Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 36
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
2.
Blood ; 144(1): 117-121, 2024 Jul 04.
Artículo en Inglés | MEDLINE | ID: mdl-38527216

RESUMEN

ABSTRACT: Intravenous injection of excess apotransferrin enhances dietary iron absorption in mice and triggers accumulation of plasma non-transferrin-bound iron. Injected fluorescent-labeled transferrin colocalizes with lamina propria macrophages, consistent with the recently proposed iron absorption checkpoint involving macrophage-mediated transferrin degradation.


Asunto(s)
Apoproteínas , Hierro de la Dieta , Transferrina , Animales , Transferrina/metabolismo , Ratones , Hierro de la Dieta/metabolismo , Hierro de la Dieta/farmacocinética , Hierro de la Dieta/administración & dosificación , Apoproteínas/metabolismo , Macrófagos/metabolismo , Absorción Intestinal , Hierro/metabolismo , Ratones Endogámicos C57BL
3.
FASEB J ; 37(12): e23257, 2023 12.
Artículo en Inglés | MEDLINE | ID: mdl-37902616

RESUMEN

Cardiomyopathy is a major complication of thalassemia, yet the precise underlying molecular mechanisms remain unclear. We examined whether altered lipid metabolism is an early driving factor in the development of cardiomyopathy using the Th3/+ mouse model of thalassemia. At age 20 weeks, male and female Th3/+ mice manifested anemia and iron overload; however, only males displayed metabolic defects and altered cardiac function. Untargeted lipidomics indicated that the circulating levels of 35 lipid species were significantly altered in Th3/+ mice compared to wild-type controls: triglycerides (TGs) with saturated fatty acids (FAs; TG42:0 and TG44:0) were elevated, while TGs with unsaturated FAs (TG(18:2_20:5_18:2 and TG54:8)) were reduced. Similarly, phosphatidylcholines (PCs) with long chain FAs (palmitic (16:0) or oleic (18:1)) were increased, while PCs with polyunsaturated FAs decreased. Circulating PC(16:0_14:0), GlcCer(d18:1/24:0) correlated significantly with iron overload and cardiac hypertrophy. 16S rRNA gene profiling revealed alterations in the intestinal microbiota of Th3/+ mice. Differentially abundant bacterial genera correlated with PC(39:6), PC(18:1_22:6), GlcCer(d18:1/24:1) and CE(14:0). These results provide new knowledge on perturbations in lipid metabolism and the gut microbiota of Th3/+ mice and identify specific factors which may represent early biomarkers or therapeutic targets to prevent development of cardiomyopathy in ß-thalassemia.


Asunto(s)
Cardiomiopatías , Microbioma Gastrointestinal , Cardiopatías , Sobrecarga de Hierro , Talasemia , Femenino , Masculino , Animales , Ratones , Metabolismo de los Lípidos , ARN Ribosómico 16S , Talasemia/complicaciones , Modelos Animales de Enfermedad , Glucosilceramidas , Sobrecarga de Hierro/complicaciones , Triglicéridos
4.
J Acquir Immune Defic Syndr ; 94(3): 273-279, 2023 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-37368933

RESUMEN

BACKGROUND: People with HIV and hepatitis C virus (HCV) coinfection experience excess mortality because of multiple causes. Identification of biomarkers associated with mortality beyond that attributable to liver fibrosis may be relevant for prognostication. Fibroblast growth factor 23 (FGF23), a phosphotropic hormone, predicts adverse outcomes in several chronic conditions. We aimed to investigate whether elevated FGF23 predicts all-cause mortality in patients with HIV/HCV coinfection. METHODS: We included patients with HIV/HCV coinfection from the Canadian Coinfection Cohort with available serum FGF23, fibrosis biomarker fibrosis-4 (FIB-4), and at least 1-year follow-up. Elevated FGF23 and advanced liver fibrosis were defined as FGF23 > 241 reference unit/mL and FIB-4 > 3.25, respectively. All-cause mortality was analyzed using survival analysis. The effect of advanced liver fibrosis as a mediator on mortality was estimated by mediation analysis. RESULTS: Three hundred twenty-one patients were included (24% with elevated FGF23, 19% with advanced liver fibrosis). During a mean follow-up period of 8.4 years, 34% of the cohort died. The incidence rate of all-cause mortality was higher in patients with elevated FGF23 (66.1 per 1000 person-years, 95% confidence interval 45.8 to 92.3) relative to patients without elevated FGF23 (37.5 per 1000 person-years, 95% confidence interval 29.6 to 46.9). After adjusting for potential confounders, elevated FGF23 was associated with significant direct and indirect effects (mediated through advanced liver fibrosis) on all-cause mortality, with 57% of deaths not mediated through advanced fibrosis. CONCLUSIONS: In patients with HIV/HCV coinfection, FGF23 may be used as prognostic biomarker for risk stratification accounting also for death causes other than those attributable to liver fibrosis.


Asunto(s)
Coinfección , Infecciones por VIH , Hepatitis C , Humanos , Infecciones por VIH/complicaciones , Hepacivirus , Factor-23 de Crecimiento de Fibroblastos , Coinfección/complicaciones , Canadá/epidemiología , Hepatitis C/complicaciones , Cirrosis Hepática/complicaciones , Biomarcadores
5.
Int J Mol Sci ; 24(6)2023 Mar 08.
Artículo en Inglés | MEDLINE | ID: mdl-36982241

RESUMEN

Hepcidin is a liver-derived hormone that controls systemic iron traffic. It is also expressed in the heart, where it acts locally. We utilized cell and mouse models to study the regulation, expression, and function of cardiac hepcidin. Hepcidin-encoding Hamp mRNA was induced upon differentiation of C2C12 cells to a cardiomyocyte-like phenotype and was not further stimulated by BMP6, BMP2, or IL-6, the major inducers of hepatic hepcidin. The mRNAs encoding hepcidin and its upstream regulator hemojuvelin (Hjv) are primarily expressed in the atria of the heart, with ~20-fold higher Hamp mRNA levels in the right vs. left atrium and negligible expression in the ventricles and apex. Hjv-/- mice, a model of hemochromatosis due to suppression of liver hepcidin, exhibit only modest cardiac Hamp deficiency and minor cardiac dysfunction. Dietary iron manipulations did not significantly affect cardiac Hamp mRNA in the atria of wild-type or Hjv-/- mice. Two weeks following myocardial infarction, Hamp was robustly induced in the liver and heart apex but not atria, possibly in response to inflammation. We conclude that cardiac Hamp is predominantly expressed in the right atrium and is partially regulated by Hjv; however, it does not respond to iron and other inducers of hepatic hepcidin.


Asunto(s)
Hemocromatosis , Hierro , Ratones , Animales , Hierro/metabolismo , Hepcidinas/genética , Hepcidinas/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Hemocromatosis/genética , Hemocromatosis/metabolismo , Proteína de la Hemocromatosis/genética , Proteína de la Hemocromatosis/metabolismo , Hígado/metabolismo , Atrios Cardíacos/metabolismo , Proteínas Ligadas a GPI/genética , Proteínas Ligadas a GPI/metabolismo
6.
Int J Mol Sci ; 24(5)2023 Feb 23.
Artículo en Inglés | MEDLINE | ID: mdl-36901833

RESUMEN

Although metabolic complications are common in thalassemia patients, there is still an unmet need to better understand underlying mechanisms. We used unbiased global proteomics to reveal molecular differences between the th3/+ mouse model of thalassemia and wild-type control animals focusing on skeletal muscles at 8 weeks of age. Our data point toward a significantly impaired mitochondrial oxidative phosphorylation. Furthermore, we observed a shift from oxidative fibre types toward more glycolytic fibre types in these animals, which was further supported by larger fibre-type cross-sectional areas in the more oxidative type fibres (type I/type IIa/type IIax hybrid). We also observed an increase in capillary density in th3/+ mice, indicative of a compensatory response. Western blotting for mitochondrial oxidative phosphorylation complex proteins and PCR analysis of mitochondrial genes indicated reduced mitochondrial content in the skeletal muscle but not the hearts of th3/+ mice. The phenotypic manifestation of these alterations was a small but significant reduction in glucose handling capacity. Overall, this study identified many important alterations in the proteome of th3/+ mice, amongst which mitochondrial defects leading to skeletal muscle remodelling and metabolic dysfunction were paramount.


Asunto(s)
Talasemia beta , Ratones , Animales , Talasemia beta/metabolismo , Proteómica , Músculo Esquelético/metabolismo , Mitocondrias/metabolismo , Oxidación-Reducción
7.
Blood ; 141(3): 271-284, 2023 01 19.
Artículo en Inglés | MEDLINE | ID: mdl-36351237

RESUMEN

Homeostatic adaptation to systemic iron overload involves transcriptional induction of bone morphogenetic protein 6 (BMP6) in liver sinusoidal endothelial cells (LSECs). BMP6 is then secreted to activate signaling of the iron hormone hepcidin (HAMP) in neighboring hepatocytes. To explore the mechanism of iron sensing by LSECs, we generated TfrcTek-Cre mice with endothelial cell-specific ablation of transferrin receptor 1 (Tfr1). We also used control Tfrcfl/fl mice to characterize the LSEC-specific molecular responses to iron using single-cell transcriptomics. TfrcTek-Cre animals tended to have modestly increased liver iron content (LIC) compared with Tfrcfl/fl controls but expressed physiological Bmp6 and Hamp messenger RNA (mRNA). Despite a transient inability to upregulate Bmp6, they eventually respond to iron challenges with Bmp6 and Hamp induction, yet occasionally to levels slightly lower relative to LIC. High dietary iron intake triggered the accumulation of serum nontransferrin bound iron (NTBI), which significantly correlated with liver Bmp6 and Hamp mRNA levels and elicited more profound alterations in the LSEC transcriptome than holo-transferrin injection. This culminated in the robust induction of Bmp6 and other nuclear factor erythroid 2-related factor 2 (Nrf2) target genes, as well as Myc target genes involved in ribosomal biogenesis and protein synthesis. LSECs and midzonal hepatocytes were the most responsive liver cells to iron challenges and exhibited the highest expression of Bmp6 and Hamp mRNAs, respectively. Our data suggest that during systemic iron overload, LSECs internalize NTBI, which promotes oxidative stress and thereby transcriptionally induces Bmp6 via Nrf2. Tfr1 appears to contribute to iron sensing by LSECs, mostly under low iron conditions.


Asunto(s)
Sobrecarga de Hierro , Hierro , Ratones , Animales , Hierro/metabolismo , Transferrina/metabolismo , Células Endoteliales/metabolismo , Proteína Morfogenética Ósea 6/genética , Proteína Morfogenética Ósea 6/metabolismo , Factor 2 Relacionado con NF-E2 , Hepatocitos/metabolismo , Hígado/metabolismo , Hepcidinas/genética , Hepcidinas/metabolismo , Sobrecarga de Hierro/genética , Sobrecarga de Hierro/metabolismo , ARN Mensajero/metabolismo
8.
Elife ; 112022 09 06.
Artículo en Inglés | MEDLINE | ID: mdl-36066082

RESUMEN

The iron hormone hepcidin is transcriptionally activated by iron or inflammation via distinct, partially overlapping pathways. We addressed how iron affects inflammatory hepcidin levels and the ensuing hypoferremic response. Dietary iron overload did not mitigate hepcidin induction in lipopolysaccharide (LPS)-treated wild type mice but prevented effective inflammatory hypoferremia. Likewise, LPS modestly decreased serum iron in hepcidin-deficient Hjv-/- mice, model of hemochromatosis. Synthetic hepcidin triggered hypoferremia in control but not iron-loaded wild type animals. Furthermore, it dramatically decreased hepatic and splenic ferroportin in Hjv-/- mice on standard or iron-deficient diet, but only triggered hypoferremia in the latter. Mechanistically, iron antagonized hepcidin responsiveness by inactivating IRPs in the liver and spleen to stimulate ferroportin mRNA translation. Prolonged LPS treatment eliminated ferroportin mRNA and permitted hepcidin-mediated hypoferremia in iron-loaded mice. Thus, de novo ferroportin synthesis is a critical determinant of serum iron and finetunes hepcidin-dependent functional outcomes. Our data uncover a crosstalk between hepcidin and IRE/IRP systems that controls tissue ferroportin expression and determines serum iron levels. Moreover, they suggest that hepcidin supplementation therapy is more efficient when combined with iron depletion.


Asunto(s)
Hepcidinas , Lipopolisacáridos , Animales , Proteínas de Transporte de Catión , Hepcidinas/genética , Hepcidinas/metabolismo , Hormonas , Lipopolisacáridos/farmacología , Ratones , ARN Mensajero/genética , Transducción de Señal/fisiología
9.
Int J Mol Sci ; 23(18)2022 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-36142654

RESUMEN

Iron regulatory protein 1 (IRP1) is a bifunctional protein with mutually exclusive RNA-binding or enzymatic activities that depend on the presence of a 4Fe-4S cluster. While IRP1 is a well-established cytosolic protein, work in a Drosophila model suggested that it may also exhibit nuclear localization. Herein, we addressed whether mammalian IRP1 can likewise translocate to the nucleus. We utilized primary cells and tissues from wild type and Irp1-/- mice, as well as human cell lines and tissue biopsy sections. IRP1 subcellular localization was analyzed by Western blotting, immunofluorescence and immunohistochemistry. We did not detect presence of nuclear IRP1 in wild type mouse embryonic fibroblasts (MEFs), primary hepatocytes or whole mouse liver. However, we observed IRP1-positive nuclei in human liver but not ovary sections. Biochemical fractionation studies revealed presence of IRP1 in the nucleus of human Huh7 and HepG2 hepatoma cells, but not HeLa cervical cancer cells. Importantly, nuclear IRP1 was only evident in iron-replete cells and disappeared following pharmacological iron chelation. These data provide the first experimental evidence for nuclear IRP1 expression in mammals, which appears to be species- and cell-specific. Furthermore, they suggest that the nuclear translocation of IRP1 is mediated by an iron-dependent mechanism.


Asunto(s)
Proteína 1 Reguladora de Hierro/metabolismo , Hierro , Animales , Fibroblastos/metabolismo , Humanos , Hierro/metabolismo , Quelantes del Hierro , Mamíferos/metabolismo , Ratones , ARN/metabolismo
10.
Proc Natl Acad Sci U S A ; 119(35): e2121251119, 2022 08 30.
Artículo en Inglés | MEDLINE | ID: mdl-35994670

RESUMEN

GCN2 (general control nonderepressible 2) is a serine/threonine-protein kinase that controls messenger RNA translation in response to amino acid availability and ribosome stalling. Here, we show that GCN2 controls erythrocyte clearance and iron recycling during stress. Our data highlight the importance of liver macrophages as the primary cell type mediating these effects. During different stress conditions, such as hemolysis, amino acid deficiency or hypoxia, GCN2 knockout (GCN2-/-) mice displayed resistance to anemia compared with wild-type (GCN2+/+) mice. GCN2-/- liver macrophages exhibited defective erythrophagocytosis and lysosome maturation. Molecular analysis of GCN2-/- cells demonstrated that the ATF4-NRF2 pathway is a critical downstream mediator of GCN2 in regulating red blood cell clearance and iron recycling.


Asunto(s)
Aminoácidos , Eritrocitos , Hierro , Hígado , Macrófagos , Proteínas Serina-Treonina Quinasas , Factor de Transcripción Activador 4/metabolismo , Aminoácidos/deficiencia , Aminoácidos/metabolismo , Anemia/metabolismo , Animales , Citofagocitosis , Eritrocitos/metabolismo , Eliminación de Gen , Hemólisis , Hipoxia/metabolismo , Hierro/metabolismo , Hígado/citología , Lisosomas/metabolismo , Macrófagos/metabolismo , Ratones , Ratones Noqueados , Factor 2 Relacionado con NF-E2/metabolismo , Proteínas Serina-Treonina Quinasas/deficiencia , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/metabolismo , Estrés Fisiológico
11.
Commun Biol ; 5(1): 153, 2022 02 22.
Artículo en Inglés | MEDLINE | ID: mdl-35194137

RESUMEN

Hemojuvelin (HJV) enhances signaling to the iron hormone hepcidin and its deficiency causes iron overload, a risk factor for hepatocellular carcinoma (HCC). We utilized Hjv-/- mice to dissect mechanisms for hepatocarcinogenesis. We show that suboptimal treatment with diethylnitrosamine (DEN) triggers HCC only in Hjv-/- but not wt mice. Liver proteomics data were obtained by mass spectrometry. Hierarchical clustering analysis revealed that Hjv deficiency and DEN elicit similar liver proteomic responses, including induction of mitochondrial proteins. Dietary iron overload of wt mice does not recapitulate the liver proteomic phenotype of Hjv-/- animals, which is only partially corrected by iron depletion. Consistent with these data, primary Hjv-/- hepatocytes exhibit mitochondrial hyperactivity, while aged Hjv-/- mice develop spontaneous HCC. Moreover, low expression of HJV or hepcidin (HAMP) mRNAs predicts poor prognosis in HCC patients. We conclude that Hjv has a hepatoprotective function and its deficiency in mice promotes mitochondrial dysfunction and hepatocarcinogenesis.


Asunto(s)
Carcinoma Hepatocelular , Neoplasias Hepáticas , Anciano , Animales , Carcinoma Hepatocelular/genética , Humanos , Neoplasias Hepáticas/genética , Proteínas de la Membrana/metabolismo , Ratones , Mitocondrias/metabolismo , Proteómica
12.
Hepatol Commun ; 5(12): 2139-2150, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34558857

RESUMEN

Hepcidin is a liver-derived peptide hormone that limits iron egress from tissues to the bloodstream. It operates by binding to the iron exporter ferroportin, which blocks iron transport and tags ferroportin for degradation. Genetic hepcidin inactivation leads to hereditary hemochromatosis, a disease of iron overload. We used wild-type and Hjv-/- mice, a model of hemochromatosis, to examine the expression of ferroportin and other proteins of iron metabolism in hepcidin target tissues. The animals were previously subjected to dietary iron manipulations. In Hjv-/- mice, hepcidin messenger RNA correlated significantly with hepatic iron load (r = 0.8211, P < 0.001), but was substantially lower compared with wild-type controls. Duodenal ferroportin and divalent metal transporter 1 (DMT1), as well as splenic and hepatic ferroportin, were overexpressed in these animals. A high-iron diet (2% carbonyl iron) suppressed duodenal DMT1 levels in both wild-type and Hjv-/- mice; however, it did not affect duodenal ferroportin expression in Hjv-/- mice, and only reduced it in wild-type mice. In contrast, the high-iron diet decreased splenic ferroportin exclusively in Hjv-/- mice, whereas it induced hepatic ferroportin exclusively in wild-type mice. Conclusion: Our data show that dietary iron differentially affects ferroportin expression in mouse tissues and are consistent with hepcidin-dependent and hepcidin-independent mechanisms for ferroportin regulation. In the Hjv-/- mouse model of hemochromatosis, duodenal ferroportin remains unresponsive to iron but DMT1 is appropriately iron-regulated.


Asunto(s)
Proteínas de Transporte de Catión/metabolismo , Hemocromatosis/metabolismo , Hierro de la Dieta/farmacología , Hierro/metabolismo , Animales , Modelos Animales de Enfermedad , Duodeno/efectos de los fármacos , Hepcidinas/metabolismo , Hígado/efectos de los fármacos , Ratones , Bazo/efectos de los fármacos
13.
Int J Mol Sci ; 21(20)2020 Oct 21.
Artículo en Inglés | MEDLINE | ID: mdl-33096618

RESUMEN

Iron is essential for energy metabolism, and states of iron deficiency or excess are detrimental for organisms and cells. Therefore, iron and carbohydrate metabolism are tightly regulated. Serum iron and glucose levels are subjected to hormonal regulation by hepcidin and insulin, respectively. Hepcidin is a liver-derived peptide hormone that inactivates the iron exporter ferroportin in target cells, thereby limiting iron efflux to the bloodstream. Insulin is a protein hormone secreted from pancreatic ß-cells that stimulates glucose uptake and metabolism via insulin receptor signaling. There is increasing evidence that systemic, but also cellular iron and glucose metabolic pathways are interconnected. This review article presents relevant data derived primarily from mouse models and biochemical studies. In addition, it discusses iron and glucose metabolism in the context of human disease.


Asunto(s)
Glucosa/metabolismo , Hierro/metabolismo , Síndrome Metabólico/metabolismo , Animales , Glucemia/metabolismo , Metabolismo Energético , Proteínas Facilitadoras del Transporte de la Glucosa/metabolismo , Humanos , Proteína 1 Reguladora de Hierro/metabolismo , Proteína 2 Reguladora de Hierro/metabolismo , Metabolómica , Ratones
14.
PLoS One ; 14(8): e0221455, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31442254

RESUMEN

Hepatic iron overload, a hallmark of hereditary hemochromatosis, triggers progressive liver disease. There is also increasing evidence for a pathogenic role of iron in non-alcoholic fatty liver disease (NAFLD), which may progress to non-alcoholic steatohepatitis (NASH), fibrosis, cirrhosis and hepatocellular cancer. Mouse models of hereditary hemochromatosis and NAFLD can be used to explore potential interactions between iron and lipid metabolic pathways. Hfe-/- mice, a model of moderate iron overload, were reported to develop early liver fibrosis in response to a high fat diet. However, this was not the case with Hjv-/- mice, a model of severe iron overload. These data raised the possibility that the Hfe gene may protect against liver injury independently of its iron regulatory function. Herein, we addressed this hypothesis in a comparative study utilizing wild type, Hfe-/-, Hjv-/- and double Hfe-/-Hjv-/- mice. The animals, all in C57BL/6J background, were fed with high fat diets for 14 weeks and developed hepatic steatosis, associated with iron overload. Hfe co-ablation did not sensitize steatotic Hjv-deficient mice to liver injury. Moreover, we did not observe any signs of liver inflammation or fibrosis even in single steatotic Hfe-/- mice. Ultrastructural studies revealed a reduced lipid and glycogen content in Hjv-/- hepatocytes, indicative of a metabolic defect. Interestingly, glycogen levels were restored in double Hfe-/-Hjv-/- mice, which is consistent with a metabolic function of Hfe. We conclude that hepatocellular iron excess does not aggravate diet-induced steatosis to steatohepatitis or early liver fibrosis in mouse models of hereditary hemochromatosis, irrespectively of the presence or lack of Hfe.


Asunto(s)
Dieta Alta en Grasa/efectos adversos , Hemocromatosis/complicaciones , Cirrosis Hepática/complicaciones , Animales , Peso Corporal , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Hígado Graso/complicaciones , Hígado Graso/patología , Hemocromatosis/patología , Hepatocitos/metabolismo , Hepatocitos/ultraestructura , Inflamación/patología , Hierro/sangre , Hígado/metabolismo , Hígado/patología , Hígado/ultraestructura , Cirrosis Hepática/patología , Ratones Endogámicos C57BL
15.
PLoS One ; 14(7): e0219835, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31295319

RESUMEN

Hepcidin is an iron regulatory peptide hormone that is secreted from hepatocytes and inhibits iron efflux from tissues to plasma. Under inflammatory conditions, hepcidin is transcriptionally induced by IL-6/STAT3 signaling and promotes hypoferremia, an innate immune response to infection. If this pathway remains unresolved, chronic overexpression of hepcidin contributes to the anemia of inflammation, a common medical condition. Previous work showed that carbon monoxide (CO) releasing drugs (CORMs) can attenuate inflammatory induction of hepcidin. Because CO is physiologically generated during heme degradation by heme oxygenase 1 (HO-1), an IL-6-inducible enzyme with anti-inflammatory properties, we hypothesized that hepatocellular HO-1 may operate as a physiological feedback regulator of hepcidin that resolves inflammatory signaling. To address this, we generated and analyzed hepatocyte-specific HO-1 knockout (Hmox1Alb-Cre) mice. We show that these animals mount appropriate hepcidin-mediated hypoferremic response to LPS-induced inflammation, with kinetics similar to those of control Hmox1fl/fl mice. Likewise, primary hepatocytes from Hmox1Alb-Cre and Hmox1fl/fl mice exhibit similar degree and kinetics of hepcidin induction following IL-6 treatment. We conclude that hepatocellular HO-1 has no physiological function on hepcidin regulation by the inflammatory pathway.


Asunto(s)
Hemo-Oxigenasa 1/deficiencia , Hepatocitos/metabolismo , Hepcidinas/metabolismo , Mediadores de Inflamación/metabolismo , Animales , Biomarcadores , Hemo-Oxigenasa 1/genética , Hemo-Oxigenasa 1/metabolismo , Hierro/metabolismo , Hígado/metabolismo , Hígado/patología , Masculino , Ratones , Ratones Noqueados
16.
Blood ; 133(4): 344-355, 2019 01 24.
Artículo en Inglés | MEDLINE | ID: mdl-30538134

RESUMEN

Transferrin receptor 1 (Tfr1) mediates uptake of circulating transferrin-bound iron to developing erythroid cells and other cell types. Its critical physiological function is highlighted by the embryonic lethal phenotype of Tfr1-knockout (Tfrc-/-) mice and the pathologies of several tissue-specific knockouts. We generated TfrcAlb-Cre mice bearing hepatocyte-specific ablation of Tfr1 to explore implications in hepatocellular and systemic iron homeostasis. TfrcAlb-Cre mice are viable and do not display any apparent liver pathology. Nevertheless, their liver iron content (LIC) is lower compared with that of control Tfrcfl/fl littermates as a result of the reduced capacity of Tfr1-deficient hepatocytes to internalize iron from transferrin. Even though liver Hamp messenger RNA (mRNA) and serum hepcidin levels do not differ between TfrcAlb-Cre and Tfrcfl/fl mice, Hamp/LIC and hepcidin/LIC ratios are significantly higher in the former. Importantly, this is accompanied by modest hypoferremia and microcytosis, and it predisposes TfrcAlb-Cre mice to iron-deficiency anemia. TfrcAlb-Cre mice appropriately regulate Hamp expression following dietary iron manipulations or holo-transferrin injection. Holo-transferrin also triggers proper induction of Hamp mRNA, ferritin, and Tfr2 in primary TfrcAlb-Cre hepatocytes. We further show that these cells can acquire 59Fe from 59Fe-transferrin, presumably via Tfr2. We conclude that Tfr1 is redundant for basal hepatocellular iron supply but essential for fine-tuning hepcidin responses according to the iron load of hepatocytes. Our data are consistent with an inhibitory function of Tfr1 on iron signaling to hepcidin via its interaction with Hfe. Moreover, they highlight hepatocellular Tfr1 as a link between cellular and systemic iron-regulatory pathways.


Asunto(s)
Antígenos CD/metabolismo , Hepatocitos/metabolismo , Hepcidinas/metabolismo , Homeostasis , Hierro/metabolismo , Receptores de Transferrina/metabolismo , Anemia Ferropénica/patología , Animales , Ferritinas/metabolismo , Eliminación de Gen , Regulación de la Expresión Génica/efectos de los fármacos , Marcación de Gen , Hepatocitos/efectos de los fármacos , Hepcidinas/genética , Homeostasis/efectos de los fármacos , Integrasas/metabolismo , Hierro de la Dieta/farmacología , Ratones Endogámicos C57BL , Receptores de Transferrina/deficiencia , Transferrina/metabolismo
17.
Blood ; 132(17): 1829-1841, 2018 10 25.
Artículo en Inglés | MEDLINE | ID: mdl-30213871

RESUMEN

Systemic iron balance is controlled by hepcidin, a liver hormone that limits iron efflux to the bloodstream by promoting degradation of the iron exporter ferroportin in target cells. Iron-dependent hepcidin induction requires hemojuvelin (HJV), a bone morphogenetic protein (BMP) coreceptor that is disrupted in juvenile hemochromatosis, causing dramatic hepcidin deficiency and tissue iron overload. Hjv-/- mice recapitulate phenotypic hallmarks of hemochromatosis but exhibit blunted hepcidin induction following lipopolysaccharide (LPS) administration. We show that Hjv-/- mice fail to mount an appropriate hypoferremic response to acute inflammation caused by LPS, the lipopeptide FSL1, or Escherichia coli infection because residual hepcidin does not suffice to drastically decrease macrophage ferroportin levels. Hfe-/- mice, a model of milder hemochromatosis, exhibit almost wild-type inflammatory hepcidin expression and associated effects, whereas double Hjv-/-Hfe-/- mice phenocopy single Hjv-/- counterparts. In primary murine hepatocytes, Hjv deficiency does not affect interleukin-6 (IL-6)/Stat, and only slightly inhibits BMP2/Smad signaling to hepcidin; however, it severely impairs BMP6/Smad signaling and thereby abolishes synergism with the IL-6/Stat pathway. Inflammatory induction of hepcidin is suppressed in iron-deficient wild-type mice and recovers after the animals are provided overnight access to an iron-rich diet. We conclude that Hjv is required for inflammatory induction of hepcidin and controls the acute hypoferremic response by maintaining a threshold of Bmp6/Smad signaling. Our data highlight Hjv as a potential pharmacological target against anemia of inflammation.


Asunto(s)
Hepatocitos/metabolismo , Hepcidinas/metabolismo , Inflamación/metabolismo , Hierro/metabolismo , Proteínas de la Membrana/metabolismo , Animales , Proteína Morfogenética Ósea 6/metabolismo , Proteínas Ligadas a GPI , Proteína de la Hemocromatosis , Sobrecarga de Hierro/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Transducción de Señal/fisiología , Proteínas Smad/metabolismo
18.
Blood ; 131(2): 236-246, 2018 01 11.
Artículo en Inglés | MEDLINE | ID: mdl-29180398

RESUMEN

Thalassemias are a heterogeneous group of red blood cell disorders, considered a major cause of morbidity and mortality among genetic diseases. However, there is still no universally available cure for thalassemias. The underlying basis of thalassemia pathology is the premature apoptotic destruction of erythroblasts causing ineffective erythropoiesis. In ß-thalassemia, ß-globin synthesis is reduced causing α-globin accumulation. Unpaired globin chains, with heme attached to them, accumulate in thalassemic erythroblasts causing oxidative stress and the premature cell death. We hypothesize that in ß-thalassemia heme oxygenase (HO) 1 could play a pathogenic role in the development of anemia and ineffective erythropoiesis. To test this hypothesis, we exploited a mouse model of ß-thalassemia intermedia, Th3/+ We observed that HO inhibition using tin protoporphyrin IX (SnPP) decreased heme-iron recycling in the liver and ameliorated anemia in the Th3/+ mice. SnPP administration led to a decrease in erythropoietin and increase in hepcidin serum levels, changes that were accompanied by an alleviation of ineffective erythropoiesis in Th3/+ mice. Additionally, the bone marrow from Th3/+ mice treated with SnPP exhibited decreased heme catabolism and diminished iron release as well as reduced apoptosis. Our results indicate that the iron released from heme because of HO activity contributes to the pathophysiology of thalassemia. Therefore, new therapies that suppress heme catabolism may be beneficial in ameliorating the anemia and ineffective erythropoiesis in thalassemias.


Asunto(s)
Inhibidores Enzimáticos/uso terapéutico , Hemo-Oxigenasa 1/antagonistas & inhibidores , Sobrecarga de Hierro/tratamiento farmacológico , Metaloporfirinas/uso terapéutico , Protoporfirinas/uso terapéutico , Talasemia beta/tratamiento farmacológico , Animales , Modelos Animales de Enfermedad , Eritropoyesis/efectos de los fármacos , Eritropoyetina/sangre , Hemo-Oxigenasa 1/análisis , Sobrecarga de Hierro/sangre , Sobrecarga de Hierro/complicaciones , Sobrecarga de Hierro/patología , Hígado/efectos de los fármacos , Hígado/patología , Ratones , Ratones Endogámicos C57BL , Talasemia beta/sangre , Talasemia beta/complicaciones , Talasemia beta/patología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...