inflamatórios não Esteroides/uso terapêutico , Araquidonato 5-Lipoxigenase/metabolismo , Aspirina/uso terapêutico , Aterosclerose/tratamento farmacológico , Aterosclerose/metabolismo , Ácidos e Sais Biliares/metabolismo , Células Cultivadas , Colesterol/sangue , HDL-Colesterol/sangue , HDL-Colesterol/metabolismo , Humanos , Leucotrienos/metabolismo , Fígado/efeitos dos fármacos , Fígado/metabolismo , Camundongos , Camundongos Endogâmicos C57BL
5.
Am J Pathol ; 184(11): 2862-7, 2014 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-25174877

RESUMO

The retina can be shielded by the blood-retinal barrier. Because photoreceptors are damaged by excess iron, it is important to understand whether the blood-retinal barrier protects against high serum iron levels. Bone morphogenic protein 6 (Bmp6) knockout mice have serum iron overload. Herein, we tested whether the previously documented retinal iron accumulation in Bmp6 knockout mice might result from the high serum iron levels or, alternatively, low levels of retinal hepcidin, an iron regulatory hormone whose transcription can be up-regulated by Bmp6. Furthermore, to determine whether increases in serum iron can elevate retinal iron levels, we i.v. injected iron into wild-type mice. Retinas were analyzed by real-time quantitative PCR and immunofluorescence to assess the levels of iron-regulated genes/proteins and oxidative stress. Retinal hepcidin mRNA levels in Bmp6 knockout retinas were the same as, or greater than, those in age-matched wild-type retinas, indicating that Bmp6 knockout does not cause retinal hepcidin deficiency. Changes in mRNA levels of L ferritin and transferrin receptor indicated increased retinal iron levels in i.v. iron-injected wild-type mice. Oxidative stress markers were elevated in photoreceptors of mice receiving i.v. iron. These findings suggest that elevated serum iron levels can overwhelm local retinal iron regulatory mechanisms.


Assuntos
Barreira Hematorretiniana/metabolismo , Sobrecarga de Ferro/metabolismo , Ferro/metabolismo , Retina/metabolismo , Animais , Proteína Morfogenética Óssea 6/genética , Proteína Morfogenética Óssea 6/metabolismo , Hepcidinas/genética , Hepcidinas/metabolismo , Ferro/sangue , Sobrecarga de Ferro/genética , Camundongos , Camundongos Knockout , Estresse Oxidativo/fisiologia
6.
Haematologica ; 99(9): 1516-24, 2014 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-24895335

RESUMO

Anemia of chronic disease is a multifactorial disorder, resulting mainly from inflammation-driven reticuloendothelial iron retention, impaired erythropoiesis, and reduced biological activity of erythropoietin. Erythropoiesis-stimulating agents have been used for the treatment of anemia of chronic disease, although with varying response rates and potential adverse effects. Serum concentrations of hepcidin, a key regulator of iron homeostasis, are increased in patients with anemia of chronic disease and linked to the pathogenesis of this disease, because hepcidin blocks cellular iron egress, thus limiting availability of iron for erythropoiesis. We tested whether serum hepcidin levels can predict and affect the therapeutic efficacy of erythropoiesis-stimulating agent treatment using a well-established rat model of anemia of chronic disease. We found that high pre-treatment hepcidin levels correlated with an impaired hematologic response to an erythropoiesis-stimulating agent in rats with anemia of chronic disease. Combined treatment with an erythropoiesis-stimulating agent and an inhibitor of hepcidin expression, LDN-193189, significantly reduced serum hepcidin levels, mobilized iron from tissue stores, increased serum iron levels and improved hemoglobin levels more effectively than did the erythropoiesis-stimulating agent or LDN-193189 monotherapy. In parallel, both the erythropoiesis-stimulating agent and erythropoiesis-stimulating agent/LDN-193189 combined reduced the expression of cytokines known to inhibit erythropoiesis. We conclude that serum hepcidin levels can predict the hematologic responsiveness to erythropoiesis-stimulating agent therapy in anemia of chronic disease. Pharmacological inhibition of hepcidin formation improves the erythropoiesis-stimulating agent's therapeutic efficacy, which may favor a reduction of erythropoiesis-stimulating agent dosages, costs and side effects.


Assuntos
Anemia/tratamento farmacológico , Eritropoetina/farmacologia , Hematínicos/farmacologia , Hepcidinas/genética , Ferro/sangue , RNA Mensageiro/genética , Anemia/sangue , Anemia/induzido quimicamente , Anemia/patologia , Animais , Biomarcadores/sangue , Doença Crônica , Combinação de Medicamentos , Sinergismo Farmacológico , Eritropoese/efeitos dos fármacos , Feminino , Expressão Gênica , Hepcidinas/antagonistas & inibidores , Hepcidinas/sangue , Humanos , Interferon gama/antagonistas & inibidores , Interferon gama/biossíntese , Ferro/agonistas , Polissacarídeos Bacterianos , Prognóstico , Pirazóis/farmacologia , Pirimidinas/farmacologia , RNA Mensageiro/antagonistas & inibidores , RNA Mensageiro/sangue , Ratos , Ratos Endogâmicos Lew , Fator de Necrose Tumoral alfa/antagonistas & inibidores , Fator de Necrose Tumoral alfa/biossíntese
7.
Haematologica ; 96(12): 1761-9, 2011 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-21859731

RESUMO

BACKGROUND: Increased levels of hepcidin, the master regulator of iron homeostasis, contribute to the diversion of iron underlying the anemia of chronic disease. Yet hepcidin levels are low in anemia of chronic disease with concomitant true iron deficiency. Here we clarify the different underlying pathways regulating hepcidin expression under these conditions in vivo. DESIGN AND METHODS: We used rat models of iron deficiency anemia, anemia of chronic disease and anemia of chronic disease with concomitant true iron deficiency and investigated upstream signaling pathways controlling hepcidin transcription in the liver. Protein and mRNA levels of iron metabolism genes and genes involved in SMAD1/5/8 and STAT3 signaling were determined by RT-PCR, Western blotting and immunohistochemistry. RESULTS: SMAD1/5/8 phosphorylation and in parallel hepcidin mRNA expression were increased in anemia of chronic disease but significantly down-regulated in anemia of chronic disease with concomitant iron deficiency, either on the basis of phlebotomy or dietary iron restriction. Iron deficiency resulted in reduced bone morphogenetic protein-6 expression and impaired SMAD1/5/8 phosphorylation and trafficking, two key events for hepcidin transcription. Reduced SMAD1/5/8 activity in association with phlebotomy was paralleled by increased expression of the inhibitory factor, SMAD7, dietary iron restriction appeared to impair hepcidin transactivating SMAD pathways via reduction of membrane bound hemojuvelin expression. CONCLUSIONS: This study evaluated hepcidin signaling pathways in anemia of chronic disease with/without concomitant iron deficiency in vivo. While iron deficiency in general decreased bone morphogenetic protein-6 expression, phlebotomy or dietary iron restriction inhibited inflammation driven SMAD1/5/8 mediated hepcidin formation by different pathways, indicating alternate hierarchic signaling networks as a function of the mode and kinetics of iron deficiency. Nonetheless, iron deficiency inducible regulatory pathways can reverse inflammation mediated stimulation of hepcidin expression.


Assuntos
Anemia Ferropriva/metabolismo , Peptídeos Catiônicos Antimicrobianos/biossíntese , Regulação da Expressão Gênica , Fígado/metabolismo , Transdução de Sinais , Anemia Ferropriva/patologia , Animais , Doença Crônica , Modelos Animais de Doenças , Feminino , Hepcidinas , Ferro/metabolismo , Fígado/patologia , Fosforilação , RNA Mensageiro/biossíntese , Ratos , Ratos Endogâmicos Lew , Fator de Transcrição STAT3/metabolismo , Proteínas Smad/metabolismo
8.
Blood ; 118(18): 4977-84, 2011 Nov 03.
Artigo em Inglês | MEDLINE | ID: mdl-21730356

RESUMO

Anemia of chronic inflammation (ACI) is the most frequent anemia in hospitalized patients and is associated with significant morbidity. A major underlying mechanism of ACI is the retention of iron within cells of the reticuloendothelial system (RES), thus making the metal unavailable for efficient erythropoiesis. This reticuloendothelial iron sequestration is primarily mediated by excess levels of the iron regulatory peptide hepcidin down-regulating the functional expression of the only known cellular iron export protein ferroportin resulting in blockade of iron egress from these cells. Using a well-established rat model of ACI, we herein provide novel evidence for effective treatment of ACI by blocking endogenous hepcidin production using the small molecule dorsomorphin derivative LDN-193189 or the protein soluble hemojuvelin-Fc (HJV.Fc) to inhibit bone morphogenetic protein-Smad mediated signaling required for effective hepcidin transcription. Pharmacologic inhibition of hepcidin expression results in mobilization of iron from the RES, stimulation of erythropoiesis and correction of anemia. Thus, hepcidin lowering agents are a promising new class of pharmacologic drugs to effectively combat ACI.


Assuntos
Anemia/tratamento farmacológico , Peptídeos Catiônicos Antimicrobianos/antagonistas & inibidores , Inflamação/tratamento farmacológico , Pirazóis/farmacologia , Pirimidinas/farmacologia , Anemia/etiologia , Anemia/genética , Animais , Peptídeos Catiônicos Antimicrobianos/genética , Peptídeos Catiônicos Antimicrobianos/metabolismo , Células Cultivadas , Doença Crônica , Modelos Animais de Doenças , Avaliação Pré-Clínica de Medicamentos , Feminino , Proteínas Ligadas por GPI , Expressão Gênica/efeitos dos fármacos , Proteína da Hemocromatose , Hepcidinas , Fragmentos Fc das Imunoglobulinas/uso terapêutico , Inflamação/complicações , Inflamação/genética , Proteínas de Membrana/imunologia , Ratos , Ratos Endogâmicos Lew , Indução de Remissão
9.
PLoS One ; 6(3): e16762, 2011 Mar 08.
Artigo em Inglês | MEDLINE | ID: mdl-21408141

RESUMO

The mouse is a valuable model for unravelling the role of hepcidin in iron homeostasis, however, such studies still report hepcidin mRNA levels as a surrogate marker for bioactive hepcidin in its pivotal function to block ferroportin-mediated iron transport. Here, we aimed to assess bioactive mouse Hepcidin-1 (Hep-1) and its paralogue Hepcidin-2 (Hep-2) at the peptide level. To this purpose, Fourier transform ion cyclotron resonance (FTICR) and tandem-MS was used for hepcidin identification, after which a time-of-flight (TOF) MS-based methodology was exploited to routinely determine Hep-1 and -2 levels in mouse serum and urine. This method was biologically validated by hepcidin assessment in: i) 3 mouse strains (C57Bl/6; DBA/2 and BABL/c) upon stimulation with intravenous iron and LPS, ii) homozygous Hfe knock out, homozygous transferrin receptor 2 (Y245X) mutated mice and double affected mice, and iii) mice treated with a sublethal hepatotoxic dose of paracetamol. The results showed that detection of Hep-1 was restricted to serum, whereas Hep-2 and its presumed isoforms were predominantly present in urine. Elevations in serum Hep-1 and urine Hep-2 upon intravenous iron or LPS were only moderate and varied considerably between mouse strains. Serum Hep-1 was decreased in all three hemochromatosis models, being lowest in the double affected mice. Serum Hep-1 levels correlated with liver hepcidin-1 gene expression, while acute liver damage by paracetamol depleted Hep-1 from serum. Furthermore, serum Hep-1 appeared to be an excellent indicator of splenic iron accumulation. In conclusion, Hep-1 and Hep-2 peptide responses in experimental mouse agree with the known biology of hepcidin mRNA regulators, and their measurement can now be implemented in experimental mouse models to provide novel insights in post-transcriptional regulation, hepcidin function, and kinetics.


Assuntos
Peptídeos Catiônicos Antimicrobianos/química , Espectrometria de Massas , Animais , Peptídeos Catiônicos Antimicrobianos/sangue , Peptídeos Catiônicos Antimicrobianos/genética , Peptídeos Catiônicos Antimicrobianos/urina , Modelos Animais de Doenças , Regulação da Expressão Gênica/efeitos dos fármacos , Hemocromatose/sangue , Hemocromatose/genética , Hepcidinas , Interleucina-6/sangue , Ferro/metabolismo , Ferro/farmacologia , Lipopolissacarídeos/farmacologia , Fígado/efeitos dos fármacos , Fígado/metabolismo , Camundongos , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Baço/efeitos dos fármacos , Baço/metabolismo
10.
Immunity ; 34(1): 61-74, 2011 Jan 28.
Artigo em Inglês | MEDLINE | ID: mdl-21256055

RESUMO

Erythropoietin (EPO) is the principal cytokine regulating erythropoiesis through its receptor, EPOR. Interestingly, EPORs are also found on immune cells with incompletely understood functions. Here, we show that EPO inhibits the induction of proinflammatory genes including tumor necrosis factor (TNF)-α and inducible nitric oxide (NO) synthase in activated macrophages, which is mechanistically attributable to blockage of nuclear factor (NF)-κB p65 activation by EPO. Accordingly, in systemic Salmonella infection, treatment of mice with EPO results in reduced survival and impaired pathogen clearance because of diminished formation of anti-microbial effector molecules such as TNF-α and NO. However, neutralization of endogenous EPO or genetic ablation of Epor promotes Salmonella elimination. In contrast, in chemically induced colitis, EPO-EPOR interaction decreases the production of NF-κB-inducible immune mediators, thus limiting tissue damage and ameliorating disease severity. These immune-modulatory effects of EPO may be of therapeutic relevance in infectious and inflammatory diseases.


Assuntos
Colite/imunologia , Eritropoetina/administração & dosagem , Macrófagos Peritoneais/efeitos dos fármacos , NF-kappa B/metabolismo , Receptores da Eritropoetina/metabolismo , Infecções por Salmonella/imunologia , Salmonella/imunologia , Animais , Linhagem Celular , Colite/induzido quimicamente , Colite/tratamento farmacológico , Sulfato de Dextrana/administração & dosagem , Humanos , Mediadores da Inflamação/metabolismo , Macrófagos Peritoneais/imunologia , Macrófagos Peritoneais/metabolismo , Macrófagos Peritoneais/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Óxido Nítrico/imunologia , Óxido Nítrico/metabolismo , Receptores da Eritropoetina/genética , Salmonella/patogenicidade , Infecções por Salmonella/tratamento farmacológico , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/genética , Transdução de Sinais/imunologia , Ácido Trinitrobenzenossulfônico/administração & dosagem , Fator de Necrose Tumoral alfa/genética , Fator de Necrose Tumoral alfa/imunologia , Fator de Necrose Tumoral alfa/metabolismo
11.
J Hepatol ; 53(6): 1101-7, 2010 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-20801540

RESUMO

BACKGROUND & AIMS: Aceruloplasminemia is a rare autosomal recessive neurodegenerative disease associated with brain and liver iron accumulation which typically presents with movement disorders, retinal degeneration, and diabetes mellitus. Ceruloplasmin is a multi-copper ferroxidase that is secreted into plasma and facilitates cellular iron export and iron binding to transferrin. RESULTS: A novel homozygous ceruloplasmin gene mutation, c.2554+1G>T, was identified as the cause of aceruloplasminemia in three affected siblings. Two siblings presented with movement disorders and diabetes. Complementary DNA sequencing showed that this mutation causes skipping of exon 14 and deletion of amino acids 809-852 while preserving the open reading frame. Western blotting of liver extracts and sera of affected patients showed retention of the abnormal protein in the liver. Aceruloplasminemia was associated with severe brain and liver iron overload, where hepatic mRNA expression of the iron hormone hepcidin was increased, corresponding to the degree of iron overload. Hepatic iron concentration normalized after 3 and 5months of iron chelation therapy with deferasirox, which was also associated with reduced insulin demands. During short term treatment there was no clinical or imaging evidence for significant effects on brain iron overload. CONCLUSIONS: Aceruloplasminemia can show an incomplete clinical penetrance but is invariably associated with iron accumulation in the liver and in the brain. Iron accumulation in aceruloplasminemia is a result of defective cellular iron export, where hepcidin regulation is appropriate for the degree of iron overload. Iron chelation with deferasirox was effective in mobilizing hepatic iron but has no effect on brain iron.


Assuntos
Benzoatos/uso terapêutico , Ceruloplasmina/genética , Quelantes de Ferro/uso terapêutico , Ferro/metabolismo , Mutação , Triazóis/uso terapêutico , Peptídeos Catiônicos Antimicrobianos/genética , Encéfalo/metabolismo , Ceruloplasmina/deficiência , Ceruloplasmina/metabolismo , Consanguinidade , Deferasirox , Feminino , Hepcidinas , Homozigoto , Humanos , Distúrbios do Metabolismo do Ferro/tratamento farmacológico , Distúrbios do Metabolismo do Ferro/genética , Distúrbios do Metabolismo do Ferro/metabolismo , Fígado/metabolismo , Masculino , Pessoa de Meia-Idade , Doenças Neurodegenerativas/tratamento farmacológico , Doenças Neurodegenerativas/genética , Doenças Neurodegenerativas/metabolismo , Linhagem , RNA Mensageiro/genética , RNA Mensageiro/metabolismo
12.
Br J Haematol ; 148(3): 449-55, 2010 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-19863534

RESUMO

Recently, the iron and erythropoiesis-controlled growth differentiation factor 15 (GDF15) has been shown to inhibit the expression of hepcidin in beta-thalassaemia patients, thereby increasing iron absorption despite iron overload. To access the diagnostic and pathogenic impact of GDF15 in inflammatory anaemia the association of GDF15 expression with serum iron parameters and hepcidin was studied in patients suffering from iron deficiency anaemia (IDA), anaemia of chronic disease (ACD) and ACD subjects with true iron deficiency (ACD/IDA). GDF15 was significantly increased in both ACD and ACD/IDA, but not in IDA subjects as compared to controls. In contrast, hepcidin levels were significantly lower in IDA and ACD/IDA subjects than in ACD patients. IDA and ACD/IDA, but not ACD, showed an association between GDF15 and soluble transferrin receptor, an indicator of iron requirement for erythropoiesis. However, GDF15 did not correlate to hepcidin in either patient group. While GDF15 levels were linked to the needs for erythropoiesis and iron homeostasis in IDA, the immunity-driven increase of GDF15 may not primarily affect iron homeostasis and hepcidin expression. This indicates that other ACD-related factors may overcome the regulatory effects of GDF15 on hepcidin expression during inflammation.


Assuntos
Anemia/sangue , Fator 15 de Diferenciação de Crescimento/sangue , Adulto , Idoso , Anemia/etiologia , Anemia Ferropriva/sangue , Peptídeos Catiônicos Antimicrobianos/sangue , Biomarcadores/sangue , Estudos de Casos e Controles , Doença Crônica , Feminino , Fator 15 de Diferenciação de Crescimento/fisiologia , Hepcidinas , Humanos , Masculino , Pessoa de Meia-Idade
13.
Blood ; 114(17): 3642-51, 2009 Oct 22.
Artigo em Inglês | MEDLINE | ID: mdl-19700664

RESUMO

Mutations of HFE are associated with hereditary hemochromatosis, but their influence on host susceptibility to infection is incompletely understood. We report that mice lacking one or both Hfe alleles are protected from septicemia with Salmonella Typhimurium, displaying prolonged survival and improved control of bacterial replication. This increased resistance is paralleled by an enhanced production of the enterochelin-binding peptide lipocalin-2 (Lcn2), which reduces the availability of iron for Salmonella within Hfe-deficient macrophages. Accordingly, Hfe(-/-)Lcn2(-/-) macrophages are unable to efficiently control the infection or to withhold iron from intracellular Salmonella. Correspondingly, the protection conferred by the Hfe defect is abolished in Hfe(-/-) mice infected with enterochelin-deficient Salmonella as well as in Hfe(-/-)Lcn2(-/-) mice infected with wild-type bacteria. Thus, by induction of the iron-capturing peptide Lcn2, absence of functional Hfe confers host resistance to systemic infection with Salmonella, thereby providing an evolutionary advantage which may account for the high prevalence of genetic hemochromatosis.


Assuntos
Proteínas de Fase Aguda/metabolismo , Antígenos de Histocompatibilidade Classe I/fisiologia , Lipocalinas/metabolismo , Proteínas de Membrana/fisiologia , Proteínas Oncogênicas/metabolismo , Salmonelose Animal/prevenção & controle , Salmonella typhimurium/fisiologia , Animais , Proteínas de Bactérias/metabolismo , Células Cultivadas , Citocinas/metabolismo , Modelos Animais de Doenças , Feminino , Regulação Bacteriana da Expressão Gênica , Proteína da Hemocromatose , Ferro/metabolismo , Lipocalina-2 , Macrófagos/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Nitritos/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Salmonelose Animal/genética , Salmonelose Animal/metabolismo , Salmonelose Animal/microbiologia
14.
Blood ; 113(21): 5277-86, 2009 May 21.
Artigo em Inglês | MEDLINE | ID: mdl-19293425

RESUMO

The anemia of chronic disease (ACD) is characterized by macrophage iron retention induced by cytokines and the master regulator hepcidin. Hepcidin controls cellular iron efflux on binding to the iron export protein ferroportin. Many patients, however, present with both ACD and iron deficiency anemia (ACD/IDA), the latter resulting from chronic blood loss. We used a rat model of ACD resulting from chronic arthritis and mimicked ACD/IDA by additional phlebotomy to define differing iron-regulatory pathways. Iron retention during inflammation occurs in macrophages and the spleen, but not in the liver. In rats and humans with ACD, serum hepcidin concentrations are elevated, which is paralleled by reduced duodenal and macrophage expression of ferroportin. Individuals with ACD/IDA have significantly lower hepcidin levels than ACD subjects, and ACD/IDA persons, in contrast to ACD subjects, were able to absorb dietary iron from the gut and to mobilize iron from macrophages. Circulating hepcidin levels affect iron traffic in ACD and ACD/IDA and are more responsive to the erythropoietic demands for iron than to inflammation. Hepcidin determination may aid to differentiate between ACD and ACD/IDA and in selecting appropriate therapy for these patients.


Assuntos
Anemia Ferropriva/patologia , Anemia/patologia , Homeostase , Ferro/metabolismo , Anemia/sangue , Anemia/metabolismo , Anemia Ferropriva/sangue , Anemia Ferropriva/metabolismo , Animais , Peptídeos Catiônicos Antimicrobianos/sangue , Estudos de Casos e Controles , Proteínas de Transporte de Cátions/análise , Doença Crônica , Modelos Animais de Doenças , Duodeno/metabolismo , Hepcidinas , Humanos , Inflamação , Macrófagos/metabolismo , Macrófagos/patologia , Ratos , Ratos Endogâmicos Lew , Baço/metabolismo , Baço/patologia
15.
J Mol Med (Berl) ; 86(7): 825-35, 2008 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-18521557

RESUMO

Hepcidin, a small cationic liver derived peptide, is a master regulator of body iron homeostasis. Cytokines and iron availability have so far been identified as regulators of hepcidin expression. Herein, we investigated the functional role of Kupffer cells for hepcidin expression because of their vicinity to the hepatocytes and their importance for iron recycling via erythrophagocytosis. We investigated C57Bl6 mice and littermates, in which Kupffer cells were eliminated in vivo upon intravenous injection of liposome-encapsulated clodronate. Primary cultures of hepatocytes and Kupffer cells were used to study direct regulatory effects ex vivo. The in vivo depletion of Kupffer cells resulted in a significant increase in liver hepcidin expression, which was paralleled by a significant reduction in serum iron levels. The same pattern of regulation by Kupffer cell depletion was observed upon injection of bacterial lipopolysaccharide into mice and in primary (Hfe -/-) and in secondary iron-overloaded mice. Accordingly, the messenger ribonucleic acid (mRNA) concentrations of the hepcidin iron-sensing molecule hemojuvelin were not significantly changed upon Kupffer cell depletion. When primary hepatocytes were cocultivated with Kupffer cells or stimulated with a Kupffer cell-conditioned medium ex vivo, a significant reduction in hepatocyte hepcidin mRNA expression was observed. Our data suggest that Kupffer cells control body iron homeostasis by exerting negative regulatory signals toward hepcidin expression, which may be primarily referred to the secretion of yet unidentified hepcidin-suppressing molecules by Kupffer cells.


Assuntos
Peptídeos Catiônicos Antimicrobianos/genética , Regulação da Expressão Gênica , Ferro/metabolismo , Células de Kupffer/metabolismo , Animais , Peptídeos Catiônicos Antimicrobianos/metabolismo , Linhagem Celular Tumoral , Hepatócitos/metabolismo , Hepcidinas , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , RNA Mensageiro/metabolismo
16.
Am J Clin Nutr ; 87(5): 1374-83, 2008 May.
Artigo em Inglês | MEDLINE | ID: mdl-18469261

RESUMO

BACKGROUND: Mild iron overload is frequently observed in nonalcoholic fatty liver disease (NAFLD). OBJECTIVE: We aimed to study putative pathways underlying iron accumulation in NAFLD. DESIGN: Hepatic and duodenal expression of critical iron molecules in NAFLD patients with (n = 32) and without (n = 29) iron overload, hereditary hemochromatosis (n = 10), and controls (n = 20) were investigated. Phlebotomy treatment was performed in 14 NAFLD patients. RESULTS: The hepatic expressions of the iron-export protein ferroportin-1 (FP-1) and of the iron-sensing molecule hemojuvelin (HJV) were significantly lower in NAFLD patients. The mRNA expression of the iron-regulatory peptide hepcidin was increased in NAFLD patients with iron overload, which was paralleled by low duodenal FP-1 expression. Hepatic mRNA and serum protein concentrations of tumor necrosis factor-alpha (TNF-alpha) were increased in NAFLD patients and were inversely correlated with both liver FP-1 and HJV mRNA and positively associated with body mass index and hepatic hepcidin mRNA. Accordingly, TNF-alpha inhibited the FP-1 and HJV mRNA formation in HepG2 cells. Phlebotomy treatment of NALFD patients reduced serum ferritin, transferrin saturation, and TNF-alpha concentrations and improved liver function tests. CONCLUSIONS: Iron accumulation in NAFLD may result from an impaired iron export due to down-regulation of FP1 and ineffective hepatic iron sensing, as indicated by low HJV expression. TNF-alpha appears to play a role in exerting these regulatory changes. Increased hepcidin formation in iron-overloaded NAFLD patients, however, results in decreased duodenal FP-1 expression, whereas a reduction in liver FP-1 may perpetuate hepatic iron retention. Phlebotomy offers a safe and efficient therapy for these metabolic disturbances.


Assuntos
Peptídeos Catiônicos Antimicrobianos/metabolismo , Proteínas de Transporte de Cátions/metabolismo , Fígado Gorduroso/metabolismo , Hemocromatose/metabolismo , Ferro/metabolismo , Proteínas de Membrana/metabolismo , Peptídeos Catiônicos Antimicrobianos/genética , Proteínas de Transporte de Cátions/genética , Regulação para Baixo , Duodeno/metabolismo , Fígado Gorduroso/fisiopatologia , Fígado Gorduroso/terapia , Feminino , Proteínas Ligadas por GPI , Expressão Gênica , Hemocromatose/genética , Hemocromatose/fisiopatologia , Hemocromatose/terapia , Proteína da Hemocromatose , Hepcidinas , Humanos , Sobrecarga de Ferro/etiologia , Masculino , Proteínas de Membrana/genética , Pessoa de Meia-Idade , Flebotomia , RNA Mensageiro/metabolismo , Fator de Necrose Tumoral alfa/metabolismo
17.
Blood ; 111(4): 2392-9, 2008 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-18073346

RESUMO

Hepcidin, a master regulator of iron homeostasis, is produced in small amounts by inflammatory monocytes/macrophages. Chronic immune activation leads to iron retention within monocytes/macrophages and the development of anemia of chronic disease (ACD). We questioned whether monocyte-derived hepcidin exerts autocrine regulation toward cellular iron metabolism. Monocyte hepcidin mRNA expression was significantly induced within 3 hours after stimulation with LPS or IL-6, and hepcidin mRNA expression was significantly higher in monocytes of ACD patients than in controls. In ACD patients, monocyte hepcidin mRNA levels were significantly correlated to serum IL-6 concentrations, and increased monocyte hepcidin mRNA levels were associated with decreased expression of the iron exporter ferroportin and iron retention in these cells. Transient transfection experiments using a ferroportin/EmGFP fusion protein construct demonstrated that LPS inducible hepcidin expression in THP-1 monocytes resulted in internalization and degradation of ferroportin. Transfection of monocytes with siRNA directed against hepcidin almost fully reversed this lipopolysaccharide-mediated effect. Using ferroportin mutation constructs, we found that ferroportin is mainly targeted by hepcidin when expressed on the cell surface. Our results suggest that ferroportin expression in inflammatory monocytes is negatively affected by autocrine formation of hepcidin, thus contributing to iron sequestration within monocytes as found in ACD.


Assuntos
Anemia/sangue , Peptídeos Catiônicos Antimicrobianos/genética , Ferro/metabolismo , Monócitos/fisiologia , Anemia/etiologia , Peptídeos Catiônicos Antimicrobianos/biossíntese , Peptídeos Catiônicos Antimicrobianos/sangue , Proteína C-Reativa/metabolismo , Doença Crônica , Feminino , Hepcidinas , Homeostase , Humanos , Masculino , RNA Mensageiro/genética
18.
Cell Microbiol ; 9(9): 2126-40, 2007 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-17466014

RESUMO

In being both, a modifier of cellular immune effector pathways and an essential nutrient for microbes, iron is a critical determinant in host-pathogen interaction. Here, we investigated the metabolic changes of macrophage iron homeostasis and immune function following the infection of RAW264.7 murine macrophages with Salmonella typhimurium. We observed an enhanced expression of the principal iron export protein, ferroportin 1, and a subsequent increase of iron efflux in Salmonella-infected phagocytes. In parallel, the expression of haem oxygenase 1 and of the siderophore-binding peptide lipocalin 2 was markedly enhanced following pathogen entry. Collectively, these modulations reduced both the cytoplasmatic labile iron and the ferritin storage iron pool within macrophages, thus restricting the acquisition of iron by intramacrophage Salmonella. Correspondingly, limitation of macrophage iron decreased microbial survival, whereas iron supplementation impaired immune response pathways in Salmonella-infected macrophages (nitric oxide formation and tumour necrosis factor-alpha production) and promoted intracellular bacterial proliferation. Our findings suggest that the enhancement of ferroportin 1-mediated iron efflux, the upregulation of the haem-degrading enzyme haem oxygenase 1 and the induction of lipocalin 2 following infection concordantly aim at withholding iron from intracellular S. typhimurium and to increase antimicrobial immune effector pathways thus limiting pathogen proliferation.


Assuntos
Homeostase , Ferro/metabolismo , Macrófagos/metabolismo , Macrófagos/microbiologia , Salmonella typhimurium/imunologia , Proteínas de Fase Aguda/metabolismo , Animais , Peptídeos Catiônicos Antimicrobianos/genética , Peptídeos Catiônicos Antimicrobianos/metabolismo , Proteínas de Transporte de Cátions/genética , Proteínas de Transporte de Cátions/metabolismo , Células Cultivadas , Ferritinas/genética , Ferritinas/metabolismo , Heme Oxigenase-1/metabolismo , Hepcidinas , Lipocalina-2 , Lipocalinas/metabolismo , Macrófagos/citologia , Camundongos , Proteínas Oncogênicas/metabolismo , Fagócitos/metabolismo , Fagócitos/microbiologia , Salmonella typhimurium/patogenicidade , Fator de Necrose Tumoral alfa/metabolismo
19.
Nat Med ; 13(4): 448-54, 2007 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-17293870

RESUMO

Hereditary hemochromatosis and transfusional iron overload are frequent clinical conditions associated with progressive iron accumulation in parenchymal tissues, leading to eventual organ failure. We have discovered a new mechanism to reverse iron overload-pharmacological modulation of the divalent metal transporter-1 (DMT-1). DMT-1 mediates intracellular iron transport during the transferrin cycle and apical iron absorption in the duodenum. Its additional functions in iron handling in the kidney and liver are less well understood. We show that the L-type calcium channel blocker nifedipine increases DMT-1-mediated cellular iron transport 10- to 100-fold at concentrations between 1 and 100 microM. Mechanistically, nifedipine causes this effect by prolonging the iron-transporting activity of DMT-1. We show that nifedipine mobilizes iron from the liver of mice with primary and secondary iron overload and enhances urinary iron excretion. Modulation of DMT-1 function by L-type calcium channel blockers emerges as a new pharmacological therapy for the treatment of iron overload disorders.


Assuntos
Bloqueadores dos Canais de Cálcio/farmacologia , Proteínas de Transporte de Cátions/metabolismo , Hemocromatose/prevenção & controle , Sobrecarga de Ferro/tratamento farmacológico , Nifedipino/farmacologia , Animais , Transporte Biológico Ativo/efeitos dos fármacos , Células COS , Bloqueadores dos Canais de Cálcio/uso terapêutico , Chlorocebus aethiops , Eletrofisiologia , Humanos , Immunoblotting , Ferro/metabolismo , Ferro/urina , Fígado/metabolismo , Camundongos , Camundongos Knockout , Análise em Microsséries , Nifedipino/uso terapêutico , Reação em Cadeia da Polimerase Via Transcriptase Reversa
20.
J Immunol ; 178(3): 1748-58, 2007 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-17237424

RESUMO

Adipocytokines are mainly adipocyte-derived cytokines regulating metabolism and as such are key regulators of insulin resistance. Some adipocytokines such as adiponectin and leptin affect immune and inflammatory functions. Visfatin (pre-B cell colony-enhancing factor) has recently been identified as a new adipocytokine affecting insulin resistance by binding to the insulin receptor. In this study, we show that recombinant visfatin activates human leukocytes and induces cytokine production. In CD14(+) monocytes, visfatin induces the production of IL-1beta, TNF-alpha, and especially IL-6. Moreover, it increases the surface expression of costimulatory molecules CD54, CD40, and CD80. Visfatin-stimulated monocytes show augmented FITC-dextran uptake and an enhanced capacity to induce alloproliferative responses in human lymphocytes. Visfatin-induced effects involve p38 as well as MEK1 pathways as determined by inhibition with MAPK inhibitors and we observed activation of NF-kappaB. In vivo, visfatin induces circulating IL-6 in BALB/c mice. In patients with inflammatory bowel disease, plasma levels of visfatin are elevated and its mRNA expression is significantly increased in colonic tissue of Crohn's and ulcerative colitis patients compared with healthy controls. Macrophages, dendritic cells, and colonic epithelial cells might be additional sources of visfatin as determined by confocal microscopy. Visfatin can be considered a new proinflammatory adipocytokine.


Assuntos
Citocinas/genética , Citocinas/farmacologia , Adipócitos , Animais , Estudos de Casos e Controles , Células Cultivadas , Citocinas/biossíntese , Citocinas/efeitos dos fármacos , Humanos , Fatores Imunológicos , Inflamação , Doenças Inflamatórias Intestinais/genética , Leucócitos/efeitos dos fármacos , Leucócitos/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Monócitos/efeitos dos fármacos , Monócitos/metabolismo , Nicotinamida Fosforribosiltransferase , RNA Mensageiro/sangue , Proteínas Recombinantes , Transdução de Sinais
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...