Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Int J Mol Sci ; 25(9)2024 Apr 29.
Artículo en Inglés | MEDLINE | ID: mdl-38732071

RESUMEN

Iron regulatory proteins (IRP1 and IRP2) are the master regulators of mammalian iron homeostasis. They bind to the iron-responsive elements (IREs) of the transcripts of iron-related genes to regulate their expression, thereby maintaining cellular iron availability. The primary method to measure the IRE-binding activity of IRPs is the electrophoresis mobility shift assay (EMSA). This method is particularly useful for evaluating IRP1 activity, since IRP1 is a bifunctional enzyme and its protein levels remain similar during conversion between the IRE-binding protein and cytosolic aconitase forms. Here, we exploited a method of using a biotinylated-IRE probe to separate IRE-binding IRPs followed by immunoblotting to analyze the IRE-binding activity. This method allows for the successful measurement of IRP activity in cultured cells and mouse tissues under various iron conditions. By separating IRE-binding IRPs from the rest of the lysates, this method increases the specificity of IRP antibodies and verifies whether a band represents an IRP, thereby revealing some previously unrecognized information about IRPs. With this method, we showed that the S711-phosphorylated IRP1 was found only in the IRE-binding form in PMA-treated Hep3B cells. Second, we found a truncated IRE-binding IRP2 isoform that is generated by proteolytic cleavage on sites in the 73aa insert region of the IRP2 protein. Third, we found that higher levels of SDS, compared to 1-2% SDS in regular loading buffer, could dramatically increase the band intensity of IRPs in immunoblots, especially in HL-60 cells. Fourth, we found that the addition of SDS or LDS to cell lysates activated protein degradation at 37 °C or room temperature, especially in HL-60 cell lysates. As this method is more practical, sensitive, and cost-effective, we believe that its application will enhance future research on iron regulation and metabolism.


Asunto(s)
Proteína 1 Reguladora de Hierro , Hierro , Humanos , Animales , Hierro/metabolismo , Proteína 1 Reguladora de Hierro/metabolismo , Proteína 1 Reguladora de Hierro/genética , Ratones , Proteína 2 Reguladora de Hierro/metabolismo , Proteína 2 Reguladora de Hierro/genética , Biotinilación , Elementos de Respuesta , Fosforilación , Proteínas Reguladoras del Hierro/metabolismo , Proteínas Reguladoras del Hierro/genética , Unión Proteica , Línea Celular Tumoral
2.
Heliyon ; 8(8): e10371, 2022 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-36061025

RESUMEN

Cardiomyopathy is a primary cause of death in Friedreich ataxia (FRDA) patients with defective iron-sulfur cluster (ISC) biogenesis due to loss of functional frataxin and in rare patients with functional loss of other ISC biogenesis factors. The mechanistic target of rapamycin (mTOR) and AKT signaling cascades that coordinate eukaryotic cell growth and metabolism with environmental inputs, including nutrients and growth factors, are crucial regulators of cardiovascular growth and homeostasis. We observed increased phosphorylation of AKT and dysregulation of multiple downstream effectors of mTORC1, including S6K1, S6, ULK1 and 4EBP1, in a cardiac/skeletal muscle specific FRDA conditional knockout (cKO) mouse model and in human cell lines depleted of ISC biogenesis factors. Knockdown of several mitochondrial metabolic proteins that are downstream targets of ISC biogenesis, including lipoyl synthase and subunit B of succinate dehydrogenase, also resulted in activation of mTOR and AKT signaling, suggesting that mTOR and AKT hyperactivations are part of the metabolic stress response to ISC deficiencies. Administration of rapamycin, a specific inhibitor of mTOR signaling, enhanced the survival of the Fxn cKO mice, providing proof of concept for the potential of mTOR inhibition to ameliorate cardiac disease in patients with defective ISC biogenesis. However, AKT phosphorylation remained high in rapamycin-treated Fxn cKO hearts, suggesting that parallel mTOR and AKT inhibition might be necessary to further improve the lifespan and healthspan of ISC deficient individuals.

3.
Blood Adv ; 2(20): 2732-2743, 2018 10 23.
Artículo en Inglés | MEDLINE | ID: mdl-30337301

RESUMEN

Heme oxygenase 1 (HMOX1), the inducible enzyme that catabolizes the degradation of heme into biliverdin, iron, and carbon monoxide, plays an essential role in the clearance of senescent and damaged red blood cells, systemic iron homeostasis, erythropoiesis, vascular hemostasis, and oxidative and inflammatory stress responses. In humans, HMOX1 deficiency causes a rare and lethal disease, characterized by severe anemia, intravascular hemolysis, as well as vascular and tissue damage. Hmox1 knockout (KO) mice recapitulated the phenotypes of HMOX1-deficiency patients and could be rescued by bone marrow (BM) transplantation that engrafted donor's hematopoietic stem cells into the recipient animals after myeloablation. To find better therapy and elucidate the contribution of macrophages to the pathogenesis of HMOX1-deficiency disease, we infused wild-type (WT) macrophages into Hmox1 KO mice. Results showed that WT macrophages engrafted and proliferated in the livers of Hmox1 KO mice, which corrected the microcytic anemia, rescued the intravascular hemolysis, restored iron homeostasis, eliminated kidney iron overload and tissue damage, and provided long-term protection. These results showed that a single macrophage infusion delivered a long-term curative effect in Hmox1 KO mice, obviating the need for BM transplantation, and suggested that the HMOX1 disease stems mainly from the loss of viable reticuloendothelial macrophages. Our work provides new insights into the etiology of HMOX1 deficiency and demonstrates the potential of infusion of WT macrophages to prevent disease in patients with HMOX1 deficiency and potentially other macrophage-related diseases.


Asunto(s)
Anemia Hemolítica/complicaciones , Anemia/genética , Trastornos del Crecimiento/complicaciones , Hemo-Oxigenasa 1/deficiencia , Hemólisis/genética , Trastornos del Metabolismo del Hierro/complicaciones , Hígado/fisiopatología , Macrófagos/metabolismo , Animales , Humanos , Ratones
4.
Blood ; 132(19): 2078-2087, 2018 11 08.
Artículo en Inglés | MEDLINE | ID: mdl-30213870

RESUMEN

Ferroportin (FPN), the only known vertebrate iron exporter, transports iron from intestinal, splenic, and hepatic cells into the blood to provide iron to other tissues and cells in vivo. Most of the circulating iron is consumed by erythroid cells to synthesize hemoglobin. Here we found that erythroid cells not only consumed large amounts of iron, but also returned significant amounts of iron to the blood. Erythroblast-specific Fpn knockout (Fpn KO) mice developed lower serum iron levels in conjunction with tissue iron overload and increased FPN expression in spleen and liver without changing hepcidin levels. Our results also showed that Fpn KO mice, which suffer from mild hemolytic anemia, were sensitive to phenylhydrazine-induced oxidative stress but were able to tolerate iron deficiency upon exposure to a low-iron diet and phlebotomy, supporting that the anemia of Fpn KO mice resulted from erythrocytic iron overload and resulting oxidative injury rather than a red blood cell (RBC) production defect. Moreover, we found that the mean corpuscular volume (MCV) values of gain-of-function FPN mutation patients were positively associated with serum transferrin saturations, whereas MCVs of loss-of-function FPN mutation patients were not, supporting that erythroblasts donate iron to blood through FPN in response to serum iron levels. Our results indicate that FPN of erythroid cells plays an unexpectedly essential role in maintaining systemic iron homeostasis and protecting RBCs from oxidative stress, providing insight into the pathophysiology of FPN diseases.


Asunto(s)
Anemia Ferropénica/genética , Proteínas de Transporte de Catión/genética , Células Eritroides/patología , Hemólisis , Hierro/sangre , Mutación con Pérdida de Función , Estrés Oxidativo , Anemia Ferropénica/sangre , Anemia Ferropénica/patología , Animales , Proteínas de Transporte de Catión/análisis , Células Eritroides/metabolismo , Mutación con Ganancia de Función , Humanos , Hierro/análisis , Deficiencias de Hierro , Sobrecarga de Hierro/sangre , Sobrecarga de Hierro/genética , Sobrecarga de Hierro/patología , Hígado/metabolismo , Hígado/patología , Ratones , Ratones Noqueados , Bazo/metabolismo , Bazo/patología
5.
Blood Adv ; 2(10): 1146-1156, 2018 05 22.
Artículo en Inglés | MEDLINE | ID: mdl-29784770

RESUMEN

Given the essential roles of iron-sulfur (Fe-S) cofactors in mediating electron transfer in the mitochondrial respiratory chain and supporting heme biosynthesis, mitochondrial dysfunction is a common feature in a growing list of human Fe-S cluster biogenesis disorders, including Friedreich ataxia and GLRX5-related sideroblastic anemia. Here, our studies showed that restriction of Fe-S cluster biogenesis not only compromised mitochondrial oxidative metabolism but also resulted in decreased overall histone acetylation and increased H3K9me3 levels in the nucleus and increased acetylation of α-tubulin in the cytosol by decreasing the lipoylation of the pyruvate dehydrogenase complex, decreasing levels of succinate dehydrogenase and the histone acetyltransferase ELP3, and increasing levels of the tubulin acetyltransferase MEC17. Previous studies have shown that the metabolic shift in Toll-like receptor (TLR)-activated myeloid cells involves rapid activation of glycolysis and subsequent mitochondrial respiratory failure due to nitric oxide (NO)-mediated damage to Fe-S proteins. Our studies indicated that TLR activation also actively suppresses many components of the Fe-S cluster biogenesis machinery, which exacerbates NO-mediated damage to Fe-S proteins by interfering with cluster recovery. These results reveal new regulatory pathways and novel roles of the Fe-S cluster biogenesis machinery in modifying the epigenome and acetylome and provide new insights into the etiology of Fe-S cluster biogenesis disorders.


Asunto(s)
Histonas/metabolismo , Proteínas Hierro-Azufre/metabolismo , Receptores Toll-Like/metabolismo , Tubulina (Proteína)/metabolismo , Acetilación , Humanos
6.
Science ; 359(6383): 1520-1523, 2018 03 30.
Artículo en Inglés | MEDLINE | ID: mdl-29599243

RESUMEN

Malaria parasites invade red blood cells (RBCs), consume copious amounts of hemoglobin, and severely disrupt iron regulation in humans. Anemia often accompanies malaria disease; however, iron supplementation therapy inexplicably exacerbates malarial infections. Here we found that the iron exporter ferroportin (FPN) was highly abundant in RBCs, and iron supplementation suppressed its activity. Conditional deletion of the Fpn gene in erythroid cells resulted in accumulation of excess intracellular iron, cellular damage, hemolysis, and increased fatality in malaria-infected mice. In humans, a prevalent FPN mutation, Q248H (glutamine to histidine at position 248), prevented hepcidin-induced degradation of FPN and protected against severe malaria disease. FPN Q248H appears to have been positively selected in African populations in response to the impact of malaria disease. Thus, FPN protects RBCs against oxidative stress and malaria infection.


Asunto(s)
Proteínas de Transporte de Catión/metabolismo , Eritrocitos/metabolismo , Hemólisis , Hierro/metabolismo , Malaria/epidemiología , Sustitución de Aminoácidos , Anemia/metabolismo , Animales , Población Negra/genética , Proteínas de Transporte de Catión/genética , Niño , Eritrocitos/efectos de los fármacos , Femenino , Hepcidinas/metabolismo , Hepcidinas/farmacología , Humanos , Hierro/administración & dosificación , Hierro/farmacología , Malaria/sangre , Malaria/genética , Masculino , Ratones , Ratones Noqueados , Mutación , Estrés Oxidativo , Riesgo , Selección Genética , Eliminación de Secuencia , Zambia/epidemiología
7.
J Clin Invest ; 128(4): 1317-1325, 2018 04 02.
Artículo en Inglés | MEDLINE | ID: mdl-29480820

RESUMEN

Chuvash polycythemia is an inherited disease caused by a homozygous germline VHLR200W mutation, which leads to impaired degradation of HIF2α, elevated levels of serum erythropoietin, and erythrocytosis/polycythemia. This phenotype is recapitulated by a mouse model bearing a homozygous VhlR200W mutation. We previously showed that iron-regulatory protein 1-knockout (Irp1-knockout) mice developed erythrocytosis/polycythemia through translational derepression of Hif2α, suggesting that IRP1 could be a therapeutic target to treat Chuvash polycythemia. Here, we fed VhlR200W mice supplemented with Tempol, a small, stable nitroxide molecule and observed that Tempol decreased erythropoietin production, corrected splenomegaly, normalized hematocrit levels, and increased the lifespans of these mice. We attribute the reversal of erythrocytosis/polycythemia to translational repression of Hif2α expression by Tempol-mediated increases in the IRE-binding activity of Irp1, as reversal of polycythemia was abrogated in VhlR200W mice in which Irp1 was genetically ablated. Thus, a new approach to the treatment of patients with Chuvash polycythemia may include dietary supplementation of Tempol, which decreased Hif2α expression and markedly reduced life-threatening erythrocytosis/polycythemia in the VhlR200W mice.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/biosíntesis , Óxidos N-Cíclicos/farmacología , Regulación de la Expresión Génica/efectos de los fármacos , Proteína 1 Reguladora de Hierro/metabolismo , Policitemia/tratamiento farmacológico , Biosíntesis de Proteínas/efectos de los fármacos , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Humanos , Proteína 1 Reguladora de Hierro/genética , Ratones , Ratones Mutantes , Policitemia/genética , Policitemia/metabolismo , Policitemia/patología , Marcadores de Spin
8.
PLoS One ; 6(10): e25404, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-22003390

RESUMEN

Genetic ablation of Iron Regulatory Protein 2 (Irp2, Ireb2), which post-transcriptionally regulates iron metabolism genes, causes a gait disorder in mice that progresses to hind-limb paralysis. Here we have demonstrated that misregulation of iron metabolism from loss of Irp2 causes lower motor neuronal degeneration with significant spinal cord axonopathy. Mitochondria in the lumbar spinal cord showed significantly decreased Complex I and II activities, and abnormal morphology. Lower motor neurons appeared to be the most adversely affected neurons, and we show that functional iron starvation due to misregulation of iron import and storage proteins, including transferrin receptor 1 and ferritin, may have a causal role in disease. We demonstrated that two therapeutic approaches were beneficial for motor neuron survival. First, we activated a homologous protein, IRP1, by oral Tempol treatment and found that axons were partially spared from degeneration. Secondly, we genetically decreased expression of the iron storage protein, ferritin, to diminish functional iron starvation. These data suggest that functional iron deficiency may constitute a previously unrecognized molecular basis for degeneration of motor neurons in mice.


Asunto(s)
Eliminación de Gen , Deficiencias de Hierro , Proteína 2 Reguladora de Hierro/deficiencia , Proteína 2 Reguladora de Hierro/genética , Mitocondrias/metabolismo , Neuronas Motoras/metabolismo , Neuronas Motoras/patología , Animales , Apoferritinas/biosíntesis , Atrofia/metabolismo , Axones/efectos de los fármacos , Axones/metabolismo , Axones/patología , Biomarcadores/metabolismo , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Encéfalo/patología , Óxidos N-Cíclicos/farmacología , Homeostasis/efectos de los fármacos , Homeostasis/genética , Hierro/metabolismo , Proteína 1 Reguladora de Hierro/deficiencia , Proteína 1 Reguladora de Hierro/metabolismo , Ratones , Mitocondrias/efectos de los fármacos , Mitocondrias/genética , Mitocondrias/patología , Neuronas Motoras/efectos de los fármacos , Estrés Oxidativo/efectos de los fármacos , Estrés Oxidativo/genética , Marcadores de Spin , Médula Espinal/efectos de los fármacos , Médula Espinal/metabolismo , Médula Espinal/patología
9.
Ann N Y Acad Sci ; 1012: 65-83, 2004 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-15105256

RESUMEN

In mammals, iron regulatory proteins 1 and 2 (IRP1 and IRP2) posttranscriptionally regulate expression of several iron metabolism proteins including ferritin and transferrin receptor. Genetically engineered mice that lack IRP2, but have the normal complement of IRP1, develop adult-onset neurodegenerative disease associated with inappropriately high expression of ferritin in degenerating neurons. Here, we report that mice that are homozygous for a targeted deletion of IRP2 and heterozygous for a targeted deletion of IRP1 (IRP1+/- IRP2-/-) develop a much more severe form of neurodegeneration, characterized by widespread axonopathy and eventually by subtle vacuolization in several areas, particularly in the substantia nigra. Axonopathy develops in white matter tracts in which marked increases in ferric iron and ferritin expression are detected. Axonal degeneration is significant and widespread before evidence for abnormalities or loss of neuronal cell bodies can be detected. Ultimately, neuronal cell bodies degenerate in the substantia nigra and some other vulnerable areas, microglia are activated, and vacuoles appear. Mice manifest gait and motor impairment at stages when axonopathy is pronounced, but neuronal cell body loss is minimal. These observations suggest that therapeutic strategies that aim to revitalize neurons by treatment with neurotrophic factors may be of value in IRP2-/- and IRP1+/- IRP2-/- mouse models of neurodegeneration.


Asunto(s)
Proteínas Reguladoras del Hierro/deficiencia , Hierro/metabolismo , Degeneración Nerviosa/metabolismo , Factores de Edad , Animales , Axones/patología , Axones/ultraestructura , Western Blotting/métodos , Encéfalo/anatomía & histología , Encéfalo/metabolismo , Encéfalo/patología , Recuento de Células/métodos , Células Cultivadas , Embrión de Mamíferos , Ferritinas/metabolismo , Fuerza de la Mano/fisiología , Inmunohistoquímica/métodos , Proteínas Reguladoras del Hierro/sangre , Proteínas Reguladoras del Hierro/genética , Proteínas Reguladoras del Hierro/metabolismo , Ratones , Ratones Noqueados , Microglía/metabolismo , Microscopía Electrónica/métodos , Degeneración Nerviosa/patología , Degeneración Nerviosa/fisiopatología , Neuronas/metabolismo , Neuronas/patología , Oligodendroglía/metabolismo , Oligodendroglía/patología , Receptores de Transferrina/metabolismo , Células Madre , Tirosina 3-Monooxigenasa/metabolismo , Ubiquitina/metabolismo , Vacuolas/patología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...