Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Am J Physiol Renal Physiol ; 322(1): F89-F103, 2022 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-34843656

RESUMO

Chronic kidney disease involves disturbances in iron metabolism including anemia caused by insufficient erythropoietin (EPO) production. However, underlying mechanisms responsible for the dysregulation of cellular iron metabolism are incompletely defined. Using the unilateral ureteral obstruction (UUO) model in Irp1+/+ and Irp1-/- mice, we asked if iron regulatory proteins (IRPs), the central regulators of cellular iron metabolism and suppressors of EPO production, contribute to the etiology of anemia in kidney failure. We identified a significant reduction in IRP protein level and RNA binding activity that associates with a loss of the iron uptake protein transferrin receptor 1 (TfR1), increased expression of the iron storage protein subunits H- and L-ferritin, and a low but overall variable level of stainable iron in the obstructed kidney. This reduction in IRP RNA binding activity and ferritin RNA levels suggests the concomitant rise in ferritin expression and iron content in kidney failure is IRP dependent. In contrast, the reduction in the Epo mRNA level in the obstructed kidney was not rescued by genetic ablation of IRP1, suggesting disruption of normal hypoxia-inducible factor (HIF)-2α regulation. Furthermore, reduced expression of some HIF-α target genes in UUO occurred in the face of increased expression of HIF-α proteins and prolyl hydroxylases 2 and 1, the latter of which is not known to be HIF-α mediated. Our results suggest that the IRP system drives changes in cellular iron metabolism that are associated with kidney failure in UUO but that the impact of IRPs on EPO production is overridden by disrupted hypoxia signaling.NEW & NOTEWORTHY This study demonstrates that iron metabolism and hypoxia signaling are dysregulated in unilateral obstructive nephropathy. Expression of iron regulatory proteins (IRPs), central regulators of cellular iron metabolism, and the iron uptake (transferrin receptor 1) and storage (ferritins) proteins they target is strongly altered. This suggests a role of IRPs in previously observed changes in iron metabolism in progressive renal disease. Hypoxia signaling is disrupted and appeared to dominate the action of IRP1 in controlling erythropoietin expression.


Assuntos
Anemia/etiologia , Ferro/metabolismo , Rim/metabolismo , Insuficiência Renal/etiologia , Obstrução Ureteral/complicações , Anemia/metabolismo , Anemia/patologia , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Hipóxia Celular , Modelos Animais de Doenças , Eritropoetina/genética , Eritropoetina/metabolismo , Ferritinas/genética , Ferritinas/metabolismo , Fibrose , Subunidade alfa do Fator 1 Induzível por Hipóxia/genética , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Prolina Dioxigenases do Fator Induzível por Hipóxia/genética , Prolina Dioxigenases do Fator Induzível por Hipóxia/metabolismo , Proteína 1 Reguladora do Ferro/genética , Proteína 1 Reguladora do Ferro/metabolismo , Rim/patologia , Masculino , Camundongos Endogâmicos C57BL , Camundongos Knockout , Pró-Colágeno-Prolina Dioxigenase/genética , Pró-Colágeno-Prolina Dioxigenase/metabolismo , Receptores da Transferrina/genética , Receptores da Transferrina/metabolismo , Insuficiência Renal/metabolismo , Insuficiência Renal/patologia , Obstrução Ureteral/metabolismo , Obstrução Ureteral/patologia
2.
J Biol Chem ; 292(38): 15976-15989, 2017 09 22.
Artigo em Inglês | MEDLINE | ID: mdl-28768766

RESUMO

Iron-regulatory protein 1 (IRP1) belongs to a family of RNA-binding proteins that modulate metazoan iron metabolism. Multiple mechanisms are employed to control the action of IRP1 in dictating changes in the uptake and metabolic fate of iron. Inactivation of IRP1 RNA binding by iron primarily involves insertion of a [4Fe-4S] cluster by the cytosolic iron-sulfur cluster assembly (CIA) system, converting it into cytosolic aconitase (c-acon), but can also involve iron-mediated degradation of IRP1 by the E3 ligase FBXL5 that also targets IRP2. How CIA and FBXL5 collaborate to maintain cellular iron homeostasis through IRP1 and other pathways is poorly understood. Because impaired Fe-S cluster biogenesis associates with human disease, we determined the importance of FBXL5 for regulating IRP1 when CIA is impaired. Suppression of FBXL5 expression coupled with induction of an IRP1 mutant (IRP13C>3S) that cannot insert the Fe-S cluster, or along with knockdown of the CIA factors NUBP2 or FAM96A, reduced cell viability. Iron supplementation reversed this growth defect and was associated with FBXL5-dependent polyubiquitination of IRP1. Phosphorylation of IRP1 at Ser-138 increased when CIA was inhibited and was required for iron rescue. Impaired CIA activity, as noted by reduced c-acon activity, was associated with enhanced FBXL5 expression and a concomitant reduction in IRP1 and IRP2 protein level and RNA-binding activity. Conversely, expression of either IRP induced FBXL5 protein level, demonstrating a negative feedback loop limiting excessive accumulation of iron-response element RNA-binding activity, whose disruption reduces cell growth. We conclude that a regulatory circuit involving FBXL5 and CIA acts through both IRPs to control iron metabolism and promote optimal cell growth.


Assuntos
Proteínas F-Box/metabolismo , Proteína 1 Reguladora do Ferro/metabolismo , Ferro/metabolismo , Complexos Ubiquitina-Proteína Ligase/metabolismo , Proteínas F-Box/genética , Ferritinas/metabolismo , Técnicas de Silenciamento de Genes , Células HEK293 , Humanos , Proteína 1 Reguladora do Ferro/química , Proteína 2 Reguladora do Ferro/metabolismo , Fosforilação , Complexo de Endopeptidases do Proteassoma/metabolismo , Proteólise , RNA/metabolismo , Serina/metabolismo , Enxofre/metabolismo , Complexos Ubiquitina-Proteína Ligase/deficiência , Complexos Ubiquitina-Proteína Ligase/genética , Ubiquitinação
3.
J Biol Chem ; 289(11): 7835-43, 2014 Mar 14.
Artigo em Inglês | MEDLINE | ID: mdl-24509859

RESUMO

Mitochondrial iron is essential for the biosynthesis of heme and iron-sulfur ([Fe-S]) clusters in mammalian cells. In developing erythrocytes, iron is imported into the mitochondria by MFRN1 (mitoferrin-1, SLC25A37). Although loss of MFRN1 in zebrafish and mice leads to profound anemia, mutant animals showed no overt signs of porphyria, suggesting that mitochondrial iron deficiency does not result in an accumulation of protoporphyrins. Here, we developed a gene trap model to provide in vitro and in vivo evidence that iron regulatory protein-1 (IRP1) inhibits protoporphyrin accumulation. Mfrn1(+/gt);Irp1(-/-) erythroid cells exhibit a significant increase in protoporphyrin levels. IRP1 attenuates protoporphyrin biosynthesis by binding to the 5'-iron response element (IRE) of alas2 mRNA, inhibiting its translation. Ectopic expression of alas2 harboring a mutant IRE, preventing IRP1 binding, in Mfrn1(gt/gt) cells mimics Irp1 deficiency. Together, our data support a model whereby impaired mitochondrial [Fe-S] cluster biogenesis in Mfrn1(gt/gt) cells results in elevated IRP1 RNA-binding that attenuates ALAS2 mRNA translation and protoporphyrin accumulation.


Assuntos
5-Aminolevulinato Sintetase/metabolismo , Regulação da Expressão Gênica , Proteína 1 Reguladora do Ferro/metabolismo , Proteínas de Membrana Transportadoras/metabolismo , Porfirias/metabolismo , Animais , Blastocisto/citologia , Diferenciação Celular , Linhagem Celular Tumoral , Feminino , Genótipo , Células HEK293 , Heme/química , Humanos , Ferro/química , Proteínas Ferro-Enxofre/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Biossíntese de Proteínas , Protoporfirinas/metabolismo , Peixe-Zebra
4.
ACS Chem Biol ; 11(1): 193-9, 2016 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-26535807

RESUMO

Collagen is the most abundant protein in animals. Its overproduction is associated with fibrosis and cancer metastasis. The stability of collagen relies on post-translational modifications, the most prevalent being the hydroxylation of collagen strands by collagen prolyl 4-hydroxylases (CP4Hs). Catalysis by CP4Hs enlists an iron cofactor to convert proline residues to 4-hydroxyproline residues, which are essential for the conformational stability of mature collagen. Ethyl 3,4-dihydroxybenzoate (EDHB) is commonly used as a "P4H" inhibitor in cells, but suffers from low potency, poor selectivity, and off-target effects that cause iron deficiency. Dicarboxylates of 2,2'-bipyridine are among the most potent known CP4H inhibitors but suffer from a high affinity for free iron. A screen of biheteroaryl compounds revealed that replacing one pyridyl group with a thiazole moiety retains potency and enhances selectivity. A diester of 2-(5-carboxythiazol-2-yl)pyridine-5-carboxylic acid is bioavailable to human cells and inhibits collagen biosynthesis at concentrations that neither cause general toxicity nor disrupt iron homeostasis. These data anoint a potent and selective probe for CP4H and a potential lead for the development of a new class of antifibrotic and antimetastatic agents.


Assuntos
Ácidos Carboxílicos/farmacologia , Pró-Colágeno-Prolina Dioxigenase/antagonistas & inibidores , Antineoplásicos/química , Antineoplásicos/farmacologia , Antineoplásicos/toxicidade , Ácidos Carboxílicos/química , Ácidos Carboxílicos/toxicidade , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Ativação Enzimática/efeitos dos fármacos , Inibidores Enzimáticos/síntese química , Inibidores Enzimáticos/química , Inibidores Enzimáticos/farmacologia , Humanos , Pró-Colágeno-Prolina Dioxigenase/química , Tiazóis/química , Tiazóis/farmacologia
5.
Sci Signal ; 8(372): ra34, 2015 Apr 14.
Artigo em Inglês | MEDLINE | ID: mdl-25872869

RESUMO

In multicellular organisms, the mechanisms by which diverse cell types acquire distinct amino acids and how cellular function adapts to their availability are fundamental questions in biology. We found that increased neutral essential amino acid (NEAA) uptake was a critical component of erythropoiesis. As red blood cells matured, expression of the amino acid transporter gene Lat3 increased, which increased NEAA import. Inadequate NEAA uptake by pharmacologic inhibition or RNAi-mediated knockdown of LAT3 triggered a specific reduction in hemoglobin production in zebrafish embryos and murine erythroid cells through the mTORC1 (mammalian target of rapamycin complex 1)/4E-BP (eukaryotic translation initiation factor 4E-binding protein) pathway. CRISPR-mediated deletion of members of the 4E-BP family in murine erythroid cells rendered them resistant to mTORC1 and LAT3 inhibition and restored hemoglobin production. These results identify a developmental role for LAT3 in red blood cells and demonstrate that mTORC1 serves as a homeostatic sensor that couples hemoglobin production at the translational level to sufficient uptake of NEAAs, particularly L-leucine.


Assuntos
Proteínas de Transporte/metabolismo , Fatores de Iniciação em Eucariotos/metabolismo , Hemoglobinas/metabolismo , Leucina/metabolismo , Complexos Multiproteicos/metabolismo , Fosfoproteínas/metabolismo , Serina-Treonina Quinases TOR/metabolismo , Proteínas Adaptadoras de Transdução de Sinal , Sistemas de Transporte de Aminoácidos Básicos/genética , Sistemas de Transporte de Aminoácidos Básicos/metabolismo , Animais , Animais Geneticamente Modificados , Sistemas CRISPR-Cas , Proteínas de Transporte/genética , Proteínas de Ciclo Celular , Linhagem Celular Tumoral , Células Cultivadas , Embrião de Mamíferos/irrigação sanguínea , Embrião de Mamíferos/embriologia , Embrião de Mamíferos/metabolismo , Embrião não Mamífero/embriologia , Embrião não Mamífero/metabolismo , Células Eritroides/metabolismo , Eritropoese/genética , Fatores de Iniciação em Eucariotos/genética , Regulação da Expressão Gênica no Desenvolvimento , Células HEK293 , Hemoglobinas/genética , Humanos , Immunoblotting , Alvo Mecanístico do Complexo 1 de Rapamicina , Camundongos , Microscopia Confocal , Complexos Multiproteicos/genética , Fosfoproteínas/genética , Interferência de RNA , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução de Sinais/genética , Serina-Treonina Quinases TOR/genética , Peixe-Zebra
6.
Cell Metab ; 17(2): 282-90, 2013 Feb 05.
Artigo em Inglês | MEDLINE | ID: mdl-23395174

RESUMO

Red blood cell production is a finely tuned process that requires coordinated oxygen- and iron-dependent regulation of cell differentiation and iron metabolism. Here, we show that translational regulation of hypoxia-inducible factor 2α (HIF-2α) synthesis by iron regulatory protein 1 (IRP1) is critical for controlling erythrocyte number. IRP1-null (Irp1(-/-)) mice display a marked transient polycythemia. HIF-2α messenger RNA (mRNA) is derepressed in kidneys of Irp1(-/-) mice but not in kidneys of Irp2(-/-) mice, leading to increased renal erythropoietin (Epo) mRNA and inappropriately elevated serum Epo levels. Expression of the iron transport genes DCytb, Dmt1, and ferroportin, as well as other HIF-2α targets, is enhanced in Irp1(-/-) duodenum. Analysis of mRNA translation state in the liver revealed IRP1-dependent dysregulation of HIF-2α mRNA translation, whereas IRP2 deficiency derepressed translation of all other known 5' iron response element (IRE)-containing mRNAs expressed in the liver. These results uncover separable physiological roles of each IRP and identify IRP1 as a therapeutic target for manipulating HIF-2α action in hematologic, oncologic, and other disorders.


Assuntos
Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Eritropoese , Proteína 1 Reguladora do Ferro/metabolismo , Ferro/metabolismo , Oxigênio/metabolismo , Transdução de Sinais , Absorção , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Duodeno/metabolismo , Duodeno/patologia , Células Eritroides/metabolismo , Células Precursoras Eritroides/metabolismo , Eritropoetina/sangue , Regulação da Expressão Gênica , Hematopoese Extramedular , Proteína 1 Reguladora do Ferro/deficiência , Camundongos , Policitemia/sangue , Policitemia/patologia , Biossíntese de Proteínas , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Baço/metabolismo
7.
J Biol Chem ; 284(19): 12701-9, 2009 May 08.
Artigo em Inglês | MEDLINE | ID: mdl-19269970

RESUMO

Iron-sulfur cluster-dependent interconversion of iron regulatory protein 1 (IRP1) between its RNA binding and cytosolic aconitase (c-acon) forms controls vertebrate iron homeostasis. Cluster removal from c-acon is thought to include oxidative demetallation as a required step, but little else is understood about the process of conversion to IRP1. In comparison with c-acon(WT), Ser(138) phosphomimetic mutants of c-acon contain an unstable [4Fe-4S] cluster and were used as tools to further define the pathway(s) of iron-sulfur cluster disassembly. Under anaerobic conditions cluster insertion into purified IRP1(S138E) and cluster loss on treatment with NO regulated aconitase and RNA binding activity over a similar range as observed for IRP1(WT). However, activation of RNA binding of c-acon(S138E) was an order of magnitude more sensitive to NO than for c-acon(WT). Consistent with this, an altered set point between RNA-binding and aconitase forms was observed for IRP1(S138E) when expressed in HEK cells. Active c-acon(S138E) could only accumulate under hypoxic conditions, suggesting enhanced cluster disassembly in normoxia. Cluster disassembly mechanisms were further probed by determining the impact of iron chelation on acon activity. Unexpectedly EDTA rapidly inhibited c-acon(S138E) activity without affecting c-acon(WT). Additional chelator experiments suggested that cluster loss can be initiated in c-acon(S138E) through a spontaneous nonoxidative demetallation process. Taken together, our results support a model wherein Ser(138) phosphorylation sensitizes IRP1/c-acon to decreased iron availability by allowing the [4Fe-4S](2+) cluster to cycle with [3Fe-4S](0) in the absence of cluster perturbants, indicating that regulation can be initiated merely by changes in iron availability.


Assuntos
Aconitato Hidratase/metabolismo , Proteína 1 Reguladora do Ferro/metabolismo , Proteínas Ferro-Enxofre/metabolismo , Ferro/metabolismo , Serina/metabolismo , Enxofre/metabolismo , Citosol/enzimologia , Espectroscopia de Ressonância de Spin Eletrônica , Humanos , Hipóxia , Quelantes de Ferro , Proteína 1 Reguladora do Ferro/genética , Proteínas Ferro-Enxofre/genética , Rim/citologia , Rim/metabolismo , Mutagênese Sítio-Dirigida , Mutação , Óxido Nítrico/metabolismo , Fosforilação , RNA/metabolismo
8.
EMBO J ; 25(3): 544-53, 2006 Feb 08.
Artigo em Inglês | MEDLINE | ID: mdl-16424901

RESUMO

The generally accepted role of iron-regulatory protein 1 (IRP1) in orchestrating the fate of iron-regulated mRNAs depends on the interconversion of its cytosolic aconitase and RNA-binding forms through assembly/disassembly of its Fe-S cluster, without altering protein abundance. Here, we show that IRP1 protein abundance can be iron-regulated. Modulation of IRP1 abundance by iron did not require assembly of the Fe-S cluster, since a mutant with all cluster-ligating cysteines mutated to serine underwent iron-induced protein degradation. Phosphorylation of IRP1 at S138 favored the RNA-binding form and promoted iron-dependent degradation. However, phosphorylation at S138 was not required for degradation. Further, degradation of an S138 phosphomimetic mutant was not blocked by mutation of cluster-ligating cysteines. These findings were confirmed in mouse models with genetic defects in cytosolic Fe-S cluster assembly/disassembly. IRP1 RNA-binding activity was primarily regulated by IRP1 degradation in these animals. Our results reveal a mechanism for regulating IRP1 action relevant to the control of iron homeostasis during cell proliferation, inflammation, and in response to diseases altering cytosolic Fe-S cluster assembly or disassembly.


Assuntos
Proteína 1 Reguladora do Ferro/metabolismo , Ferro/fisiologia , Animais , Linhagem Celular , Hemina/metabolismo , Humanos , Proteína 1 Reguladora do Ferro/genética , Camundongos , Camundongos Knockout , Mutação , Fosforilação , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Proteínas de Ligação a RNA/genética , Proteínas de Ligação a RNA/metabolismo , Serina/metabolismo
9.
Hum Mol Genet ; 15(6): 953-64, 2006 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-16467350

RESUMO

Proteins with iron-sulfur (Fe-S) clusters participate in multiple metabolic pathways throughout the cell. The mitochondrial ABC half-transporter Abcb7, which is mutated in X-linked sideroblastic anemia with ataxia in humans, is a functional ortholog of yeast Atm1p and is predicted to export a mitochondrially derived metabolite required for cytosolic Fe-S cluster assembly. Using an inducible Cre/loxP system to delete exons 9 and 10 of the Abcb7 gene, we examined the phenotype of mice deficient in Abcb7. We found that Abcb7 was essential in extra-embryonic tissues early in gestation and that the mutant allele exhibits an X-linked parent-of-origin lethality effect. Furthermore, using X-chromosome inactivation assays and tissue-specific deletions, Abcb7 was found to be essential for the development and function of numerous other cell types and tissues. A notable exception to this was liver, where loss of Abcb7 impaired cytosolic Fe-S cluster assembly but was not lethal. In this situation, control of iron regulatory protein 1, a key cytosolic modulator of iron metabolism, which is responsive to the availability of cytosolic Fe-S clusters, was impaired and contributed to the dysregulation of hepatocyte iron metabolism. Altogether, these studies demonstrate the essential nature of Abcb7 in mammals and further substantiate a central role for mitochondria in the biogenesis of cytosolic Fe-S proteins.


Assuntos
Transportadores de Cassetes de Ligação de ATP/fisiologia , Citosol/metabolismo , Proteínas Ferro-Enxofre/biossíntese , Proteínas Mitocondriais/fisiologia , Transportadores de Cassetes de Ligação de ATP/genética , Animais , Linhagem Celular , Linhagem da Célula/genética , Genes Letais , Hepatócitos/metabolismo , Hepatócitos/ultraestrutura , Ferro/metabolismo , Proteína 1 Reguladora do Ferro/metabolismo , Proteína 2 Reguladora do Ferro/metabolismo , Masculino , Camundongos , Proteínas Mitocondriais/deficiência , Proteínas Mitocondriais/genética , Cromossomo X/genética
10.
Inorg Chem ; 41(4): 669-77, 2002 Feb 25.
Artigo em Inglês | MEDLINE | ID: mdl-11849065

RESUMO

A 6000-fold rate enhancement has been observed for the hydrolysis of bis(p-nitrophenyl)phosphate (BNPP) in the presence of 0.2 mM Cu(i-Pr(3)[9]aneN(3))(2+) at pH 9.2 and 50 degrees C. In a direct comparison, the rate of hydrolysis of BNPP is accelerated at least 60-fold over the previously reported catalyst Cu([9]aneN(3))(2+). As observed for Cu([9]aneN(3))(2+), hydrolysis is selective for diesters over monoesters. Hydrolysis of BNPP by Cu(i-Pr(3)[9]aneN(3))(2+) is catalytic, exhibiting both rate enhancement and turnover. The reaction is inhibited by both p-nitrophenyl phosphate and inorganic phosphate. The reaction is first-order in substrate and half-order in metal complex, with a k(1.5) of 0.060 +/- 0.004 M(-1/2) s(-1) at 50 degrees C. The temperature dependence of the rate constant results in a calculated activation enthalpy (Delta H(++) of 51 +/- 2 kJ mol(-1) and activation entropy (Delta S(++)) of -110 +/- 6 J mol(-1) K(-1). The kinetic pK(a) of 7.8 +/- 0.2 is close to the thermodynamic pK(a) of 7.9 +/- 0.2, consistent with deprotonation of a coordinated water molecule in the active form of the catalyst. The active catalyst [Cu(i-Pr(3)[9]aneN(3))(OH)(OH(2))](+) is in equilibrium with an inactive dimer, and the formation constant for this dimer is between 216 and 1394 M(-1) at pH 9.2 and 50 degrees C. Temperature dependence of the dimer formation constant K(f) indicates an endothermic enthalpy of formation for the dimer of 27 +/- 3 kJ mol(-1). The time course of anaerobic DNA cleavage by Cu(i-Pr(3)[9]aneN(3))(2+) is presented over a wide range of concentrations at pH 7.8 at 50 degrees C. The concentration dependence of DNA cleavage by Cu([9]aneN(3))(2+) and Cu(i-Pr(3)[9]aneN(3))(2+) reveals a maximum cleavage efficiency at sub-micromolar concentrations of cleavage agent. DNA cleavage by Cu(i-Pr(3)[9]aneN(3))(2+) is twice as efficient at pH 7.8 as at pH 7.2.


Assuntos
Cobre/química , DNA/química , Compostos Organometálicos/química , Catálise , Eletroforese em Gel de Poliacrilamida , Concentração de Íons de Hidrogênio , Hidrólise , Cinética , Estrutura Molecular , Nitrofenóis/química , Compostos Organometálicos/metabolismo , Plasmídeos , Temperatura
11.
Proc Natl Acad Sci U S A ; 101(30): 10907-12, 2004 Jul 27.
Artigo em Inglês | MEDLINE | ID: mdl-15263083

RESUMO

Iron-regulatory protein 1 (IRP1) is a dual-function protein with mutually exclusive roles as a posttranscriptional regulator of animal-cell iron metabolism or as the cytosolic isoform of the iron-sulfur enzyme aconitase (c-acon). Much effort has focused on the role of IRP1 in posttranscriptional gene regulation and in factors that influence its interconversion with c-acon, but little is known about the metabolic function and regulation of c-acon. The role of PKC-dependent phosphorylation of S711 on IRP1/c-acon function was examined. Phosphorylation state-specific antibodies revealed that S711 is phosphorylated by PKC in vitro and in human embryonic kidney cells treated with a PKC activator. In aco1 yeast, the phosphomimetic mutants S711D and S711E exhibited severely impaired aconitase function, whereas S711A and S711T were unaffected relative to the WT protein. Aconitase activity in yeast extracts displayed a similar pattern when assayed for capacity to convert citrate to isocitrate: WT, S711A, and S711T were active, but S711D and S711E activity was undetectable. In contrast, when measured by the conversion of isocitrate to cis-aconitate, S711D and S711E displayed substantial activity, indicating that phosphorylation impairs the citrate but not isocitrate mode of aconitase function. This possibility was confirmed in vivo by demonstrating that S711D and S711E specifically antagonized the requirement for isocitrate in two metabolic scenarios. Iron-responsive element RNA-binding affinity was unaffected by S711 mutations. Our results show that S711 is a target of phosphorylation capable of conferring distinct effects on c-acon function potentially dictating changes in cytosolic citrate/isocitrate metabolism.


Assuntos
Citratos/metabolismo , Proteína 1 Reguladora do Ferro/antagonistas & inibidores , Isocitratos/metabolismo , Serina , Substituição de Aminoácidos , Citosol/enzimologia , Proteína 1 Reguladora do Ferro/química , Proteína 1 Reguladora do Ferro/genética , Cinética , Mutagênese Sítio-Dirigida , Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/antagonistas & inibidores , Proteínas de Saccharomyces cerevisiae/química , Proteínas de Saccharomyces cerevisiae/genética
12.
J Cell Physiol ; 190(2): 218-26, 2002 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-11807826

RESUMO

Iron regulatory proteins (IRPs), the cytosolic proteins involved in the maintenance of cellular iron homeostasis, bind to stem loop structures found in the mRNA of key proteins involved iron uptake, storage, and metabolism and regulate the expression of these proteins in response to changes in cellular iron needs. We have shown previously that HFE-expressing fWTHFE/tTA HeLa cells have slightly increased transferrin receptor levels and dramatically reduced ferritin levels when compared to the same clonal cell line without HFE (Gross et al., 1998, J Biol Chem 273:22068-22074). While HFE does not alter transferrin receptor trafficking or non-transferrin mediated iron uptake, it does specifically reduce (55)Fe uptake from transferrin (Roy et al., 1999, J Biol Chem 274:9022-9028). In this report, we show that IRP RNA binding activity is increased by up to 5-fold in HFE-expressing cells through the activation of both IRP isoforms. Calcein measurements show a 45% decrease in the intracellular labile iron pool in HFE-expressing cells, which is in keeping with the IRP activation. These results all point to the direct effect of the interaction of HFE with transferrin receptor in lowering the intracellular labile iron pool and establishing a new set point for iron regulation within the cell.


Assuntos
Antígenos HLA/metabolismo , Antígenos de Histocompatibilidade Classe I/metabolismo , Proteínas Ferro-Enxofre/metabolismo , Ferro/metabolismo , Proteínas de Membrana , Proteínas de Ligação a RNA/metabolismo , RNA/metabolismo , Ferritinas/antagonistas & inibidores , Ferritinas/biossíntese , Fluoresceínas , Corantes Fluorescentes , Células HeLa , Proteína da Hemocromatose , Humanos , Membranas Intracelulares/metabolismo , Proteína 1 Reguladora do Ferro , Proteína 2 Reguladora do Ferro , Proteínas Reguladoras de Ferro
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA