Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
1.
Hepatology ; 65(5): 1512-1525, 2017 05.
Artículo en Inglés | MEDLINE | ID: mdl-28027576

RESUMEN

Ferroportin (FPN1) is the sole iron exporter in mammals, but its cell-specific function and regulation are still elusive. This study examined FPN1 expression in human macrophages, the cells that are primarily responsible on a daily basis for plasma iron turnover and are central in the pathogenesis of ferroportin disease (FD), the disease attributed to lack-of-function FPN1 mutations. We characterized FPN1 protein expression and traffic by confocal microscopy, western blotting, gel filtration, and immunoprecipitation studies in macrophages from control blood donors (donor) and patients with either FPN1 p.A77D, p.G80S, and p.Val162del lack-of-function or p.A69T gain-of-function mutations. We found that in normal macrophages, FPN1 cycles in the early endocytic compartment does not multimerize and is promptly degraded by hepcidin (Hepc), its physiological inhibitor, within 3-6 hours. In FD macrophages, endogenous FPN1 showed a similar localization, except for greater accumulation in lysosomes. However, in contrast with previous studies using overexpressed mutant protein in cell lines, FPN1 could still reach the cell surface and be normally internalized and degraded upon exposure to Hepc. However, when FD macrophages were exposed to large amounts of heme iron, in contrast to donor and p.A69T macrophages, FPN1 could no longer reach the cell surface, leading to intracellular iron retention. CONCLUSION: FPN1 cycles as a monomer within the endocytic/plasma membrane compartment and responds to its physiological inhibitor, Hepc, in both control and FD cells. However, in FD, FPN1 fails to reach the cell surface when cells undergo high iron turnover. Our findings provide a basis for the FD characterized by a preserved iron transfer in the enterocytes (i.e., cells with low iron turnover) and iron retention in cells exposed to high iron flux, such as liver and spleen macrophages. (Hepatology 2017;65:1512-1525).


Asunto(s)
Proteínas de Transporte de Catión/deficiencia , Macrófagos/metabolismo , Animales , Estudios de Casos y Controles , Células Hep G2 , Hepcidinas/metabolismo , Humanos , Hierro/metabolismo , Ratones
2.
Gastroenterology ; 146(4): 1060-9, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24361124

RESUMEN

BACKGROUND & AIMS: Hepatic gluconeogenesis provides fuel during starvation, and is abnormally induced in obese individuals or those with diabetes. Common metabolic disorders associated with active gluconeogenesis and insulin resistance (obesity, metabolic syndrome, diabetes, and nonalcoholic fatty liver disease) have been associated with alterations in iron homeostasis that disrupt insulin sensitivity and promote disease progression. We investigated whether gluconeogenic signals directly control Hepcidin, an important regulator of iron homeostasis, in starving mice (a model of persistently activated gluconeogenesis and insulin resistance). METHODS: We investigated hepatic regulation of Hepcidin expression in C57BL/6Crl, 129S2/SvPas, BALB/c, and Creb3l3-/- null mice. Mice were fed a standard, iron-balanced chow diet or an iron-deficient diet for 9 days before death, or for 7 days before a 24- to 48-hour starvation period; liver and spleen tissues then were collected and analyzed by quantitative reverse-transcription polymerase chain reaction and immunoblot analyses. Serum levels of iron, hemoglobin, Hepcidin, and glucose also were measured. We analyzed human hepatoma (HepG2) cells and mouse primary hepatocytes to study transcriptional control of Hamp (the gene that encodes Hepcidin) in response to gluconeogenic stimuli using small interfering RNA, luciferase promoter, and chromatin immunoprecipitation analyses. RESULTS: Starvation led to increased transcription of the gene that encodes phosphoenolpyruvate carboxykinase 1 (a protein involved in gluconeogenesis) in livers of mice, increased levels of Hepcidin, and degradation of Ferroportin, compared with nonstarved mice. These changes resulted in hypoferremia and iron retention in liver tissue. Livers of starved mice also had increased levels of Ppargc1a mRNA and Creb3l3 mRNA, which encode a transcriptional co-activator involved in energy metabolism and a liver-specific transcription factor, respectively. Glucagon and a cyclic adenosine monophosphate analog increased promoter activity and transcription of Hamp in cultured liver cells; levels of Hamp were reduced after administration of small interfering RNAs against Ppargc1a and Creb3l3. PPARGC1A and CREB3L3 bound the Hamp promoter to activate its transcription in response to a cyclic adenosine monophosphate analog. Creb3l3-/- mice did not up-regulate Hamp or become hypoferremic during starvation. CONCLUSIONS: We identified a link between glucose and iron homeostasis, showing that Hepcidin is a gluconeogenic sensor in mice during starvation. This response is involved in hepatic metabolic adaptation to increased energy demands; it preserves tissue iron for vital activities during food withdrawal, but can cause excessive iron retention and hypoferremia in disorders with persistently activated gluconeogenesis and insulin resistance.


Asunto(s)
Gluconeogénesis , Hepatocitos/metabolismo , Hepcidinas/sangre , Hierro/sangre , Hígado/metabolismo , Transducción de Señal , Inanición/sangre , Animales , Sitios de Unión , Glucemia/metabolismo , Proteínas de Transporte de Catión/metabolismo , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/deficiencia , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/genética , Modelos Animales de Enfermedad , Hemoglobinas/metabolismo , Células Hep G2 , Homeostasis , Humanos , Resistencia a la Insulina , Masculino , Ratones , Ratones de la Cepa 129 , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma , Fosfoenolpiruvato Carboxiquinasa (GTP)/genética , Fosfoenolpiruvato Carboxiquinasa (GTP)/metabolismo , Regiones Promotoras Genéticas , Interferencia de ARN , Bazo/metabolismo , Inanición/genética , Factores de Tiempo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Transcripción Genética , Transfección , Regulación hacia Arriba
3.
J Mol Med (Berl) ; 101(9): 1113-1124, 2023 09.
Artículo en Inglés | MEDLINE | ID: mdl-37493829

RESUMEN

Hepcidin, the hepatic iron hormone, is the central regulator of iron homeostasis. Cyclic AMP-Responsive Element-Binding protein 3-like 3 (CREB3L3/CREB-H) is a liver homeostatic regulator of essential nutrients (i.e. glucose and lipids) and has been previously involved in hepcidin response to pathologic stress signals. Here, we asked whether CREB-H has also a physiologic role in iron homeostasis through hepcidin. To this end, we analyzed hepcidin gene expression and regulation in the liver of wild type and Creb3l3 knockout mice during early postnatal development, as a model of "physiologic" stressful condition. The effect of iron challenge in vivo and BMP6 stimulation in vitro have been also addressed. In addition, we investigated the BMP signaling pathway and hepcidin promoter activity following CREB3L3 silencing and hepcidin promoter mutation in HepG2 cells. Creb3l3 knockout suckling and young-adult mice showed a prominent serum and hepatic iron accumulation, respectively, due to impaired hepcidin mRNA expression which progressively returned to normal level in adult mice. Interestingly, upon iron challenge, while the upstream BMP/SMAD signaling pathway controlling hepcidin was equally responsive in both strains, hepcidin gene expression was impaired in knockout mice and more iron accumulated in the liver. Accordingly, hepcidin gene response to BMP6 was blunted in primary CREB-H knockout hepatocytes and in HepG2 cells transfected with CREB-H siRNA or carrying a hepcidin promoter mutated in the CREB-H binding site. In conclusion, CREB-H has a role in maintaining the homeostatic balance of iron traffic through hepcidin during the critical postnatal period and in response to iron challenge. KEY MESSAGES: CREB-H KO mice develop liver iron overload shortly after weaning that normalizes in adulthood. CHEB-H is involved in hepcidin gene response to oral iron in vivo. CREB-H loss hampers hepcidin promoter response to BMP6. CREB-H is a key stress-sensor controlling hepcidin gene transcription in physiologic and pathophysiologic states.


Asunto(s)
Hepcidinas , Hígado , Ratones , Animales , Hígado/metabolismo , Hierro/metabolismo , Proteína Morfogenética Ósea 6/genética , Proteína Morfogenética Ósea 6/metabolismo , Ratones Noqueados , Expresión Génica , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/metabolismo
4.
Gastroenterology ; 139(1): 315-22.e1, 2010 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-20338170

RESUMEN

BACKGROUND & AIMS: Hemochromatosis is a common hereditary disease caused by mutations in HFE and characterized by increased absorption of iron in the intestine. However, the intestine does not appear to be the site of mutant HFE activity in the disease; we investigated the role of the liver-the source of the iron regulatory hormone hepcidin-in pathogenesis in mice. METHODS: We exchanged livers between Hfe wild-type (+/+) and Hfe null (-/-) mice by orthotopic liver transplantation (OLT) and assessed histopathology, serum and tissue iron parameters, and hepatic hepcidin messenger RNA expression. RESULTS: At 6-8 months after OLT, Hfe(-/-) mice that received Hfe(-/-) livers maintained the hemochromatosis phenotype: iron accumulation in hepatocytes but not Kupffer cells (KC), increased transferrin levels, and low levels of iron in the spleen. Hfe(+/+) mice that received Hfe(-/-) livers had increased levels of iron in serum and liver and low levels of iron in spleen. However, they did not develop the iron-poor KCs that characterize hemochromatosis: KCs appeared iron rich, although hepatic hepcidin expression was low. Transplantation of Hfe(+/+) livers into Hfe(-/-) mice prevented hepatic iron accumulation but did not return spleen and plasma levels of iron to normal; KCs still appeared to be iron poor, despite normal hepcidin expression. CONCLUSIONS: In Hfe(-/-) mice, transplantation of livers from Hfe(+/+) mice reversed the iron-loading phenotype associated with hemochromatosis (regardless of Hfe expression in intestine). However, KCs still had low levels of iron that were not affected by hepatic hepcidin expression. These findings indicate an independent, iron-modifying effect of HFE in KCs.


Asunto(s)
Péptidos Catiónicos Antimicrobianos/fisiología , Antígenos de Histocompatibilidad Clase I/fisiología , Hierro/metabolismo , Macrófagos del Hígado/fisiología , Trasplante de Hígado , Proteínas de la Membrana/fisiología , Animales , Proteína de la Hemocromatosis , Hepcidinas , Hígado/metabolismo , Macrófagos/fisiología , Masculino , Ratones , Fenotipo
5.
Gastroenterology ; 139(5): 1721-9, 2010 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-20682319

RESUMEN

BACKGROUND & AIMS: Abnormal hepcidin regulation is central to the pathogenesis of HFE hemochromatosis. Hepatic bone morphogenetic protein 6 (BMP6)-SMAD signaling is a main regulatory mechanism controlling hepcidin expression, and this pathway was recently shown to be impaired in Hfe knockout (Hfe(-/-)) mice. To more definitively determine whether HFE regulates hepcidin expression through an interaction with the BMP6-SMAD signaling pathway, we investigated whether hepatic Hfe overexpression activates the BMP6-SMAD pathway to induce hepcidin expression. We then investigated whether excess exogenous BMP6 administration overcomes the BMP6-SMAD signaling impairment and ameliorates hemochromatosis in Hfe(-/-) mice. METHODS: The BMP6-SMAD pathway and the effects of neutralizing BMP6 antibody were examined in Hfe transgenic mice (Hfe Tg) compared with wild-type (WT) mice. Hfe(-/-) and WT mice were treated with exogenous BMP6 and analyzed for hepcidin expression and iron parameters. RESULTS: Hfe Tg mice exhibited hepcidin excess and iron deficiency anemia. Hfe Tg mice also exhibited increased hepatic BMP6-SMAD target gene expression compared with WT mice, whereas anti-BMP6 antibody administration to Hfe Tg mice improved the hepcidin excess and iron deficiency. In Hfe(-/-) mice, supraphysiologic doses of exogenous BMP6 improved hepcidin deficiency, reduced serum iron, and redistributed tissue iron to appropriate storage sites. CONCLUSIONS: HFE interacts with the BMP6-SMAD signaling pathway to regulate hepcidin expression, but HFE is not necessary for hepcidin induction by BMP6. Exogenous BMP6 treatment in mice compensates for the molecular defect underlying Hfe hemochromatosis, and BMP6-like agonists may have a role as an alternative therapeutic strategy for this disease.


Asunto(s)
Péptidos Catiónicos Antimicrobianos/genética , Proteína Morfogenética Ósea 6/uso terapéutico , Regulación de la Expresión Génica , Hemocromatosis/prevención & control , Antígenos de Histocompatibilidad Clase I/metabolismo , Hierro/metabolismo , Proteínas de la Membrana/metabolismo , ARN/genética , Animales , Péptidos Catiónicos Antimicrobianos/biosíntesis , Péptidos Catiónicos Antimicrobianos/efectos de los fármacos , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Femenino , Hemocromatosis/metabolismo , Hemocromatosis/patología , Proteína de la Hemocromatosis , Hepcidinas , Masculino , Ratones , Ratones Noqueados , Reacción en Cadena de la Polimerasa
6.
Hepatology ; 51(2): 654-9, 2010 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-20017200

RESUMEN

UNLABELLED: Hereditary hemochromatosis (HC) is commonly associated with homozygosity for the cysteine-to-tyrosine substitution at position 282 (C282Y) of the HFE protein. This mutation prevents HFE from binding beta(2)-microglobulin (beta(2)M) and reaching the cell surface. We have discovered that a widely used hepatoma cell line, Huh-7, carries a HFE mutation similar to that associated with human HC. By HFE gene sequencing of Huh-7 genomic DNA, we found a TAC nucleotide deletion (c. 691_693del) responsible for loss of a tyrosine at position 231 (p. Y231del) of the HFE protein. This mutation affects a conserved hydrophobic region in a loop connecting two beta strands that make up the alpha3 domain of HFE, not far from the 282 site. HFE was detected by western blot in HepG2 but not in Huh-7 cell membrane fractions. In WRL-68 cells expressing wild-type HFE, the HFE protein was largely found at the plasma membrane where it colocalizes with beta(2)M. On the contrary, the HFE-Y231del mutant, similarly to an exogenously expressed HFE-C282Y mutant, failed to reach the plasma membrane and did not colocalize with membrane-expressed beta(2)M. C282Y mutant HFE in HC is associated with inadequate hepcidin expression. We found that Huh-7 cells display lower hepcidin messenger RNA levels as compared to HepG2 cells, which carry a wild-type HFE. Interestingly, hepcidin messenger RNA levels increased significantly in Huh-7 cells stably expressing exogenous wild-type HFE at the plasma membrane. CONCLUSION: Huh-7 cells may represent a novel and valuable tool to investigate the role of altered HFE traffic in iron metabolism and pathogenesis of human HFE HC.


Asunto(s)
Línea Celular Tumoral , Antígenos de Histocompatibilidad Clase I/genética , Proteínas de la Membrana/genética , Mutación , Proteína de la Hemocromatosis , Humanos
7.
Gastroenterology ; 137(4): 1489-97, 2009 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-19591830

RESUMEN

BACKGROUND AND AIMS: Mutations in HFE are the most common cause of the iron-overload disorder hereditary hemochromatosis. Levels of the main iron regulatory hormone, hepcidin, are inappropriately low in hereditary hemochromatosis mouse models and patients with HFE mutations, indicating that HFE regulates hepcidin. The bone morphogenetic protein 6 (BMP6)-SMAD signaling pathway is an important endogenous regulator of hepcidin expression. We investigated whether HFE is involved in BMP6-SMAD regulation of hepcidin expression. METHODS: The BMP6-SMAD pathway was examined in Hfe knockout (KO) mice and in wild-type (WT) mice as controls. Mice were placed on diets of varying iron content. Hepcidin induction by BMP6 was examined in primary hepatocytes from Hfe KO mice; data were compared with those of WT mice. RESULTS: Liver levels of Bmp6 messenger RNA (mRNA) were higher in Hfe KO mice; these were appropriate for the increased hepatic levels of iron in these mice, compared with WT mice. However, levels of hepatic phosphorylated Smad 1/5/8 protein (an intracellular mediator of Bmp6 signaling) and Id1 mRNA (a target gene of Bmp6) were inappropriately low for the body iron burden and Bmp6 mRNA levels in Hfe KO, compared with WT mice. BMP6 induction of hepcidin expression was reduced in Hfe KO hepatocytes compared with WT hepatocytes. CONCLUSIONS: HFE is not involved in regulation of BMP6 by iron, but does regulate the downstream signals of BMP6 that are triggered by iron.


Asunto(s)
Proteína Morfogenética Ósea 6/metabolismo , Hemocromatosis/metabolismo , Hígado/metabolismo , Proteínas de la Membrana/deficiencia , Transducción de Señal , Animales , Péptidos Catiónicos Antimicrobianos/metabolismo , Proteína Morfogenética Ósea 6/genética , Células Cultivadas , Modelos Animales de Enfermedad , Femenino , Hemocromatosis/genética , Proteína de la Hemocromatosis , Hepcidinas , Antígenos de Histocompatibilidad Clase I/genética , Proteína 1 Inhibidora de la Diferenciación/metabolismo , Hierro de la Dieta/metabolismo , Masculino , Proteínas de la Membrana/genética , Ratones , Ratones Noqueados , Fosforilación , ARN Mensajero/metabolismo , Transducción de Señal/genética , Proteína Smad1/metabolismo , Proteína Smad5/metabolismo , Proteína Smad8/metabolismo
8.
Mol Cell Endocrinol ; 513: 110817, 2020 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-32439416

RESUMEN

The Osteocyte, recognized as a major orchestrator of osteoblast and osteoclast activity, is the most important key player during bone remodeling processes. Imbalances occurring during bone remodeling, caused by hormone perturbations or by mechanical loading alterations, can induce bone pathologies such as osteoporosis. Recently, the active fraction of parathormone, PTH (1-34) or Teriparatide (TPTD), was chosen as election treatment for osteoporosis. The effect of such therapy is dependent on the temporal manner of administration. The molecular reasons why the type of administration regimen is so critical for the fate of bone remodeling are numerous and not yet well known. Our study attempts to analyze diverse signaling pathways directly activated in osteocytes upon TPTD treatment. By means of gene array analysis, we found many molecules upregulated or downregulated in osteocytes. Later, we paid attention to Wisp-2, a protein involved in the Wnt pathway, that is secreted by MLO-Y4 cells and increases upon TPTD treatment and that is able to positively influence the early phases of osteogenic differentiation. We also confirmed the pro osteogenic property of Wisp-2 during mesenchymal stem cell differentiation into the preliminary osteoblast phenotype. The same results were confirmed with an in vivo approach confirming a remarkable Wisp-2 expression in metaphyseal trabecular bone. These results highlighted the anabolic roles unrolled by osteocytes in controlling the action of neighboring cells, suggesting that the perturbation of certain signaling cascades, such as the Wnt pathway, is crucial for the positive regulation of bone formation.


Asunto(s)
Péptidos y Proteínas de Señalización Intracelular/genética , Osteoblastos/efectos de los fármacos , Teriparatido/farmacología , Animales , Remodelación Ósea/efectos de los fármacos , Diferenciación Celular/efectos de los fármacos , Células Cultivadas , Regulación de la Expresión Génica/efectos de los fármacos , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Masculino , Ratones , Osteoblastos/fisiología , Osteocitos/efectos de los fármacos , Osteocitos/fisiología , Osteogénesis/efectos de los fármacos , Osteogénesis/genética , Ratas , Ratas Sprague-Dawley
9.
Endocrinology ; 157(10): 3935-3945, 2016 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-27483343

RESUMEN

Hepcidin, the iron hormone, is regulated by a number of stimulatory and inhibitory signals. The cAMP responsive element binding protein 3-like 3 (CREB3L3) mediates hepcidin response to endoplasmic reticulum (ER) stress. In this study we asked whether hepcidin response to ER stress also requires the small mother against decapentaplegic (SMAD)-1/5/8 pathway, which has a major role in hepcidin regulation in response to iron and other stimuli. We analyzed hepcidin mRNA expression and promoter activity in response to ER stressors in HepG2 cells in the presence of the bone morphogenetic protein (BMP) type I receptor inhibitor LDN-193189, mutated hepcidin promoter or small interfering RNA against different SMAD proteins. We then used a similar approach in vivo in wild-type, Smad1/5, or Creb3l3-/- animals undergoing ER stress. In vitro, LDN-193189 prevented hepcidin mRNA induction by different ER stressors. Seemingly, mutation of a BMP-responsive element in the hepcidin promoter prevented ER stress-mediated up-regulation. Moreover, in vitro silencing of SMAD proteins by small interfering RNA, in particular SMAD5, blunted hepcidin response to ER stress. On the contrary, hepcidin induction by ER stress was maintained when using antibodies against canonical BMP receptor ligands. In vivo, hepcidin was induced by ER stress and prevented by LDN-193189. In addition, in Smad1/5 knockout mice, ER stress was unable to induce hepcidin expression. Finally, in Creb3l3 knockout mice, in response to ER stress, SMAD1/5 were correctly phosphorylated and hepcidin induction was still appreciable, although to a lesser extent as compared with the control mice. In conclusion, our study indicates that hepcidin induction by ER stress involves the central regulatory SMAD1/5 pathway.


Asunto(s)
Estrés del Retículo Endoplásmico , Hepcidinas/metabolismo , Proteínas Smad Reguladas por Receptores/metabolismo , Animales , Células Hep G2 , Humanos , Masculino , Ratones Endogámicos C57BL , Transducción de Señal
10.
Science ; 325(5942): 877-80, 2009 Aug 14.
Artículo en Inglés | MEDLINE | ID: mdl-19679815

RESUMEN

Hepcidin is a peptide hormone that is secreted by the liver and controls body iron homeostasis. Hepcidin overproduction causes anemia of inflammation, whereas its deficiency leads to hemochromatosis. Inflammation and iron are known extracellular stimuli for hepcidin expression. We found that endoplasmic reticulum (ER) stress also induces hepcidin expression and causes hypoferremia and spleen iron sequestration in mice. CREBH (cyclic AMP response element-binding protein H), an ER stress-activated transcription factor, binds to and transactivates the hepcidin promoter. Hepcidin induction in response to exogenously administered toxins or accumulation of unfolded protein in the ER is defective in CREBH knockout mice, indicating a role for CREBH in ER stress-regulated hepcidin expression. The regulation of hepcidin by ER stress links the intracellular response involved in protein quality control to innate immunity and iron homeostasis.


Asunto(s)
Péptidos Catiónicos Antimicrobianos/genética , Péptidos Catiónicos Antimicrobianos/metabolismo , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/metabolismo , Retículo Endoplásmico/fisiología , Hierro/metabolismo , Estrés Fisiológico , Células 3T3 , Animales , Línea Celular Tumoral , Hepcidinas , Homeostasis , Humanos , Inmunidad Innata , Hierro/sangre , Hígado/metabolismo , Ratones , Ratones Noqueados , Mutación , Regiones Promotoras Genéticas , Pliegue de Proteína , Interferencia de ARN , Bazo/metabolismo , Activación Transcripcional
11.
Blood Cells Mol Dis ; 35(3): 315-8, 2005.
Artículo en Inglés | MEDLINE | ID: mdl-16135412

RESUMEN

Ferroportin-associated iron overload (also known as the ferroportin disease) is a common cause of hereditary hyperferritinemia. It was originally proposed that loss-of-protein function mutations account for iron overload in the FD. This hypothesis is consistent with the phenotype reported in most patients with FD of early iron accumulation in tissues, particularly in macrophages, in spite of relatively normal-low circulatory iron. It was still unclear, however, how FPN mutations would affect iron retention in enterocytes. We studied histologically the intestine of six patients with different FPN mutations as compared to other subjects with various iron disorders. We found that regardless of the underlying FPN mutation, no iron accumulation was found in absorbing enterocytes while, intestinal villi showed marked signs of iron accumulation in the cells of lamina propria. Not surprisingly, in the liver, iron excess was found mainly in Kupffer cells. These results indicate that FPN haploinsufficiency is not limiting for iron export from enterocytes.


Asunto(s)
Proteínas de Transporte de Catión/genética , Enterocitos/metabolismo , Sobrecarga de Hierro/genética , Sobrecarga de Hierro/metabolismo , Hierro/metabolismo , Adulto , Anciano , Enterocitos/patología , Femenino , Humanos , Sobrecarga de Hierro/patología , Macrófagos del Hígado/metabolismo , Masculino , Persona de Mediana Edad , Mutación
12.
Hepatology ; 41(3): 545-52, 2005 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-15726660

RESUMEN

Hepcidin, the iron hormone, is produced by the liver in response to iron and inflammation. Its synthesis during inflammation is triggered by cytokines, but the details of iron activation are obscure. We tested the role of Kupffer cells and macrophages by studying iron-loaded or inflamed mice with selective inactivation of Kupffer cells or the in vitro effect of conditioned human macrophages on hepcidin expression. Hepcidin messenger RNA (mRNA) expression was studied by Northern blot and reverse transcriptase polymerase chain reaction analysis in mice that were treated with 40 mg/kg gadolinium (III) chloride (GdCl(3)) as a Kupffer cell inactivating agent and subjected to inflammatory challenges with either lipopolysaccharide (LPS) and turpentine or iron overload by iron-dextran administration. Similar analyses were performed in human hepatoma cells (HepG2) cultured with medium from LPS- or iron-conditioned macrophages from blood donors or patients with HFE-linked hereditary hemochromatosis (HH). In vivo, LPS and particularly turpentine stimulated hepcidin mRNA expression, and this effect was prevented by the inactivation of Kupffer cells. Also, iron overload markedly upregulated hepatic hepcidin mRNA, but this activity persisted in spite of Kupffer cell blockade. In vitro, the medium of LPS-treated normal or hemocromatotic macrophages turned on hepcidin expression. On the contrary, medium of iron-manipulated macrophages, regardless of their HFE status, did not affect hepcidin mRNA steady-state levels. In conclusion, Kupffer cells are required for the activation of hepcidin synthesis during inflammation, and HH inflamed macrophages are capable of mounting a normal response, eventually leading to hepcidin stimulation. However, both Kupffer cells and human macrophages are dispensable for the regulatory activity exerted by iron on hepatic hepcidin.


Asunto(s)
Péptidos Catiónicos Antimicrobianos/genética , Sobrecarga de Hierro/metabolismo , Macrófagos del Hígado/fisiología , Hígado/metabolismo , Macrófagos/fisiología , Adulto , Animales , Células Cultivadas , Femenino , Gadolinio/farmacología , Proteína de la Hemocromatosis , Hepcidinas , Antígenos de Histocompatibilidad Clase I/fisiología , Humanos , Lipopolisacáridos/farmacología , Masculino , Proteínas de la Membrana/fisiología , Ratones , Ratones Endogámicos BALB C , Persona de Mediana Edad , ARN Mensajero/análisis
13.
J Hepatol ; 39(5): 710-5, 2003 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-14568251

RESUMEN

BACKGROUND/AIMS: MTP1/Ferroportin1/IREG1, the product of the SLC40A1 gene, is a main iron export protein in mammals. However, the way this gene is regulated by iron is still unclear. The aim of this study was to investigate the functional role of genomic SLC40A1 elements in response to iron. METHODS: Vectors containing either reverse similar 2.6 kb 5' flanking region or deletion constructs, including one devoid of an iron responsive element (SLC40A1-DeltaIRE-Luc), were analyzed by luciferase reporter gene in transfected HepG2, CaCO2 and U937 cells. Expression of iron genes and activity of the iron regulatory protein were also studied. RESULTS: Iron increased and desferrioxamine decreased luciferase activity in all the cell types using both the full-length construct and the promoter deletion constructs, in the absence of changes in SLC40A1 or luciferase mRNA levels. To test the role of the SLC40A1 5' untranslated region, we first demonstrated that wild type and not SLC40A1-DeltaIRE-Luc could bind iron regulatory protein. Then, in cells transfected with SLC40A1-DeltaIRE-Luc, we found that, in spite of iron regulatory protein activation, the response to iron manipulation was lost. CONCLUSIONS: We demonstrate that the iron responsive element in the SLC40A1 gene is functional and that it controls gene expression through the cytoplasmic iron regulatory protein system.


Asunto(s)
Proteínas de Transporte de Catión/genética , Regulación de la Expresión Génica , Hierro/fisiología , Elementos de Respuesta/fisiología , Regiones no Traducidas 5'/genética , Regiones no Traducidas 5'/fisiología , Animales , Línea Celular Tumoral , Deferoxamina/farmacología , Eliminación de Gen , Expresión Génica/efectos de los fármacos , Humanos , Hierro/farmacología , Quelantes del Hierro/farmacología , Luciferasas/genética , Luciferasas/metabolismo , Ratones , Regiones Promotoras Genéticas/genética , ARN Mensajero/metabolismo , Elementos de Respuesta/genética
14.
J Bioenerg Biomembr ; 34(1): 67-79, 2002 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-11860182

RESUMEN

Hepatic fibrosis due to iron overload is mediated by oxidant stress. The basic mechanisms underlying this process in vivo are still little understood. Acutely iron-dosed gerbils were assayed for lobular accumulation of hepatic lipid peroxidation by-products, oxidant-stress gene response, mitochondrial energy-dependent functions, and fibrogenesis. Iron overload in nonparenchymal cells caused an activation of hepatic stellate cells and fibrogenesis. Oxidant-stress gene response and accumulation of malondialdehyde-protein adducts were restricted to iron-filled nonparenchymal cells, sparing nearby hepatocytes. Concomitantly, a significant rise in the mitochondrial desferrioxamine-chelatable iron pool associated with the impairment of mitochondrial oxidative metabolism and the hepatic ATP decrease, was detected. Ultrastructural mitochondrial alterations were observed only in nonparenchymal cells. All biochemical and functional derangements were hindered by in vivo silybin administration which blocked completely fibrogenesis. Iron-induced oxidant stress in nonparenchymal cells appeared to bring about irreversible mitochondrial derangement associated with the onset of hepatic fibrosis.


Asunto(s)
Hepatocitos/metabolismo , Hierro/toxicidad , Estrés Oxidativo/efectos de los fármacos , Adenosina Trifosfato/biosíntesis , Animales , Enfermedad Hepática Inducida por Sustancias y Drogas , Transporte de Electrón , Complejo IV de Transporte de Electrones/metabolismo , Fibrosis/inducido químicamente , Fibrosis/patología , Fibrosis/prevención & control , Gerbillinae , Hepatocitos/efectos de los fármacos , Hepatocitos/patología , Hierro/administración & dosificación , Sobrecarga de Hierro/complicaciones , Sobrecarga de Hierro/patología , Hepatopatías/patología , Hepatopatías/prevención & control , Masculino , Potenciales de la Membrana , Mitocondrias Hepáticas/efectos de los fármacos , Mitocondrias Hepáticas/metabolismo , Mitocondrias Hepáticas/patología , Enfermedades Mitocondriales/inducido químicamente , Enfermedades Mitocondriales/prevención & control , Estrés Oxidativo/fisiología , Silimarina/farmacología , Silimarina/uso terapéutico
15.
Blood Cells Mol Dis ; 31(3): 299-304, 2003.
Artículo en Inglés | MEDLINE | ID: mdl-14636642

RESUMEN

The product of the SLC40A1 gene, ferroportin 1, is a main iron export protein. Pathogenic mutations in ferroportin 1 lead to an autosomal dominant hereditary iron overload syndrome characterized by high serum ferritin concentration, normal transferrin saturation, iron accumulation predominantly in macrophages, and marginal anemia. Iron overload occurs in both the African and the African-American populations, but a possible genetic basis has not been established. We analyzed the ferroportin 1 gene in 19 unrelated patients from southern Africa (N = 15) and the United States (N = 4) presenting with primary iron overload. We found a new c. 744 C-->T (Q248H) mutation in the SLC40A1 gene in 4 of these patients (3 Africans and 1 African-American). Among 22 first degree family members, 10 of whom were Q248H heterozygotes, the mutation was associated with a trend to higher serum ferritin to amino aspartate transferase ratios (means of 14.8 versus 4.3 microg/U; P = 0.1) and lower hemoglobin concentrations (means of 11.8 versus 13.2 g/dL; P = 0.1). The ratio corrects serum ferritin concentration for alcohol-induced hepatocellular damage. We also found heterozygosity for the Q248H mutation in 7 of 51 (14%) southern African community control participants selected because they had a serum ferritin concentration below 400 microg/L and in 5 of 100 (5%) anonymous African-Americans, but we did not find the change in 300 Caucasians with normal iron status and 25 Caucasians with non-HFE iron overload. The hemoglobin concentration was significantly lower in the African community controls with the Q248H mutation than in those without it. We conclude that the Q248H mutation is a common polymorphism in the ferroportin 1 gene in African populations that may be associated with mild anemia and a tendency to iron loading.


Asunto(s)
Población Negra/genética , Negro o Afroamericano/genética , Proteínas de Transporte de Catión/genética , Sobrecarga de Hierro/genética , Mutación/genética , Secuencia de Bases , Femenino , Ferritinas/sangre , Glutamina/genética , Glutamina/metabolismo , Pruebas Hematológicas , Histidina/genética , Histidina/metabolismo , Humanos , Hierro/sangre , Sobrecarga de Hierro/sangre , Persona de Mediana Edad , Datos de Secuencia Molecular , Polimorfismo Genético/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA