Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Heliyon ; 8(8): e10371, 2022 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-36061025

RESUMO

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.

2.
Nat Commun ; 11(1): 6310, 2020 12 09.
Artigo em Inglês | MEDLINE | ID: mdl-33298951

RESUMO

Heme biosynthesis and iron-sulfur cluster (ISC) biogenesis are two major mammalian metabolic pathways that require iron. It has long been known that these two pathways interconnect, but the previously described interactions do not fully explain why heme biosynthesis depends on intact ISC biogenesis. Herein we identify a previously unrecognized connection between these two pathways through our discovery that human aminolevulinic acid dehydratase (ALAD), which catalyzes the second step of heme biosynthesis, is an Fe-S protein. We find that several highly conserved cysteines and an Ala306-Phe307-Arg308 motif of human ALAD are important for [Fe4S4] cluster acquisition and coordination. The enzymatic activity of human ALAD is greatly reduced upon loss of its Fe-S cluster, which results in reduced heme biosynthesis in human cells. As ALAD provides an early Fe-S-dependent checkpoint in the heme biosynthetic pathway, our findings help explain why heme biosynthesis depends on intact ISC biogenesis.


Assuntos
Heme/biossíntese , Proteínas Ferro-Enxofre/metabolismo , Ferro/metabolismo , Sintase do Porfobilinogênio/metabolismo , Enxofre/metabolismo , Motivos de Aminoácidos , Vias Biossintéticas , Linhagem Celular , Coenzimas/metabolismo , Cisteína/metabolismo , Humanos , Proteínas Ferro-Enxofre/genética , Sintase do Porfobilinogênio/genética , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo
3.
Blood Adv ; 2(10): 1146-1156, 2018 05 22.
Artigo em Inglês | MEDLINE | ID: mdl-29784770

RESUMO

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.


Assuntos
Histonas/metabolismo , Proteínas Ferro-Enxofre/metabolismo , Receptores Toll-Like/metabolismo , Tubulina (Proteína)/metabolismo , Acetilação , Humanos
4.
Methods Enzymol ; 547: 275-307, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25416363

RESUMO

Iron is a heavily utilized element in organisms and numerous mechanisms accordingly regulate the trafficking, metabolism, and storage of iron. Despite the high regulation of iron homeostasis, several diseases and mutations can lead to the misregulation and often accumulation of iron in the cytosol or mitochondria of tissues. To understand the genesis of iron overload, it is necessary to employ various techniques to quantify iron in organisms and mitochondria. This chapter discusses techniques for determining the total iron content of tissue samples, ranging from colorimetric determination of iron concentrations, atomic absorption spectroscopy, inductively coupled plasma-optical emission spectroscopy, and inductively coupled plasma-mass spectrometry. In addition, we discuss in situ techniques for analyzing iron including electron microscopic nonheme iron histochemistry, electron energy loss spectroscopy, synchrotron X-ray fluorescence imaging, and confocal Raman microscopy. Finally, we discuss biophysical methods for studying iron in isolated mitochondria, including ultraviolet-visible, electron paramagnetic resonance, X-ray absorbance, and Mössbauer spectroscopies. This chapter should aid researchers to select and interpret mitochondrial iron quantifications.


Assuntos
Bioquímica/métodos , Biofísica/métodos , Ferro/metabolismo , Mitocôndrias/metabolismo , Animais , Colorimetria/métodos , Espectroscopia de Ressonância de Spin Eletrônica/métodos , Humanos , Espectrometria de Massas/métodos , Microscopia Eletrônica/métodos , Espectrofotometria Atômica/métodos , Espectrofotometria Ultravioleta/métodos , Espectroscopia de Mossbauer/métodos , Análise Espectral/métodos , Análise Espectral Raman/métodos
5.
Cell Metab ; 19(3): 445-57, 2014 Mar 04.
Artigo em Inglês | MEDLINE | ID: mdl-24606901

RESUMO

Iron sulfur (Fe-S) clusters, preassembled on the ISCU scaffold, are transferred to target proteins or to intermediate scaffolds by a dedicated chaperone-cochaperone system. However, the molecular mechanisms that underlie substrate discrimination and guide delivery of nascent clusters to specific subsets of Fe-S recipients are poorly understood. Here, we identified interacting partners of the cochaperone HSC20 and discovered that LYR motifs are molecular signatures of specific recipient Fe-S proteins or accessory factors that assist Fe-S cluster delivery. In succinate dehydrogenase B, two LYR motifs engage the ISCU-HSC20-HSPA9 complex to aid incorporation of three Fe-S clusters within the final structure of complex II. Moreover, we show that members of the LYR motif family which assist assembly of complexes II or III, SDHAF1 and LYRM7, respectively, are HSC20 binding partners. Our studies unveil a network of interactions between HSC20 and LYR motif-containing proteins that are key to the assembly and function of complexes I, II, and III.


Assuntos
Proteínas Ferro-Enxofre/metabolismo , Chaperonas Moleculares/metabolismo , Motivos de Aminoácidos , Sequência de Aminoácidos , Sítios de Ligação , Células HEK293 , Proteínas de Choque Térmico HSP70/antagonistas & inibidores , Proteínas de Choque Térmico HSP70/genética , Proteínas de Choque Térmico HSP70/metabolismo , Células HeLa , Humanos , Proteínas Ferro-Enxofre/química , Proteínas Mitocondriais/antagonistas & inibidores , Proteínas Mitocondriais/genética , Proteínas Mitocondriais/metabolismo , Chaperonas Moleculares/antagonistas & inibidores , Chaperonas Moleculares/química , Chaperonas Moleculares/genética , Dados de Sequência Molecular , Ligação Proteica , Estrutura Terciária de Proteína , Interferência de RNA , RNA Interferente Pequeno/metabolismo
6.
Hum Mol Genet ; 23(1): 24-39, 2014 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-23943793

RESUMO

Iron-sulfur (Fe-S) clusters are ancient enzyme cofactors found in virtually all life forms. We evaluated the physiological effects of chronic Fe-S cluster deficiency in human skeletal muscle, a tissue that relies heavily on Fe-S cluster-mediated aerobic energy metabolism. Despite greatly decreased oxidative capacity, muscle tissue from patients deficient in the Fe-S cluster scaffold protein ISCU showed a predominance of type I oxidative muscle fibers and higher capillary density, enhanced expression of transcriptional co-activator PGC-1α and increased mitochondrial fatty acid oxidation genes. These Fe-S cluster-deficient muscles showed a dramatic up-regulation of the ketogenic enzyme HMGCS2 and the secreted protein FGF21 (fibroblast growth factor 21). Enhanced muscle FGF21 expression was reflected by elevated circulating FGF21 levels in the patients, and robust FGF21 secretion could be recapitulated by respiratory chain inhibition in cultured myotubes. Our findings reveal that mitochondrial energy starvation elicits a coordinated response in Fe-S-deficient skeletal muscle that is reflected systemically by increased plasma FGF21 levels.


Assuntos
Acidose Láctica/congênito , Fatores de Crescimento de Fibroblastos/metabolismo , Hidroximetilglutaril-CoA Sintase/metabolismo , Proteínas Ferro-Enxofre/metabolismo , Músculo Esquelético/metabolismo , Doenças Musculares/congênito , Fatores de Transcrição/genética , Acidose Láctica/genética , Acidose Láctica/metabolismo , Acidose Láctica/patologia , Adulto , Idoso , Estudos de Casos e Controles , Células Cultivadas , Metabolismo Energético , Feminino , Fatores de Crescimento de Fibroblastos/genética , Regulação da Expressão Gênica , Humanos , Hidroximetilglutaril-CoA Sintase/genética , Proteínas Ferro-Enxofre/genética , Masculino , Pessoa de Meia-Idade , Mitocôndrias Musculares/metabolismo , Mitocôndrias Musculares/patologia , Doenças Musculares/genética , Doenças Musculares/metabolismo , Doenças Musculares/patologia , Coativador 1-alfa do Receptor gama Ativado por Proliferador de Peroxissomo , Fatores de Transcrição/metabolismo
7.
Cell Metab ; 17(2): 271-81, 2013 Feb 05.
Artigo em Inglês | MEDLINE | ID: mdl-23395173

RESUMO

Iron regulatory proteins (Irps) 1 and 2 posttranscriptionally control the expression of transcripts that contain iron-responsive element (IRE) sequences, including ferritin, ferroportin, transferrin receptor, and hypoxia-inducible factor 2α (HIF2α). We report here that mice with targeted deletion of Irp1 developed pulmonary hypertension and polycythemia that was exacerbated by a low-iron diet. Hematocrits increased to 65% in iron-starved mice, and many polycythemic mice died of abdominal hemorrhages. Irp1 deletion enhanced HIF2α protein expression in kidneys of Irp1(-/-) mice, which led to increased erythropoietin (EPO) expression, polycythemia, and concomitant tissue iron deficiency. Increased HIF2α expression in pulmonary endothelial cells induced high expression of endothelin-1, likely contributing to the pulmonary hypertension of Irp1(-/-) mice. Our results reveal why anemia is an early physiological consequence of iron deficiency, highlight the physiological significance of Irp1 in regulating erythropoiesis and iron distribution, and provide important insights into the molecular pathogenesis of pulmonary hypertension.


Assuntos
Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Deleção de Genes , Hipertensão Pulmonar/complicações , Proteína 1 Reguladora do Ferro/metabolismo , Policitemia/complicações , Biossíntese de Proteínas , Animais , Dieta , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/metabolismo , Células Endoteliais/patologia , Endotelina-1/genética , Endotelina-1/metabolismo , Eritropoetina/sangue , Hemorragia Gastrointestinal/sangue , Hemorragia Gastrointestinal/complicações , Hemorragia Gastrointestinal/patologia , Hematopoese Extramedular/efeitos dos fármacos , Hipertensão Pulmonar/sangue , Hipertensão Pulmonar/patologia , Ferro/farmacologia , Proteína 1 Reguladora do Ferro/deficiência , Proteína 2 Reguladora do Ferro/metabolismo , Longevidade , Camundongos , Modelos Biológicos , Degeneração Neural/sangue , Degeneração Neural/complicações , Degeneração Neural/patologia , Especificidade de Órgãos/efeitos dos fármacos , Policitemia/sangue , Policitemia/patologia , Biossíntese de Proteínas/efeitos dos fármacos , Ativação Transcricional/efeitos dos fármacos , Ativação Transcricional/genética
8.
J Biol Chem ; 287(48): 40119-30, 2012 Nov 23.
Artigo em Inglês | MEDLINE | ID: mdl-23035118

RESUMO

BACKGROUND: ISCU myopathy is a disease caused by muscle-specific deficiency of the Fe-S cluster scaffold protein ISCU. RESULTS: MyoD expression enhanced ISCU mRNA mis-splicing, and oxidative stress exacerbated ISCU depletion in patient cells. CONCLUSION: ISCU protein deficiency in patients results from muscle-specific mis-splicing as well as oxidative stress. SIGNIFICANCE: Oxidative stress negatively influences the mammalian Fe-S cluster assembly machinery by destabilization of ISCU. Iron-sulfur (Fe-S) cluster cofactors are formed on the scaffold protein ISCU. ISCU myopathy is a disease caused by an intronic mutation that leads to abnormally spliced ISCU mRNA. We found that two predominant mis-spliced ISCU mRNAs produce a truncated and short-lived ISCU protein product in multiple patient cell types. Expression of the muscle-specific transcription factor MyoD further diminished normal splicing of ISCU mRNA in patient myoblasts, demonstrating that the process of muscle differentiation enhances the loss of normal ISCU mRNA splicing. ISCU protein was nearly undetectable in patient skeletal muscle, but was higher in patient myoblasts, fibroblasts, and lymphoblasts. We next treated patient cells with pro-oxidants to mimic the oxidative stress associated with muscle activity. Brief hydrogen peroxide treatment or incubation in an enriched oxygen atmosphere led to a marked further reduction of ISCU protein levels, which could be prevented by pretreatment with the antioxidant ascorbate. Thus, we conclude that skeletal muscle differentiation of patient cells causes a higher degree of abnormal ISCU splicing and that oxidative stress resulting from skeletal muscle work destabilizes the small amounts of normal ISCU protein generated in patient skeletal muscles.


Assuntos
Diferenciação Celular , Proteínas Ferro-Enxofre/genética , Doenças Mitocondriais/metabolismo , Músculo Esquelético/citologia , Estresse Oxidativo , Splicing de RNA , Adulto , Idoso , Animais , Feminino , Humanos , Proteínas Ferro-Enxofre/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Pessoa de Meia-Idade , Doenças Mitocondriais/genética , Doenças Mitocondriais/fisiopatologia , Músculo Esquelético/metabolismo , Proteína MyoD/genética , Proteína MyoD/metabolismo , Especificidade de Órgãos , Adulto Jovem
9.
Cancer Cell ; 20(3): 315-27, 2011 Sep 13.
Artigo em Inglês | MEDLINE | ID: mdl-21907923

RESUMO

Inactivation of the TCA cycle enzyme, fumarate hydratase (FH), drives a metabolic shift to aerobic glycolysis in FH-deficient kidney tumors and cell lines from patients with hereditary leiomyomatosis renal cell cancer (HLRCC), resulting in decreased levels of AMP-activated kinase (AMPK) and p53 tumor suppressor, and activation of the anabolic factors, acetyl-CoA carboxylase and ribosomal protein S6. Reduced AMPK levels lead to diminished expression of the DMT1 iron transporter, and the resulting cytosolic iron deficiency activates the iron regulatory proteins, IRP1 and IRP2, and increases expression of the hypoxia inducible factor HIF-1α, but not HIF-2α. Silencing of HIF-1α or activation of AMPK diminishes invasive activities, indicating that alterations of HIF-1α and AMPK contribute to the oncogenic growth of FH-deficient cells.


Assuntos
Proteínas Quinases Ativadas por AMP/metabolismo , Fumarato Hidratase/deficiência , Deficiências de Ferro , Neoplasias Renais/metabolismo , Leiomiomatose/congênito , Acetilcoenzima A/biossíntese , Acetil-CoA Carboxilase/biossíntese , Acetil-CoA Carboxilase/metabolismo , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos/biossíntese , Proteínas de Transporte de Cátions/biossíntese , Linhagem Celular Tumoral , Fumarato Hidratase/metabolismo , Glicólise/efeitos dos fármacos , Humanos , Subunidade alfa do Fator 1 Induzível por Hipóxia/biossíntese , Subunidade alfa do Fator 1 Induzível por Hipóxia/genética , Proteína 1 Reguladora do Ferro/biossíntese , Proteína 1 Reguladora do Ferro/metabolismo , Proteína 2 Reguladora do Ferro/biossíntese , Proteína 2 Reguladora do Ferro/metabolismo , Neoplasias Renais/enzimologia , Neoplasias Renais/patologia , Leiomiomatose/metabolismo , Leiomiomatose/patologia , Camundongos , NADP/biossíntese , Síndromes Neoplásicas Hereditárias , Ribose/biossíntese , Proteína S6 Ribossômica/biossíntese , Proteína S6 Ribossômica/metabolismo , Neoplasias Cutâneas , Tenoiltrifluoracetona/farmacologia , Proteína Supressora de Tumor p53/biossíntese , Neoplasias Uterinas
10.
Am J Hum Genet ; 89(4): 486-95, 2011 Oct 07.
Artigo em Inglês | MEDLINE | ID: mdl-21944046

RESUMO

Severe combined deficiency of the 2-oxoacid dehydrogenases, associated with a defect in lipoate synthesis and accompanied by defects in complexes I, II, and III of the mitochondrial respiratory chain, is a rare autosomal recessive syndrome with no obvious causative gene defect. A candidate locus for this syndrome was mapped to chromosomal region 2p14 by microcell-mediated chromosome transfer in two unrelated families. Unexpectedly, analysis of genes in this area identified mutations in two different genes, both of which are involved in [Fe-S] cluster biogenesis. A homozygous missense mutation, c.545G>A, near the splice donor of exon 6 in NFU1 predicting a p.Arg182Gln substitution was found in one of the families. The mutation results in abnormal mRNA splicing of exon 6, and no mature protein could be detected in fibroblast mitochondria. A single base-pair duplication c.123dupA was identified in BOLA3 in the second family, causing a frame shift that produces a premature stop codon (p.Glu42Argfs(∗)13). Transduction of fibroblast lines with retroviral vectors expressing the mitochondrial, but not the cytosolic isoform of NFU1 and with isoform 1, but not isoform 2 of BOLA3 restored both respiratory chain function and oxoacid dehydrogenase complexes. NFU1 was previously proposed to be an alternative scaffold to ISCU for the biogenesis of [Fe-S] centers in mitochondria, and the function of BOLA3 was previously unknown. Our results demonstrate that both play essential roles in the production of [Fe-S] centers for the normal maturation of lipoate-containing 2-oxoacid dehydrogenases, and for the assembly of the respiratory chain complexes.


Assuntos
Proteínas de Transporte/genética , Mutação , Oxirredutases/metabolismo , Proteínas/genética , Citosol/metabolismo , Transporte de Elétrons , Éxons , Saúde da Família , Feminino , Fibroblastos/metabolismo , Homozigoto , Humanos , Proteínas Ferro-Enxofre/metabolismo , Masculino , Mitocôndrias/metabolismo , Proteínas Mitocondriais , Mutação de Sentido Incorreto
11.
Blood ; 116(1): 97-108, 2010 Jul 08.
Artigo em Inglês | MEDLINE | ID: mdl-20407036

RESUMO

Human red cell differentiation requires the action of erythropoietin on committed progenitor cells. In iron deficiency, committed erythroid progenitors lose responsiveness to erythropoietin, resulting in hypoplastic anemia. To address the basis for iron regulation of erythropoiesis, we established primary hematopoietic cultures with transferrin saturation levels that restricted erythropoiesis but permitted granulopoiesis and megakaryopoiesis. Experiments in this system identified as a critical regulatory element the aconitases, multifunctional iron-sulfur cluster proteins that metabolize citrate to isocitrate. Iron restriction suppressed mitochondrial and cytosolic aconitase activity in erythroid but not granulocytic or megakaryocytic progenitors. An active site aconitase inhibitor, fluorocitrate, blocked erythroid differentiation in a manner similar to iron deprivation. Exogenous isocitrate abrogated the erythroid iron restriction response in vitro and reversed anemia progression in iron-deprived mice. The mechanism for aconitase regulation of erythropoiesis most probably involves both production of metabolic intermediates and modulation of erythropoietin signaling. One relevant signaling pathway appeared to involve protein kinase Calpha/beta, or possibly protein kinase Cdelta, whose activities were regulated by iron, isocitrate, and erythropoietin.


Assuntos
Células Precursoras Eritroides/efeitos dos fármacos , Eritropoese/efeitos dos fármacos , Proteína 1 Reguladora do Ferro/metabolismo , Ferro/farmacologia , Anemia Ferropriva/sangue , Anemia Ferropriva/etiologia , Anemia Ferropriva/metabolismo , Animais , Diferenciação Celular/efeitos dos fármacos , Células Cultivadas , Células Precursoras Eritroides/citologia , Células Precursoras Eritroides/metabolismo , Feminino , Citometria de Fluxo , Humanos , Immunoblotting , Deficiências de Ferro , Proteína 1 Reguladora do Ferro/genética , Isocitratos/administração & dosagem , Células K562 , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Interferência de RNA , Transdução de Sinais/efeitos dos fármacos
12.
Blood ; 115(4): 860-9, 2010 Jan 28.
Artigo em Inglês | MEDLINE | ID: mdl-19965627

RESUMO

Mammalian ferrochelatase, the terminal enzyme in the heme biosynthetic pathway, possesses an iron-sulfur [2Fe-2S] cluster that does not participate in catalysis. We investigated ferrochelatase expression in iron-deficient erythropoietic tissues of mice lacking iron regulatory protein 2, in iron-deficient murine erythroleukemia cells, and in human patients with ISCU myopathy. Ferrochelatase activity and protein levels were dramatically decreased in Irp2(-/-) spleens, whereas ferrochelatase mRNA levels were increased, demonstrating posttranscriptional regulation of ferrochelatase in vivo. Translation of ferrochelatase mRNA was unchanged in iron-depleted murine erythroleukemia cells, and the stability of mature ferrochelatase protein was also unaffected. However, the stability of newly formed ferrochelatase protein was dramatically decreased during iron deficiency. Ferrochelatase was also severely depleted in muscle biopsies and cultured myoblasts from patients with ISCU myopathy, a disease caused by deficiency of a scaffold protein required for Fe-S cluster assembly. Together, these data suggest that decreased Fe-S cluster availability because of cellular iron depletion or impaired Fe-S cluster assembly causes reduced maturation and stabilization of apo-ferrochelatase, providing a direct link between Fe-S biogenesis and completion of heme biosynthesis. We propose that decreased heme biosynthesis resulting from impaired Fe-S cluster assembly can contribute to the pathogenesis of diseases caused by defective Fe-S cluster biogenesis.


Assuntos
Anemia Ferropriva/metabolismo , Ferroquelatase/metabolismo , Heme/biossíntese , Ferro/metabolismo , Miopatias Mitocondriais/metabolismo , Enxofre/metabolismo , Anemia Ferropriva/genética , Anemia Ferropriva/patologia , Animais , Biópsia , Linhagem Celular Tumoral , Eritrócitos/citologia , Eritrócitos/enzimologia , Ferroquelatase/genética , Regulação Enzimológica da Expressão Gênica , Humanos , Proteína 2 Reguladora do Ferro/genética , Leucemia Eritroblástica Aguda/patologia , Camundongos , Camundongos Mutantes , Miopatias Mitocondriais/genética , Miopatias Mitocondriais/patologia , Músculo Esquelético/enzimologia , Músculo Esquelético/patologia , Estresse Oxidativo/fisiologia , Processamento de Proteína Pós-Traducional , Processamento Pós-Transcricional do RNA , RNA Mensageiro/metabolismo
13.
Cell Metab ; 10(2): 80-1, 2009 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-19656484

RESUMO

A large-scale computational and genetic analysis study by Nilsson et al. (2009) has identified five genes that coexpress with heme biosynthetic enzymes and are required for normal heme synthesis. Several are implicated in iron-sulfur cluster biogenesis, and malfunction of these genes may repress heme synthesis by activating the IRE/IRP posttranscriptional regulatory system.


Assuntos
Heme/biossíntese , Proteínas Ferro-Enxofre/metabolismo , Animais , Heme/metabolismo , Humanos , Proteínas Reguladoras de Ferro/metabolismo , Proteínas de Transporte da Membrana Mitocondrial/genética , Proteínas de Transporte da Membrana Mitocondrial/metabolismo , Família Multigênica , Peixe-Zebra
14.
Hum Mol Genet ; 18(16): 3014-25, 2009 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-19454487

RESUMO

The LYR family consists of proteins of diverse functions that contain the conserved tripeptide 'LYR' near the N-terminus, and it includes Isd11, which was previously observed to have an important role in iron-sulfur (Fe-S) cluster biogenesis in Saccharomyces cerevisiae. Here, we have cloned and characterized human ISD11 and shown that human ISD11 forms a stable complex in vivo with the human cysteine desulfurase (ISCS), which generates the inorganic sulfur needed for Fe-S protein biogenesis. Similar to ISCS, we have found that ISD11 localizes to the mitochondrial compartment, as expected, but also to the nucleus of mammalian cells. Using RNA-interference techniques, we have shown that suppression of human ISD11 inactivated mitochondrial and cytosolic aconitases. In addition, ISD11 suppression activated iron-responsive element-binding activity of iron regulatory protein 1, increased protein levels of iron regulatory protein 2, and resulted in abnormal punctate ferric iron accumulations in cells. These results indicate that ISD11 is important in the biogenesis of Fe-S clusters in mammalian cells, and its loss disrupts normal mitochondrial and cytosolic iron homeostasis.


Assuntos
Homeostase , Proteínas Reguladoras de Ferro/metabolismo , Ferro/metabolismo , Enxofre/metabolismo , Sequência de Aminoácidos , Liases de Carbono-Enxofre/genética , Liases de Carbono-Enxofre/metabolismo , Citosol/química , Citosol/metabolismo , Células HeLa , Humanos , Proteína 1 Reguladora do Ferro/genética , Proteína 1 Reguladora do Ferro/metabolismo , Proteína 2 Reguladora do Ferro/genética , Proteína 2 Reguladora do Ferro/metabolismo , Proteínas Reguladoras de Ferro/química , Proteínas Reguladoras de Ferro/genética , Mitocôndrias/química , Mitocôndrias/metabolismo , Dados de Sequência Molecular , Transporte Proteico , Alinhamento de Sequência
15.
Proc Natl Acad Sci U S A ; 105(33): 12028-33, 2008 Aug 19.
Artigo em Inglês | MEDLINE | ID: mdl-18685102

RESUMO

In mammals, two homologous cytosolic regulatory proteins, iron regulatory protein 1 (also known as IRP1 and Aco1) and iron regulatory protein 2 (also known as IRP2 and Ireb2), sense cytosolic iron levels and posttranscriptionally regulate iron metabolism genes, including transferrin receptor 1 (TfR1) and ferritin H and L subunits, by binding to iron-responsive elements (IREs) within target transcripts. Mice that lack IRP2 develop microcytic anemia and neurodegeneration associated with functional cellular iron depletion caused by low TfR1 and high ferritin expression. IRP1 knockout (IRP1(-/-)) animals do not significantly misregulate iron metabolism, partly because IRP1 is an iron-sulfur protein that functions mainly as a cytosolic aconitase in mammalian tissues and IRP2 activity increases to compensate for loss of the IRE binding form of IRP1. The neurodegenerative disease of IRP2(-/-) animals progresses slowly as the animals age. In this study, we fed IRP2(-/-) mice a diet supplemented with a stable nitroxide, Tempol, and showed that the progression of neuromuscular impairment was markedly attenuated. In cell lines derived from IRP2(-/-) animals, and in the cerebellum, brainstem, and forebrain of animals maintained on the Tempol diet, IRP1 was converted from a cytosolic aconitase to an IRE binding protein that stabilized the TfR1 transcript and repressed ferritin synthesis. We suggest that Tempol protected IRP2(-/-) mice by disassembling the cytosolic iron-sulfur cluster of IRP1 and activating IRE binding activity, which stabilized the TfR1 transcript, repressed ferritin synthesis, and partially restored normal cellular iron homeostasis in the brain.


Assuntos
Proteína 2 Reguladora do Ferro/deficiência , Proteína 2 Reguladora do Ferro/metabolismo , Doenças Neurodegenerativas/metabolismo , Doenças Neurodegenerativas/prevenção & controle , Animais , Linhagem Celular , Óxidos N-Cíclicos/química , Óxidos N-Cíclicos/farmacologia , Progressão da Doença , Ativação Enzimática , Humanos , Proteína 1 Reguladora do Ferro/metabolismo , Proteína 2 Reguladora do Ferro/genética , Camundongos , Camundongos Knockout , Estrutura Molecular , Doenças Neurodegenerativas/genética , Doenças Neurodegenerativas/patologia , Ligação Proteica , Receptores da Transferrina/metabolismo , Marcadores de Spin
16.
Trends Genet ; 24(8): 398-407, 2008 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-18606475

RESUMO

Iron-sulfur (Fe-S) clusters are essential for numerous biological processes, including mitochondrial respiratory chain activity and various other enzymatic and regulatory functions. Human Fe-S cluster assembly proteins are frequently encoded by single genes, and inherited defects in some of these genes cause disease. Recently, the spectrum of diseases attributable to abnormal Fe-S cluster biogenesis has extended beyond Friedreich ataxia to include a sideroblastic anemia with deficiency of glutaredoxin 5 and a myopathy associated with a deficiency of a Fe-S cluster assembly scaffold protein, ISCU. Mutations within other mammalian Fe-S cluster assembly genes could be causative for human diseases that manifest distinctive combinations of tissue-specific impairments. Thus, defects in the iron-sulfur cluster biogenesis pathway could underlie many human diseases.


Assuntos
Proteínas Ferro-Enxofre/biossíntese , Proteínas Ferro-Enxofre/genética , Transportadores de Cassetes de Ligação de ATP/genética , Anemia Sideroblástica/genética , Ataxia de Friedreich/genética , Glutarredoxinas/genética , Humanos , Proteínas de Ligação ao Ferro/genética , Modelos Biológicos , Doenças Musculares/genética , Mutação , Frataxina
17.
Am J Hum Genet ; 82(3): 652-60, 2008 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-18304497

RESUMO

A myopathy with severe exercise intolerance and myoglobinuria has been described in patients from northern Sweden, with associated deficiencies of succinate dehydrogenase and aconitase in skeletal muscle. We identified the gene for the iron-sulfur cluster scaffold protein ISCU as a candidate within a region of shared homozygosity among patients with this disease. We found a single mutation in ISCU that likely strengthens a weak splice acceptor site, with consequent exon retention. A marked reduction of ISCU mRNA and mitochondrial ISCU protein in patient muscle was associated with a decrease in the iron regulatory protein IRP1 and intracellular iron overload in skeletal muscle, consistent with a muscle-specific alteration of iron homeostasis in this disease. ISCU interacts with the Friedreich ataxia gene product frataxin in iron-sulfur cluster biosynthesis. Our results therefore extend the range of known human diseases that are caused by defects in iron-sulfur cluster biogenesis.


Assuntos
Tolerância ao Exercício/genética , Proteínas Ferro-Enxofre/genética , Miopatias Mitocondriais/genética , Sítios de Splice de RNA/genética , Aconitato Hidratase/deficiência , Adulto , Idoso , Sequência de Aminoácidos , Sequência de Bases , Análise Mutacional de DNA , Homozigoto , Humanos , Mitocôndrias/enzimologia , Miopatias Mitocondriais/enzimologia , Dados de Sequência Molecular , Mutação , Linhagem , Polimorfismo de Nucleotídeo Único , RNA Mensageiro/metabolismo , Succinato Desidrogenase/deficiência , Suécia
18.
Biometals ; 20(3-4): 549-64, 2007 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-17205209

RESUMO

Iron and citrate are essential for the metabolism of most organisms, and regulation of iron and citrate biology at both the cellular and systemic levels is critical for normal physiology and survival. Mitochondrial and cytosolic aconitases catalyze the interconversion of citrate and isocitrate, and aconitase activities are affected by iron levels, oxidative stress and by the status of the Fe-S cluster biogenesis apparatus. Assembly and disassembly of Fe-S clusters is a key process not only in regulating the enzymatic activity of mitochondrial aconitase in the citric acid cycle, but also in controlling the iron sensing and RNA binding activities of cytosolic aconitase (also known as iron regulatory protein IRP1). This review discusses the central role of aconitases in intermediary metabolism and explores how iron homeostasis and Fe-S cluster biogenesis regulate the Fe-S cluster switch and modulate intracellular citrate flux.


Assuntos
Aconitato Hidratase/metabolismo , Ácido Cítrico/metabolismo , Proteínas Ferro-Enxofre/metabolismo , Ferro/metabolismo , Aconitato Hidratase/antagonistas & inibidores , Animais , Linhagem Celular , Quelantes/metabolismo , Citoplasma/metabolismo , Metabolismo Energético , Humanos , Mitocôndrias/metabolismo , Estresse Oxidativo
19.
Cell Metab ; 3(3): 199-210, 2006 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-16517407

RESUMO

Iron-sulfur (Fe-S) clusters are required for the functions of mitochondrial aconitase, mammalian iron regulatory protein 1, and many other proteins in multiple subcellular compartments. Recent studies in Saccharomyces cerevisiae indicated that Fe-S cluster biogenesis also has an important role in mitochondrial iron homeostasis. Here we report the functional analysis of the mitochondrial and cytosolic isoforms of the human Fe-S cluster scaffold protein, ISCU. Suppression of human ISCU by RNAi not only inactivated mitochondrial and cytosolic aconitases in a compartment-specific manner but also inappropriately activated the iron regulatory proteins and disrupted intracellular iron homeostasis. Furthermore, endogenous ISCU levels were suppressed by iron deprivation. These results provide evidence for a coordinated response to iron deficiency that includes activation of iron uptake, redistribution of intracellular iron, and decreased utilization of iron in Fe-S proteins.


Assuntos
Citosol/metabolismo , Homeostase , Proteínas Ferro-Enxofre/metabolismo , Ferro/metabolismo , Mitocôndrias/metabolismo , Aconitato Hidratase/metabolismo , Células Cultivadas , Regulação da Expressão Gênica , Inativação Gênica , Células HeLa , Humanos , Proteína 1 Reguladora do Ferro/metabolismo , Proteína 2 Reguladora do Ferro/metabolismo , Modelos Biológicos , Ligação Proteica , Isoformas de Proteínas/metabolismo , RNA Interferente Pequeno/genética , Elementos de Resposta
20.
J Biol Chem ; 281(18): 12344-51, 2006 May 05.
Artigo em Inglês | MEDLINE | ID: mdl-16527810

RESUMO

Iron-sulfur clusters are prosthetic groups composed of sulfur and iron that are found in respiratory chain complexes and numerous enzymes. Iron-sulfur clusters are synthesized in a multistep process that utilizes cysteine desulfurases, scaffold proteins, chaperones, and iron donors. Assembly of iron-sulfur clusters occurs in the mitochondrial matrix of mammalian cells, but cytosolic isoforms of three major mammalian iron-sulfur cluster (ISC) assembly components have been found, raising the possibility that de novo iron-sulfur cluster biogenesis also occurs in cytosol. The human cysteine desulfurase, ISCS, has two isoforms, one of which targets to the mitochondria, whereas the other less abundant form is cytosolic and nuclear. The open-reading frame of cytosolic mammalian ISCS begins at the second AUG of the transcript and lacks mitochondrial targeting information. Yeast complementation experiments have suggested that the human cytosolic ISCS isoform (c-ISCS) cannot be functional. To evaluate function of c-ISCS, we overexpressed the human cytosolic ISCS in yeast Pichia pastoris and showed that the cytosolic form of ISCS is an active cysteine desulfurase that covalently binds 35S acquired from desulfuration of radiolabeled cysteine. Human cytosolic ISCS dimerized as efficiently as bacterial ISCS and formed a complex in vitro with overexpressed cytosolic human ISCU. When incubated with iron regulatory protein 1, cysteine, and iron, the cytosolic forms of ISCS and ISCU facilitated efficient formation of a [4Fe-4S] cluster on IRP1. Thus, the cytosolic form of ISCS is a functional cysteine desulfurase that can collaborate with cytosolic ISCU to promote de novo iron-sulfur cluster formation.


Assuntos
Liases de Carbono-Enxofre/fisiologia , Citosol/enzimologia , Proteínas de Escherichia coli/fisiologia , Proteínas Ferro-Enxofre/química , Aconitato Hidratase/química , Sequência de Aminoácidos , Citosol/metabolismo , Transporte de Elétrons , Escherichia coli/metabolismo , Humanos , Ferro/metabolismo , Proteínas Ferro-Enxofre/fisiologia , Dados de Sequência Molecular , Pichia/metabolismo , Homologia de Sequência de Aminoácidos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...