Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Sci Rep ; 12(1): 6695, 2022 04 23.
Artigo em Inglês | MEDLINE | ID: mdl-35461329

RESUMO

In chronic kidney disease, ferric citrate has been shown to be an effective phosphate binder and source of enteral iron; however, the effects of ferric citrate on the kidney have been less well-studied. Here, in Col4α3 knockout mice-a murine model of progressive chronic kidney disease, we evaluated the effects of five weeks of 1% ferric citrate dietary supplementation. As expected, ferric citrate lowered serum phosphate concentrations and increased serum iron levels in the Col4α3 knockout mice. Consistent with decreased enteral phosphate absorption and possibly improved iron status, ferric citrate greatly reduced circulating fibroblast growth factor 23 levels. Interestingly, ferric citrate also lessened systemic inflammation, improved kidney function, reduced albuminuria, and decreased kidney inflammation and fibrosis, suggesting renoprotective effects of ferric citrate in the setting of chronic kidney disease. The factors mediating possible ferric citrate renoprotection, the mechanisms by which they may act, and whether ferric citrate affects chronic kidney disease progression in humans deserves further study.


Assuntos
Compostos Férricos , Insuficiência Renal Crônica , Animais , Modelos Animais de Doenças , Feminino , Compostos Férricos/farmacologia , Humanos , Inflamação , Ferro , Masculino , Camundongos , Camundongos Knockout , Fosfatos
2.
Kidney Int ; 101(4): 711-719, 2022 04.
Artigo em Inglês | MEDLINE | ID: mdl-34838540

RESUMO

Ferric citrate is approved as an iron replacement product in patients with non-dialysis chronic kidney disease and iron deficiency anemia. Ferric citrate-delivered iron is enterally absorbed, but the specific mechanisms involved have not been evaluated, including the possibilities of conventional, transcellular ferroportin-mediated absorption and/or citrate-mediated paracellular absorption. Here, we first demonstrate the efficacy of ferric citrate in high hepcidin models, including Tmprss6 knockout mice (characterized by iron-refractory iron deficiency anemia) with and without adenine diet-induced chronic kidney disease. Next, to assess whether or not enteral ferric citrate absorption is dependent on ferroportin, we evaluated the effects of ferric citrate in a tamoxifen-inducible, enterocyte-specific ferroportin knockout murine model (Villin-Cre-ERT2, Fpnflox/flox). In this model, ferroportin deletion was efficient, as tamoxifen injection induced a 4000-fold decrease in duodenum ferroportin mRNA expression, with undetectable ferroportin protein on Western blot of duodenal enterocytes, resulting in a severe iron deficiency anemia phenotype. In ferroportin-deficient mice, three weeks of 1% ferric citrate dietary supplementation, a dose that prevented iron deficiency in control mice, did not improve iron status or rescue the iron deficiency anemia phenotype. We repeated the conditional ferroportin knockout experiment in the setting of uremia, using an adenine nephropathy model, where three weeks of 1% ferric citrate dietary supplementation again failed to improve iron status or rescue the iron deficiency anemia phenotype. Thus, our data suggest that enteral ferric citrate absorption is dependent on conventional enterocyte iron transport by ferroportin and that, in these models, significant paracellular absorption does not occur.


Assuntos
Anemia Ferropriva , Proteínas de Transporte de Cátions , Anemia Ferropriva/tratamento farmacológico , Animais , Proteínas de Transporte de Cátions/genética , Compostos Férricos/farmacologia , Hepcidinas/metabolismo , Humanos , Ferro/metabolismo , Camundongos
3.
Kidney Int ; 100(1): 79-89, 2021 07.
Artigo em Inglês | MEDLINE | ID: mdl-33811979

RESUMO

Vadadustat is an investigational hypoxia-inducible factor prolyl hydroxylase inhibitor that increases endogenous erythropoietin production and has been shown to decrease hepcidin levels, ameliorate iron restriction, and increase hemoglobin concentrations in anemic patients with chronic kidney disease (CKD). In studies of physiological responses to other erythropoietic stimuli, erythropoietin induced erythroblast secretion of erythroferrone (ERFE), which acts on the liver to suppress hepcidin production and mobilize iron for erythropoiesis. We therefore investigated whether vadadustat effects on erythropoiesis and iron metabolism are dependent on ERFE. Wild type and ERFE knockout mice with and without CKD were treated with vadadustat or vehicle. In both wild type and ERFE knockout CKD models, vadadustat was similarly effective, as evidenced by normalized hemoglobin concentrations, increased expression of duodenal iron transporters, lower serum hepcidin levels, and decreased tissue iron concentrations. This is consistent with ERFE-independent increased iron mobilization. Vadadustat treatment also lowered serum urea nitrogen and creatinine concentrations and decreased expression of kidney fibrosis markers. Lastly, vadadustat affected fibroblast growth factor 23 (FGF23) profiles: in non-CKD mice, vadadustat increased plasma total FGF23 out of proportion to intact FGF23, consistent with the known effects of hypoxia-inducible factor-1α and erythropoietin on FGF23 production and metabolism. However, in the mice with CKD, vadadustat markedly decreased both total and intact FGF23, effects likely contributed to by the reduced loss of kidney function. Thus, in this CKD model, vadadustat ameliorated anemia independently of ERFE, improved kidney parameters, and decreased FGF23. How vadadustat affects CKD progression in humans warrants future studies.


Assuntos
Anemia , Eritropoetina , Insuficiência Renal Crônica , Anemia/tratamento farmacológico , Anemia/etiologia , Animais , Fator de Crescimento de Fibroblastos 23 , Glicina/análogos & derivados , Hepcidinas , Humanos , Rim , Camundongos , Camundongos Knockout , Ácidos Picolínicos , Insuficiência Renal Crônica/complicações , Insuficiência Renal Crônica/tratamento farmacológico
4.
Nephrol Dial Transplant ; 34(12): 2057-2065, 2019 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-30007314

RESUMO

BACKGROUND: Erythropoietin (EPO) has been reported as a novel determinant of fibroblast growth factor 23 (FGF23) production; however, it is unknown whether FGF23 is stimulated by chronic exposure to EPO or by EPO administration in nonpolycystic chronic kidney disease (CKD) models. METHODS: We analyzed the effects of chronic EPO on FGF23 in murine models with chronically high EPO levels and normal kidney function. We studied the effects of exogenous EPO on FGF23 in wild-type mice, with and without CKD, injected with EPO. Also, in four independent human CKD cohorts, we evaluated associations between FGF23 and serum EPO levels or exogenous EPO dose. RESULTS: Mice with high endogenous EPO have elevated circulating total FGF23, increased disproportionately to intact FGF23, suggesting coupling of increased FGF23 production with increased proteolytic cleavage. Similarly, in wild-type mice with and without CKD, a single exogenous EPO dose acutely increases circulating total FGF23 out of proportion to intact FGF23. In these murine models, the bone marrow is shown to be a novel source of EPO-stimulated FGF23 production. In humans, serum EPO levels and recombinant human EPO dose are positively and independently associated with total FGF23 levels across the spectrum of CKD and after kidney transplantation. In our largest cohort of 680 renal transplant recipients, serum EPO levels are associated with total FGF23, but not intact FGF23, consistent with the effects of EPO on FGF23 production and metabolism observed in our murine models. CONCLUSION: EPO affects FGF23 production and metabolism, which may have important implications for CKD patients.


Assuntos
Eritropoetina/metabolismo , Fatores de Crescimento de Fibroblastos/metabolismo , Regulação da Expressão Gênica , Insuficiência Renal Crônica/patologia , Talassemia beta/patologia , Animais , Estudos de Coortes , Feminino , Fator de Crescimento de Fibroblastos 23 , Humanos , Transplante de Rim , Masculino , Camundongos , Camundongos Transgênicos , Pessoa de Meia-Idade , Prognóstico , Insuficiência Renal Crônica/metabolismo , Insuficiência Renal Crônica/cirurgia , Talassemia beta/metabolismo
5.
Physiol Rep ; 6(19): e13890, 2018 09.
Artigo em Inglês | MEDLINE | ID: mdl-30315639

RESUMO

Erythropoietin (EPO) acts on erythroid progenitor cells to promote their survival and differentiation to mature erythrocytes. Along with this canonical role, EPO is also reported to modulate energy metabolism, resulting in improved glucose tolerance and insulin sensitivity. EPO also stimulates the production of the hormone erythroferrone (ERFE) which acts to suppress hepcidin production, thus increasing dietary iron absorption and mobilizing stored iron for use in erythropoiesis. ERFE (initially termed myonectin) was also reported have an effect on systemic lipid metabolism by promoting the clearance of nonesterifed fatty acids (NEFA) from circulation. As increased levels of circulating NEFA blunt insulin sensitivity and impair glucose tolerance, ERFE-induced clearance of NEFA after EPO administration would have a beneficial effect on glucose metabolism. The aim of this study was to determine if the known metabolic effect of EPO treatment on glucose homeostasis is mediated by ERFE, produced in response to EPO. Mice lacking Erfe did not differ from wild-type mice in blood lipid parameters, blood glucose, and glucose or insulin tolerance at baseline or after chronic EPO treatment. Additionally, hepcidin suppression and the response of erythrocyte parameters to chronic EPO treatment were unaffected by the absence of Erfe. These findings suggest that the known beneficial effects of EPO on glucose metabolism are not attributable to an accompanying increase in ERFE production, and that Erfe is dispensable for normal glucose homeostasis. Furthermore, our data indicate that ERFE-independent mechanisms can suppress hepcidin in response to chronically elevated EPO levels.


Assuntos
Glicemia/efeitos dos fármacos , Glicemia/metabolismo , Citocinas/deficiência , Eritropoetina/administração & dosagem , Fármacos Hematológicos/administração & dosagem , Proteínas Musculares/deficiência , Animais , Feminino , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout
7.
Blood ; 130(3): 245-257, 2017 07 20.
Artigo em Inglês | MEDLINE | ID: mdl-28465342

RESUMO

The iron-regulatory hormone hepcidin is induced early in infection, causing iron sequestration in macrophages and decreased plasma iron; this is proposed to limit the replication of extracellular microbes, but could also promote infection with macrophage-tropic pathogens. The mechanisms by which hepcidin and hypoferremia modulate host defense, and the spectrum of microbes affected, are poorly understood. Using mouse models, we show that hepcidin was selectively protective against siderophilic extracellular pathogens (Yersinia enterocolitica O9) by controlling non-transferrin-bound iron (NTBI) rather than iron-transferrin concentration. NTBI promoted the rapid growth of siderophilic but not nonsiderophilic bacteria in mice with either genetic or iatrogenic iron overload and in human plasma. Hepcidin or iron loading did not affect other key components of innate immunity, did not indiscriminately promote intracellular infections (Mycobacterium tuberculosis), and had no effect on extracellular nonsiderophilic Y enterocolitica O8 or Staphylococcus aureus Hepcidin analogs may be useful for treatment of siderophilic infections.


Assuntos
Infecções Relacionadas a Cateter/imunologia , Hemocromatose/imunologia , Hepcidinas/imunologia , Sobrecarga de Ferro/imunologia , Ferro/metabolismo , Infecções Estafilocócicas/imunologia , Animais , Ligação Competitiva , Infecções Relacionadas a Cateter/metabolismo , Infecções Relacionadas a Cateter/microbiologia , Infecções Relacionadas a Cateter/mortalidade , Modelos Animais de Doenças , Resistência à Doença , Expressão Gênica , Hemocromatose/metabolismo , Hemocromatose/microbiologia , Hemocromatose/mortalidade , Hepcidinas/agonistas , Hepcidinas/deficiência , Hepcidinas/genética , Humanos , Ferro/imunologia , Sobrecarga de Ferro/metabolismo , Sobrecarga de Ferro/microbiologia , Sobrecarga de Ferro/mortalidade , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Mycobacterium tuberculosis/efeitos dos fármacos , Mycobacterium tuberculosis/crescimento & desenvolvimento , Mycobacterium tuberculosis/metabolismo , Oligopeptídeos/farmacologia , Ligação Proteica , Infecções Estafilocócicas/metabolismo , Infecções Estafilocócicas/microbiologia , Infecções Estafilocócicas/mortalidade , Staphylococcus aureus , Análise de Sobrevida , Transferrina/genética , Transferrina/metabolismo , Yersinia enterocolitica/efeitos dos fármacos , Yersinia enterocolitica/crescimento & desenvolvimento , Yersinia enterocolitica/metabolismo
8.
Shock ; 48(4): 490-497, 2017 10.
Artigo em Inglês | MEDLINE | ID: mdl-28452907

RESUMO

Intensive care unit (ICU) anemia is an extreme version of anemia of inflammation that occurs commonly in critically ill patients and is associated with increased morbidity and mortality. Currently available therapies for ICU anemia have shown inconsistent efficacies in clinical trials. We conducted a systematic study of the effects of early versus delayed iron (Fe) and/or erythropoietin (EPO) therapy in our previously characterized mouse model of ICU anemia based on an injection of heat-killed Brucella abortus. To study the effects of ongoing inflammation on the response to therapy, inflamed wild-type (WT) and hepcidin knockout (HKO) mice were treated at either early (days 1 and 2) or delayed (days 7 and 8) time points after the inflammatory stimulus. In the early treatment group, Fe and/or EPO therapy did not increase hemoglobin (Hgb) levels or reticulocyte production in either the inflamed WT or HKO groups. In the delayed treatment group, combination Fe + EPO therapy did increase Hgb and reticulocyte production in WT mice (mean ΔHgb in WT saline group -9.2 g/dL vs. Fe/EPO -5.5 g/dL; P < 0.001). The HKO mice in the delayed treatment group did not improve their Hgb, but HKO mice in all treatment groups developed a milder anemia than the WT mice. Our findings indicate that combination Fe + EPO therapy is effective in partially reversing ICU anemia when administered after the phase of acute inflammation. Hepcidin ablation alone was more effective in attenuating ICU anemia than Fe + EPO therapy, which indicates the potential of antihepcidin therapeutics in treating ICU anemia.


Assuntos
Anemia/tratamento farmacológico , Eritropoetina/farmacologia , Hepcidinas/antagonistas & inibidores , Ferro/farmacologia , Sepse/tratamento farmacológico , Anemia/genética , Anemia/metabolismo , Anemia/patologia , Animais , Modelos Animais de Doenças , Feminino , Hepcidinas/genética , Masculino , Camundongos , Camundongos Knockout , Sepse/genética , Sepse/metabolismo , Sepse/patologia
11.
Am J Physiol Renal Physiol ; 311(6): F1369-F1377, 2016 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-27733366

RESUMO

In the setting of normal kidney function, iron deficiency is associated with increased FGF23 production and cleavage, altering circulating FGF23 levels. Our objective was to determine how chronic kidney disease (CKD) and dietary iron intake affect FGF23 production and metabolism in wild-type (WT) and hepcidin knockout (HKO) mice. For 8 wk, the mice were fed diets that contained adenine (to induce CKD) or no adenine (control group), with either low-iron (4 ppm) or standard-iron (335 ppm) concentrations. The low-iron diet induced iron deficiency anemia in both the WT and HKO mice. Among the WT mice, in both the control and CKD groups, a low-iron compared with a standard-iron diet increased bone Fgf23 mRNA expression, C-terminal FGF23 (cFGF23) levels, and FGF23 cleavage as manifested by a lower percentage intact FGF23 (iFGF23). Independent of iron status, CKD was associated with inhibition of FGF23 cleavage. Similar results were observed in the HKO control and CKD groups. Dietary iron content was more influential on FGF23 parameters than the presence or absence of hepcidin. In the CKD mice (WT and HKO, total n = 42), independent of the effects of serum phosphate, iron deficiency was associated with increased FGF23 production but also greater cleavage, whereas worse kidney function was associated with increased FGF23 production but decreased cleavage. Therefore, in both the WT and HKO mouse models, dietary iron content and CKD affected FGF23 production and metabolism.


Assuntos
Fatores de Crescimento de Fibroblastos/metabolismo , Ferro da Dieta/administração & dosagem , Rim/efeitos dos fármacos , Insuficiência Renal Crônica/metabolismo , Adenina , Animais , Fator de Crescimento de Fibroblastos 23 , Hepcidinas/genética , Hepcidinas/metabolismo , Rim/metabolismo , Camundongos , Camundongos Knockout , Insuficiência Renal Crônica/induzido quimicamente
12.
Blood Cells Mol Dis ; 56(1): 31-6, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-26603720

RESUMO

Acute and severe anemia of inflammation (AI) is a common complication of various clinical syndromes, including fulminant infections, critical illness with multiorgan failure, and exacerbations of autoimmune diseases. Building on recent data showing beneficial results with isocitrate treatment for chronic low-grade AI in a rat model, we used a mouse model of acute and severe AI induced by intraperitoneal heat-killed Brucella abortus to determine if isocitrate would be effective in this more stringent application. Inflamed mice treated with isocitrate developed an early but transient improvement in hemoglobin compared to solvent-treated controls, with a robust improvement on day 7, and only a trend towards improvement by day 14. Reticulocyte counts were increased in treated mice transiently, with no significant difference by day 21. Serum erythropoietin (EPO) levels were similar in treated versus control mice, indicating that isocitrate increased sensitivity to EPO. Serum and tissue iron levels showed no significant differences between the treated and control mice, ruling out improved iron availability as the cause of the increased response to endogenous EPO. Compared to the milder rat model, much higher doses of isocitrate were required for a relatively modest benefit.


Assuntos
Anemia/complicações , Anemia/tratamento farmacológico , Inflamação/complicações , Inflamação/tratamento farmacológico , Isocitratos/uso terapêutico , Doença Aguda , Anemia/sangue , Anemia/microbiologia , Animais , Brucella abortus/isolamento & purificação , Brucelose/sangue , Brucelose/complicações , Brucelose/tratamento farmacológico , Brucelose/microbiologia , Modelos Animais de Doenças , Eritropoese/efeitos dos fármacos , Eritropoetina/sangue , Hepcidinas/análise , Inflamação/sangue , Inflamação/microbiologia , Ferro/análise , Masculino , Camundongos
13.
Blood ; 126(17): 2031-7, 2015 Oct 22.
Artigo em Inglês | MEDLINE | ID: mdl-26276665

RESUMO

Inherited anemias with ineffective erythropoiesis, such as ß-thalassemia, manifest inappropriately low hepcidin production and consequent excessive absorption of dietary iron, leading to iron overload. Erythroferrone (ERFE) is an erythroid regulator of hepcidin synthesis and iron homeostasis. Erfe expression was highly increased in the marrow and spleen of Hbb(Th3/+) mice (Th3/+), a mouse model of thalassemia intermedia. Ablation of Erfe in Th3/+ mice restored normal levels of circulating hepcidin at 6 weeks of age, suggesting ERFE could be a factor suppressing hepcidin production in ß-thalassemia. We examined the expression of Erfe and the consequences of its ablation in thalassemic mice from 3 to 12 weeks of age. The loss of ERFE in thalassemic mice led to full restoration of hepcidin mRNA expression at 3 and 6 weeks of age, and significant reduction in liver and spleen iron content at 6 and 12 weeks of age. Ablation of Erfe slightly ameliorated ineffective erythropoiesis, as indicated by reduced spleen index, red cell distribution width, and mean corpuscular volume, but did not improve the anemia. Thus, ERFE mediates hepcidin suppression and contributes to iron overload in a mouse model of ß-thalassemia.


Assuntos
Citocinas/fisiologia , Modelos Animais de Doenças , Hepcidinas/metabolismo , Sobrecarga de Ferro/etiologia , Proteínas Musculares/fisiologia , Talassemia beta/complicações , Talassemia beta/patologia , Animais , Ensaio de Imunoadsorção Enzimática , Eritropoese/fisiologia , Feminino , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , RNA Mensageiro/genética , Reação em Cadeia da Polimerase em Tempo Real , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Talassemia beta/genética
14.
Cell Host Microbe ; 17(1): 47-57, 2015 Jan 14.
Artigo em Inglês | MEDLINE | ID: mdl-25590758

RESUMO

Hereditary hemochromatosis, an iron overload disease caused by a deficiency in the iron-regulatory hormone hepcidin, is associated with lethal infections by siderophilic bacteria. To elucidate the mechanisms of this susceptibility, we infected wild-type and hepcidin-deficient mice with the siderophilic bacterium Vibrio vulnificus and found that hepcidin deficiency results in increased bacteremia and decreased survival of infected mice, which can be partially ameliorated by dietary iron depletion. Additionally, timely administration of hepcidin agonists to hepcidin-deficient mice induces hypoferremia that decreases bacterial loads and rescues these mice from death, regardless of initial iron levels. Studies of Vibrio vulnificus growth ex vivo show that high iron sera from hepcidin-deficient mice support extraordinarily rapid bacterial growth and that this is inhibited in hypoferremic sera. Our findings demonstrate that hepcidin-mediated hypoferremia is a host defense mechanism against siderophilic pathogens and suggest that hepcidin agonists may improve infection outcomes in patients with hereditary hemochromatosis or thalassemia.


Assuntos
Bacteriemia/imunologia , Hepcidinas/metabolismo , Ferro/metabolismo , Vibrioses/imunologia , Vibrio vulnificus/crescimento & desenvolvimento , Vibrio vulnificus/imunologia , Animais , Bacteriemia/microbiologia , Carga Bacteriana , Mecanismos de Defesa , Hepcidinas/deficiência , Ferro/sangue , Camundongos Endogâmicos C57BL , Camundongos Knockout , Vibrioses/microbiologia
15.
PLoS One ; 9(3): e93283, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24681760

RESUMO

Anemia of cancer (AC) may contribute to cancer-related fatigue and impair quality of life. Improved understanding of the pathogenesis of AC could facilitate better treatment, but animal models to study AC are lacking. We characterized four syngeneic C57BL/6 mouse cancers that cause AC. Mice with two different rapidly-growing metastatic lung cancers developed the characteristic findings of anemia of inflammation (AI), with dramatically different degrees of anemia. Mice with rapidly-growing metastatic melanoma also developed a severe anemia by 14 days, with hematologic and inflammatory parameters similar to AI. Mice with a slow-growing peritoneal ovarian cancer developed an iron-deficiency anemia, likely secondary to chronically impaired nutrition and bleeding into the peritoneal cavity. Of the four models, hepcidin mRNA levels were increased only in the milder lung cancer model. Unlike in our model of systemic inflammation induced by heat-killed Brucella abortus, ablation of hepcidin in the ovarian cancer and the milder lung cancer mouse models did not affect the severity of anemia. Hepcidin-independent mechanisms play an important role in these murine models of AC.


Assuntos
Anemia Ferropriva/etiologia , Anemia Ferropriva/patologia , Neoplasias/complicações , Neoplasias/patologia , Anemia Ferropriva/genética , Animais , Brucella abortus/metabolismo , Modelos Animais de Doenças , Feminino , Hepcidinas/genética , Inflamação/genética , Inflamação/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Neoplasias/genética , Qualidade de Vida , RNA Mensageiro/genética
16.
Infect Immun ; 82(2): 745-52, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24478088

RESUMO

Hepcidin, the iron-regulatory hormone, is increased during infection or inflammation, causing hypoferremia. This response is thought to be a host defense mechanism that restricts iron availability to invading pathogens. It is not known if hepcidin is differentially induced by bacterial versus viral infections, whether the stimulation of pattern recognition receptors directly regulates hepcidin transcription, or which of the proposed signaling pathways are essential for hepcidin increase during infection. We analyzed hepcidin induction and its dependence on interleukin-6 (IL-6) in response to common bacterial or viral infections in mice or in response to a panel of pathogen-derived molecules (PAMPs) in mice and human primary hepatocytes. In wild-type (WT) mice, hepcidin mRNA was induced several hundred-fold both by a bacterial (Streptococcus pneumoniae) and a viral infection (influenza virus PR8) within 2 to 5 days. Treatment of mice and human primary hepatocytes with most Toll-like receptor ligands increased hepcidin mRNA within 6 h. Hepcidin induction by microbial stimuli was IL-6 dependent. IL-6 knockout mice failed to increase hepcidin in response to S. pneumoniae or influenza infection and had greatly diminished hepcidin response to PAMPs. In vitro, hepcidin induction by PAMPs in primary human hepatocytes was abolished by the addition of neutralizing IL-6 antibodies. Our results support the key role of IL-6 in hepcidin regulation in response to a variety of infectious and inflammatory stimuli.


Assuntos
Hepatócitos/microbiologia , Hepatócitos/virologia , Hepcidinas/biossíntese , Interleucina-6/metabolismo , Animais , Hepatócitos/imunologia , Hepatócitos/metabolismo , Humanos , Interleucina-6/imunologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Orthomyxoviridae/imunologia , Streptococcus pneumoniae/imunologia
17.
Blood ; 123(8): 1129-36, 2014 Feb 20.
Artigo em Inglês | MEDLINE | ID: mdl-24357728

RESUMO

Anemia is a common complication of infections and inflammatory diseases, but the few mouse models of this condition are not well characterized. We analyzed in detail the pathogenesis of anemia induced by an injection of heat-killed Brucella abortus and examined the contribution of hepcidin by comparing wild-type (WT) to iron-depleted hepcidin-1 knockout (Hamp-KO) mice. B abortus-treated WT mice developed severe anemia with a hemoglobin nadir at 14 days and partial recovery by 28 days. After an early increase in inflammatory markers and hepcidin, WT mice manifested hypoferremia, despite iron accumulation in the liver. Erythropoiesis was suppressed between days 1 and 7, and erythrocyte destruction was increased as evidenced by schistocytes on blood smears and shortened red blood cell lifespan. Erythropoietic recovery began after 14 days but was iron restricted. In B abortus-treated Hamp-KO compared with WT mice, anemia was milder, not iron restricted, and had a faster recovery. Similarly to severe human anemia of inflammation, the B abortus model shows multifactorial pathogenesis of inflammatory anemia including iron restriction from increased hepcidin, transient suppression of erythropoiesis, and shortened erythrocyte lifespan. Ablation of hepcidin relieves iron restriction and improves the anemia.


Assuntos
Anemia/imunologia , Brucella abortus , Brucelose/imunologia , Hepcidinas/imunologia , Inflamação/imunologia , Doença Aguda , Anemia/genética , Animais , Doença Crônica , Modelos Animais de Doenças , Eritropoese/imunologia , Hemólise/imunologia , Hepcidinas/genética , Temperatura Alta , Humanos , Inflamação/genética , Inflamação/microbiologia , Ferro/farmacologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout
18.
Cell Rep ; 5(5): 1436-42, 2013 Dec 12.
Artigo em Inglês | MEDLINE | ID: mdl-24316081

RESUMO

Hepcidin, the iron-regulatory hormone and acute phase reactant, is proposed to contribute to the pathogenesis of atherosclerosis by promoting iron accumulation in plaque macrophages, leading to increased oxidative stress and inflammation in the plaque (the "iron hypothesis"). Hepcidin and iron may thus represent modifiable risk factors in atherosclerosis. We measured hepcidin expression in Apoe(-/-) mice with varying diets and ages. To assess the role of macrophage iron in atherosclerosis, we generated Apoe(-/-) mice with macrophage-specific iron accumulation by introducing the ferroportin ffe mutation. Macrophage iron loading was also enhanced by intravenous iron injection. Contrary to the iron hypothesis, we found that hepatic hepcidin expression was not increased at any stage of the atherosclerosis progression in Apoe(-/-) or Apoe/ffe mice and that the atherosclerotic plaque size was not increased in mice with elevated macrophage iron. Our results strongly argue against any significant role of macrophage iron in atherosclerosis progression in mice.


Assuntos
Aterosclerose/metabolismo , Ferro/metabolismo , Animais , Apolipoproteínas E/genética , Aterosclerose/genética , Aterosclerose/patologia , Proteínas de Transporte de Cátions/genética , Proteínas de Transporte de Cátions/metabolismo , Dieta Hiperlipídica/efeitos adversos , Modelos Animais de Doenças , Hepcidinas/genética , Hepcidinas/metabolismo , Ferro da Dieta/efeitos adversos , Macrófagos/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Mutação , Placa Aterosclerótica/etiologia , Placa Aterosclerótica/metabolismo
19.
Hepatology ; 53(4): 1333-41, 2011 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-21480335

RESUMO

UNLABELLED: In response to iron loading, hepcidin synthesis is homeostatically increased to limit further absorption of dietary iron and its release from stores. Mutations in HFE, transferrin receptor 2 (Tfr2), hemojuvelin (HJV), or bone morphogenetic protein 6 (BMP6) prevent appropriate hepcidin response to iron, allowing increased absorption of dietary iron, and eventually iron overload. To understand the role each of these proteins plays in hepcidin regulation by iron, we analyzed hepcidin messenger RNA (mRNA) responsiveness to short and long-term iron challenge in iron-depleted Hfe, Tfr2, Hjv, and Bmp6 mutant mice. After 1-day (acute) iron challenge, Hfe(-/-) mice showed a smaller hepcidin increase than their wild-type strain-matched controls, Bmp6(-/-) mice showed nearly no increase, and Tfr2 and Hjv mutant mice showed no increase in hepcidin expression, indicating that all four proteins participate in hepcidin regulation by acute iron changes. After a 21-day (chronic) iron challenge, Hfe and Tfr2 mutant mice increased hepcidin expression to nearly wild-type levels, but a blunted increase of hepcidin was seen in Bmp6(-/-) and Hjv(-/-) mice. BMP6, whose expression is also regulated by iron, may mediate hepcidin regulation by iron stores. None of the mutant strains (except Bmp6(-/-) mice) had impaired BMP6 mRNA response to chronic iron loading. CONCLUSION: TfR2, HJV, BMP6, and, to a lesser extent, HFE are required for the hepcidin response to acute iron loading, but are partially redundant for hepcidin regulation during chronic iron loading and are not involved in the regulation of BMP6 expression. Our findings support a model in which acute increases in holotransferrin concentrations transmitted through HFE, TfR2, and HJV augment BMP receptor sensitivity to BMPs. A distinct regulatory mechanism that senses hepatic iron may modulate hepcidin response to chronic iron loading.


Assuntos
Peptídeos Catiônicos Antimicrobianos/genética , Ferro/farmacologia , Animais , Proteína Morfogenética Óssea 6/genética , Modelos Animais de Doenças , Regulação da Expressão Gênica , Hemocromatose/fisiopatologia , Proteína da Hemocromatose , Hepcidinas , Antígenos de Histocompatibilidade Classe I/genética , Ferro/metabolismo , Proteínas de Membrana/genética , Camundongos , RNA Mensageiro/metabolismo , Receptores da Transferrina/genética , Transferrina/farmacologia
20.
Blood Cells Mol Dis ; 40(1): 122-31, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-17869549

RESUMO

As the principal iron-regulatory hormone, hepcidin plays an important role in systemic iron homeostasis. The regulation of hepcidin expression by iron loading appears to be unexpectedly complex and has attracted much interest. The GPI-linked membrane protein hemojuvelin (GPI-hemojuvelin) is an essential upstream regulator of hepcidin expression. A soluble form of hemojuvelin (s-hemojuvelin) exists in blood and acts as antagonist of GPI-hemojuvelin to downregulate hepcidin expression. The release of s-hemojuvelin is negatively regulated by both transferrin-bound iron (holo-Tf) and non-transferrin-bound iron (FAC), indicating s-hemojuvelin could be one of the mediators of hepcidin regulation by iron. In this report, we investigate the proteinase involved in the release of s-hemojuvelin and show that s-hemojuvelin is released by a proprotein convertase through the cleavage at a conserved polybasic RNRR site.


Assuntos
Aminoácidos Básicos , Proteínas de Membrana/metabolismo , Pró-Proteína Convertases/metabolismo , Sequência de Aminoácidos , Animais , Peptídeos Catiônicos Antimicrobianos/antagonistas & inibidores , Sítios de Ligação , Células Cultivadas , Sequência Conservada , Proteínas Ligadas por GPI , Proteína da Hemocromatose , Hepcidinas , Humanos , Camundongos , Solubilidade
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...