Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 104
Filtrar
1.
J Biol Chem ; 298(3): 101643, 2022 03.
Artigo em Inglês | MEDLINE | ID: mdl-35093382

RESUMO

Heme is a critical biomolecule that is synthesized in vivo by several organisms such as plants, animals, and bacteria. Reflecting the importance of this molecule, defects in heme biosynthesis underlie several blood disorders in humans. Aminolevulinic acid synthase (ALAS) initiates heme biosynthesis in α-proteobacteria and nonplant eukaryotes. Debilitating and painful diseases such as X-linked sideroblastic anemia and X-linked protoporphyria can result from one of more than 91 genetic mutations in the human erythroid-specific enzyme ALAS2. This review will focus on recent structure-based insights into human ALAS2 function in health and how it dysfunctions in disease. We will also discuss how certain genetic mutations potentially result in disease-causing structural perturbations. Furthermore, we use thermodynamic and structural information to hypothesize how the mutations affect the human ALAS2 structure and categorize some of the unique human ALAS2 mutations that do not respond to typical treatments, that have paradoxical in vitro activity, or that are highly intolerable to changes. Finally, we will examine where future structure-based insights into the family of ALA synthases are needed to develop additional enzyme therapeutics.


Assuntos
5-Aminolevulinato Sintetase , Anemia Sideroblástica , Doenças Genéticas Ligadas ao Cromossomo X , 5-Aminolevulinato Sintetase/química , 5-Aminolevulinato Sintetase/genética , 5-Aminolevulinato Sintetase/metabolismo , Ácido Aminolevulínico/química , Ácido Aminolevulínico/metabolismo , Anemia Sideroblástica/enzimologia , Anemia Sideroblástica/genética , Animais , Heme , Humanos , Relação Estrutura-Atividade
2.
Protein Expr Purif ; 183: 105860, 2021 07.
Artigo em Inglês | MEDLINE | ID: mdl-33689857

RESUMO

The ATP-binding cassette sub-family B member 7 (ABCB7) is a membrane transport protein located on the inner membrane of mitochondria, which could be involved in the transport of heme from the mitochondria to the cytosol. ABCB7 also plays a central role in the maturation of cytosolic iron-sulfur (Fe/S) cluster-containing proteins, and mutations can cause a series of mitochondrial defects. X-linked sideroblastic anemia and ataxia (XLSA-A) is a rare cause of early onset ataxia, which may be overlooked due to the usually mild asymptomatic anemia. The genetic defect has been identified as a mutation in the ABCB7 gene at Xq12-q13. Here, we report the expression, purification and the 2D projections derived from negatively stained electron micrographs of recombinant H. sapiens ABCB7 (hABCB7), paving the way from an atomic structure determination of ABCB7.


Assuntos
Transportadores de Cassetes de Ligação de ATP , Mutação , Transportadores de Cassetes de Ligação de ATP/biossíntese , Transportadores de Cassetes de Ligação de ATP/química , Transportadores de Cassetes de Ligação de ATP/genética , Transportadores de Cassetes de Ligação de ATP/isolamento & purificação , Anemia Sideroblástica/enzimologia , Anemia Sideroblástica/genética , Doenças Genéticas Ligadas ao Cromossomo X/enzimologia , Doenças Genéticas Ligadas ao Cromossomo X/genética , Humanos , Conformação Proteica , Proteínas Recombinantes/biossíntese , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/isolamento & purificação , Ataxias Espinocerebelares/enzimologia , Ataxias Espinocerebelares/genética
3.
Mol Genet Metab ; 128(3): 309-313, 2019 11.
Artigo em Inglês | MEDLINE | ID: mdl-31395332

RESUMO

Erythropoietic protoporphyria (EPP), the most common porphyria of childhood and the third most common porphyria of adulthood, is characterized clinically by painful, non-blistering cutaneous photosensitivity. Two distinct inheritance patterns involving mutations affecting genes that encode enzymes of the heme biosynthetic pathway underlie the clinical phenotype. Aminolevulinic acid synthase 2 (ALAS2), the rate limiting enzyme of the heme pathway in the erythron, is a therapeutic target in EPP because inhibiting enzyme function would reduce downstream production of protoporphyrin IX (PPIX), preventing accumulation of the toxic molecule and thereby ameliorating symptoms. Isoniazid (INH) is widely used for treatment of latent and active M. tuberculosis (TB). Sideroblastic anemia is observed in some patients taking INH, and studies have shown that this process is a consequence of inhibition of ALAS2 by INH. Based on these observations, we postulated that INH might have therapeutic activity in patients with EPP. We challenged this hypothesis in a murine model of EPP and showed that, after 4 weeks of treatment with INH, both plasma PPIX and hepatic PPIX were significantly reduced. Next, we tested the effect of INH on patients with EPP. After eight weeks, no significant difference in plasma or red cell PPIX was observed among the 15 patients enrolled in the study. These results demonstrate that while INH can lower PPIX in an animal model of EPP, the standard dose used to treat TB is insufficient to affect levels in humans.


Assuntos
5-Aminolevulinato Sintetase/antagonistas & inibidores , Isoniazida/uso terapêutico , Protoporfiria Eritropoética/tratamento farmacológico , Protoporfirinas/sangue , Anemia Sideroblástica/enzimologia , Animais , Modelos Animais de Doenças , Feminino , Humanos , Fígado/química , Fígado/efeitos dos fármacos , Masculino , Camundongos , Projetos Piloto , Estudo de Prova de Conceito , Protoporfiria Eritropoética/genética , Protoporfirinas/metabolismo
4.
Mol Genet Metab ; 128(3): 190-197, 2019 11.
Artigo em Inglês | MEDLINE | ID: mdl-30737140

RESUMO

Recently, new genes and molecular mechanisms have been identified in patients with porphyrias and sideroblastic anemias (SA). They all modulate either directly or indirectly the δ-aminolevulinic acid synthase (ALAS) activity. ALAS, is encoded by two genes: the erythroid-specific (ALAS2), and the ubiquitously expressed (ALAS1). In the liver, ALAS1 controls the rate-limiting step in the production of heme and hemoproteins that are rapidly turned over in response to metabolic needs. Several heme regulatory targets have been identified as regulators of ALAS1 activity: 1) transcriptional repression via a heme-responsive element, 2) post-transcriptional destabilization of ALAS1 mRNA, 3) post-translational inhibition via a heme regulatory motif, 4) direct inhibition of the activity of the enzyme and 5) breakdown of ALAS1 protein via heme-mediated induction of the protease Lon peptidase 1. In erythroid cells, ALAS2 is a gatekeeper of production of very large amounts of heme necessary for hemoglobin synthesis. The rate of ALAS2 synthesis is transiently increased during the period of active heme synthesis. Its gene expression is determined by trans-activation of nuclear factor GATA1, CACC box and NF-E2-binding sites in the promoter areas. ALAS2 mRNA translation is also regulated by the iron-responsive element (IRE)/iron regulatory proteins (IRP) binding system. In patients, ALAS enzyme activity is affected in most of the mutations causing non-syndromic SA and in several porphyrias. Decreased ALAS2 activity results either directly from loss-of-function ALAS2 mutations as seen in X-linked sideroblastic anemia (XLSA) or from defect in the availability of one of its two mitochondrial substrates: glycine in SLC25A38 mutations and succinyl CoA in GLRX5 mutations. Moreover, ALAS2 gain of function mutations is responsible for X-linked protoporphyria and increased ALAS1 activity lead to acute attacks of hepatic porphyrias. A missense dominant mutation in the Walker A motif of the ATPase binding site in the gene coding for the mitochondrial protein unfoldase CLPX also contributes to increasing ALAS and subsequently protoporphyrinemia. Altogether, these recent data on human ALAS have informed our understanding of porphyrias and sideroblastic anemias pathogeneses and may contribute to new therapeutic strategies.


Assuntos
5-Aminolevulinato Sintetase/genética , Ácido Aminolevulínico/metabolismo , Anemia Sideroblástica/genética , Regulação da Expressão Gênica , Porfirias/genética , 5-Aminolevulinato Sintetase/metabolismo , Anemia Sideroblástica/enzimologia , Animais , Sítios de Ligação , Fator de Transcrição GATA1/genética , Heme/biossíntese , Humanos , Fígado/metabolismo , Camundongos , Mutação de Sentido Incorreto , Porfirias/enzimologia , Regiões Promotoras Genéticas
5.
Mol Genet Metab ; 128(3): 342-351, 2019 11.
Artigo em Inglês | MEDLINE | ID: mdl-30660387

RESUMO

Non-syndromic microcytic congenital sideroblastic anemia (cSA) is predominantly caused by defective genes encoding for either ALAS2, the first enzyme of heme biosynthesis pathway or SLC25A38, the mitochondrial importer of glycine, an ALAS2 substrate. Herein we explored a new case of cSA with two mutations in GLRX5, a gene for which only two patients have been reported so far. The patient was a young female with biallelic compound heterozygous mutations in GLRX5 (p.Cys67Tyr and p.Met128Lys). Three-D structure analysis confirmed the involvement of Cys67 in the coordination of the [2Fe2S] cluster and suggested a potential role of Met128 in partner interactions. The protein-level of ferrochelatase, the terminal-enzyme of heme process, was increased both in patient-derived lymphoblastoid and CD34+ cells, however, its activity was drastically decreased. The activity of ALAS2 was found altered and possibly related to a defect in the biogenesis of its co-substrate, the succinyl-CoA. Thus, the patient exhibits both a very low ferrochelatase activity without any accumulation of porphyrins precursors in contrast to what is reported in erythropoietic protoporphyria with solely impaired ferrochelatase activity. A significant oxidative stress was evidenced by decreased reduced glutathione and aconitase activity, and increased MnSOD protein expression. This oxidative stress depleted and damaged mtDNA, decreased complex I and IV activities and depleted ATP content. Collectively, our study demonstrates the key role of GLRX5 in modulating ALAS2 and ferrochelatase activities and in maintaining mitochondrial function.


Assuntos
5-Aminolevulinato Sintetase/genética , Anemia Sideroblástica/genética , Ferroquelatase/genética , Doenças Genéticas Ligadas ao Cromossomo X/genética , Glutarredoxinas/genética , Heme/biossíntese , Mutação de Sentido Incorreto , 5-Aminolevulinato Sintetase/metabolismo , Aconitato Hidratase/metabolismo , Adolescente , Sequência de Aminoácidos , Anemia Sideroblástica/enzimologia , Linhagem Celular Transformada , Feminino , Ferroquelatase/metabolismo , Doenças Genéticas Ligadas ao Cromossomo X/enzimologia , Glutationa/metabolismo , Humanos , Mitocôndrias/enzimologia , Estresse Oxidativo , Linhagem , Estrutura Terciária de Proteína
6.
Haematologica ; 103(12): 2008-2015, 2018 12.
Artigo em Inglês | MEDLINE | ID: mdl-30026338

RESUMO

YARS2 variants have previously been described in patients with myopathy, lactic acidosis and sideroblastic anemia 2 (MLASA2). YARS2 encodes the mitochondrial tyrosyl-tRNA synthetase, which is responsible for conjugating tyrosine to its cognate mt-tRNA for mitochondrial protein synthesis. Here we describe 14 individuals from 11 families presenting with sideroblastic anemia and YARS2 variants that we identified using a sideroblastic anemia gene panel or exome sequencing. The phenotype of these patients ranged from MLASA to isolated congenital sideroblastic anemia. As in previous cases, inter- and intra-familial phenotypic variability was observed, however, this report includes the first cases with isolated sideroblastic anemia and patients with biallelic YARS2 variants that have no clinically ascertainable phenotype. We identified ten novel YARS2 variants and three previously reported variants. In vitro amino-acylation assays of five novel missense variants showed that three had less effect on the catalytic activity of YARS2 than the most commonly reported variant, p.(Phe52Leu), associated with MLASA2, which may explain the milder phenotypes in patients with these variants. However, the other two missense variants had a more severe effect on YARS2 catalytic efficiency. Several patients carried the common YARS2 c.572 G>T, p.(Gly191Val) variant (minor allele frequency =0.1259) in trans with a rare deleterious YARS2 variant. We have previously shown that the p.(Gly191Val) variant reduces YARS2 catalytic activity. Consequently, we suggest that biallelic YARS2 variants, including severe loss-of-function alleles in trans of the common p.(Gly191Val) variant, should be considered as a cause of isolated congenital sideroblastic anemia, as well as the MLASA syndromic phenotype.


Assuntos
Acidose Láctica/genética , Anemia Sideroblástica/genética , Doenças Genéticas Ligadas ao Cromossomo X/genética , Mutação em Linhagem Germinativa , Síndrome MELAS/genética , Proteínas Mitocondriais/genética , Tirosina-tRNA Ligase/genética , Acidose Láctica/enzimologia , Adolescente , Anemia Sideroblástica/enzimologia , Feminino , Estudos de Associação Genética , Doenças Genéticas Ligadas ao Cromossomo X/enzimologia , Humanos , Lactente , Síndrome MELAS/enzimologia , Masculino , Pessoa de Meia-Idade , Mutação de Sentido Incorreto , Adulto Jovem
8.
Biochim Biophys Acta Mol Basis Dis ; 1863(2): 428-439, 2017 02.
Artigo em Inglês | MEDLINE | ID: mdl-27838491

RESUMO

Mutations in the C-terminus of human erythroid 5-aminolevulinate synthase (hALAS2), a pyridoxal 5'-phosphate (PLP)-dependent enzyme, are associated with two different blood disorders, X-linked sideroblastic anemia (XLSA) and X-linked protoporphyria (XLPP). XLSA-causing mutations yield hALAS2 variants with decreased activity, while XLPP-causing mutations result in a gain-of-function of hALAS2. There are no specific treatments for XLPP. Isonicotinic acid hydrazide (isoniazid, INH), an antituberculosis agent, can cause sideroblastic anemia as a side-effect, by limiting PLP availability to hALAS2, via inhibition of pyridoxal kinase or reaction with pyridoxal to form pyridoxal isonicotinoyl hydrazone. We hypothesized that INH also binds and directly inhibits hALAS2. Using fluorescence-activated cell sorting and confocal fluorescence microscopy, we demonstrate that INH reduces protoporphyrin IX levels in HeLa cells expressing either wild-type hALAS2 or XLPP variants. In addition, PLP and pyridoxamine 5'-phosphate (PMP) reversed the cellular inhibition of hALAS2 activity by INH. Steady-state kinetic analyses with purified hALAS2 indicated that INH directly inhibits the enzyme, noncompetitively or uncompetitively, with an apparent Ki of 1.2µM. Circular dichroism spectroscopy revealed that INH triggered tertiary structural changes in hALAS2 that altered the microenvironment of the PLP cofactor and hampered the association of PLP with apo-hALAS2. Treatment of four XLPP patients with INH (5mg·kg-1·day-1) over a six-month period was well tolerated but without statistically significant modification of PPIX levels. These results, taken together, permit us to further an INH inhibition kinetic mechanism for ALAS, which suggests the possible use of INH-derived drugs in treating patients with XLPP and potentially other protoporphyrin-accumulating porphyrias.


Assuntos
5-Aminolevulinato Sintetase/deficiência , Inibidores Enzimáticos/farmacologia , Doenças Genéticas Ligadas ao Cromossomo X/tratamento farmacológico , Isoniazida/farmacologia , Protoporfiria Eritropoética/tratamento farmacológico , 5-Aminolevulinato Sintetase/antagonistas & inibidores , 5-Aminolevulinato Sintetase/sangue , 5-Aminolevulinato Sintetase/química , 5-Aminolevulinato Sintetase/metabolismo , Anemia Sideroblástica/enzimologia , Inibidores Enzimáticos/uso terapêutico , Doenças Genéticas Ligadas ao Cromossomo X/sangue , Doenças Genéticas Ligadas ao Cromossomo X/enzimologia , Células HeLa , Humanos , Isoniazida/uso terapêutico , Ligação Proteica/efeitos dos fármacos , Estrutura Terciária de Proteína/efeitos dos fármacos , Protoporfiria Eritropoética/sangue , Protoporfiria Eritropoética/enzimologia , Protoporfirinas/sangue , Fosfato de Piridoxal/metabolismo , Piridoxina/farmacologia , Complexo Vitamínico B/farmacologia
9.
Blood ; 124(18): 2867-71, 2014 Oct 30.
Artigo em Inglês | MEDLINE | ID: mdl-25193871

RESUMO

Mutations in genes encoding proteins that are involved in mitochondrial heme synthesis, iron-sulfur cluster biogenesis, and mitochondrial protein synthesis have previously been implicated in the pathogenesis of the congenital sideroblastic anemias (CSAs). We recently described a syndromic form of CSA associated with B-cell immunodeficiency, periodic fevers, and developmental delay (SIFD). Here we demonstrate that SIFD is caused by biallelic mutations in TRNT1, the gene encoding the CCA-adding enzyme essential for maturation of both nuclear and mitochondrial transfer RNAs. Using budding yeast lacking the TRNT1 homolog, CCA1, we confirm that the patient-associated TRNT1 mutations result in partial loss of function of TRNT1 and lead to metabolic defects in both the mitochondria and cytosol, which can account for the phenotypic pleiotropy.


Assuntos
Anemia Sideroblástica/congênito , Anemia Sideroblástica/genética , Deficiências do Desenvolvimento/complicações , Febre/complicações , Doenças Genéticas Ligadas ao Cromossomo X/genética , Síndromes de Imunodeficiência/complicações , Mutação/genética , RNA Nucleotidiltransferases/genética , Alelos , Anemia Sideroblástica/complicações , Anemia Sideroblástica/enzimologia , Deficiências do Desenvolvimento/genética , Febre/genética , Doenças Genéticas Ligadas ao Cromossomo X/complicações , Doenças Genéticas Ligadas ao Cromossomo X/enzimologia , Células HEK293 , Humanos , Síndromes de Imunodeficiência/genética
10.
Rom J Morphol Embryol ; 54(4): 1177-82, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24399021

RESUMO

In the WHO classification, there is a provisional entity called Myelodysplastic/Myeloproliferative Neoplasm, Unclassifiable (MDS/MPN, U). Refractory anemia with ringed sideroblasts associated with marked thrombocytosis (RARS-T) was included in this category. Recently published studies report a small percentage of patients with RARS-T. Sixty percent of these have JAK2 V617F mutation, which can suggest the coexistence of two pathological conditions (MDS and MPN). In this paper, we analyzed three patients diagnosed with RARS-T in the Department of Hematology, "Fundeni" Clinical Institute, Bucharest, Romania, during the period 2005-2011. The patients were investigated with cytogenetic exam and molecular biology. In these three cases were identified morphological features of multilineage dysplasia (two-lineage dysplasia in two cases and three-lineage dysplasia in one case). In two cases, thrombocytosis was under 1000×10(3)/µL and clinical evolution was similar to the myelodysplastic syndrome (transfusion dependent anemia with response to administration of erythropoietin). In the third case, the platelets were over 1000×10(3)/µL and with response to the treatment with Hydrea, which improved anemia. JAK2 V617F mutation was not identified in any case. RARS-T remains a provisional entity and requires a complex investigation of patients for the correct diagnosis of these patients. Therapeutic options should be personalized to each case in part because there is not yet a standardized treatment of these patients.


Assuntos
Anemia Refratária/complicações , Anemia Sideroblástica/complicações , Janus Quinase 2/genética , Mutação/genética , Trombocitose/complicações , Trombocitose/genética , Adulto , Idoso de 80 Anos ou mais , Substituição de Aminoácidos , Anemia Refratária/enzimologia , Anemia Refratária/genética , Anemia Sideroblástica/enzimologia , Anemia Sideroblástica/genética , Medula Óssea/patologia , Eritropoese , Feminino , Humanos , Masculino , Trombocitose/enzimologia , Adulto Jovem
11.
J Biol Chem ; 287(34): 28943-55, 2012 Aug 17.
Artigo em Inglês | MEDLINE | ID: mdl-22740690

RESUMO

Mutations in the erythroid-specific aminolevulinic acid synthase gene (ALAS2) cause X-linked sideroblastic anemia (XLSA) by reducing mitochondrial enzymatic activity. Surprisingly, a patient with the classic XLSA phenotype had a novel exon 11 mutation encoding a recombinant enzyme (p.Met567Val) with normal activity, kinetics, and stability. Similarly, both an expressed adjacent XLSA mutation, p.Ser568Gly, and a mutation (p.Phe557Ter) lacking the 31 carboxyl-terminal residues also had normal or enhanced activity, kinetics, and stability. Because ALAS2 binds to the ß subunit of succinyl-CoA synthetase (SUCLA2), the mutant proteins were tested for their ability to bind to this protein. Wild type ALAS2 bound strongly to a SUCLA2 affinity column, but the adjacent XLSA mutant enzymes and the truncated mutant did not bind. In contrast, vitamin B6-responsive XLSA mutations p.Arg452Cys and p.Arg452His, with normal in vitro enzyme activity and stability, did not interfere with binding to SUCLA2 but instead had loss of positive cooperativity for succinyl-CoA binding, an increased K(m) for succinyl-CoA, and reduced vitamin B6 affinity. Consistent with the association of SUCLA2 binding with in vivo ALAS2 activity, the p.Met567GlufsX2 mutant protein that causes X-linked protoporphyria bound strongly to SUCLA2, highlighting the probable role of an ALAS2-succinyl-CoA synthetase complex in the regulation of erythroid heme biosynthesis.


Assuntos
5-Aminolevulinato Sintetase/metabolismo , Anemia Sideroblástica , Doenças Genéticas Ligadas ao Cromossomo X , Mutação de Sentido Incorreto , Succinato-CoA Ligases/metabolismo , 5-Aminolevulinato Sintetase/genética , Adulto , Substituição de Aminoácidos , Anemia Sideroblástica/enzimologia , Anemia Sideroblástica/genética , Estabilidade Enzimática/genética , Doenças Genéticas Ligadas ao Cromossomo X/enzimologia , Doenças Genéticas Ligadas ao Cromossomo X/genética , Heme/biossíntese , Heme/genética , Humanos , Masculino , Ligação Proteica , Protoporfiria Eritropoética/enzimologia , Protoporfiria Eritropoética/genética , Succinato-CoA Ligases/genética , Vitamina B 6/genética , Vitamina B 6/metabolismo
12.
Acta Haematol ; 125(4): 193-7, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21252495

RESUMO

BACKGROUND/AIMS: Congenital sideroblastic anemias (SA) are characterized by the presence of ringed sideroblasts in the bone marrow. The most common form is X-linked SA, which results from mutations in erythroid-specific δ-aminolevulinate synthase (ALAS2), the first enzyme in heme biosynthesis. In addition, autosomal recessive mutations in the erythroid-specific mitochondrial transporter SLC25A38 and glutaredoxin 5 (GLRX5) have recently been identified in SA patients with isolated erythroid phenotype. MATERIALS AND METHODS: We studied 5 young males with congenital SA from the Czech Republic. Mutation analysis was performed on the complete coding regions of 3 candidate genes (ALAS2, SLC25A38 and GLRX5), and the enzyme activity of ALAS2 was measured by a continuous spectrophotometric assay. RESULTS: We found the previously published R452H and R452C ALAS2 mutations in 3 patients. A novel K156E substitution in ALAS2 was discovered in 1 pyridoxine-responsive patient. The functional study showed that this substitution severely decreases ALAS2 enzyme activity. In 1 pyridoxine-refractory patient, no mutations were detected in ALAS2, SLC25A38 or GLRX5. CONCLUSION: Our report extends the list of known ALAS2 mutations, with the addition of a novel K156E substitution that is responsive to pyridoxine treatment and contributes to the general knowledge of congenital SA cases characterized worldwide.


Assuntos
5-Aminolevulinato Sintetase/genética , 5-Aminolevulinato Sintetase/metabolismo , Adulto , Anemia Sideroblástica/tratamento farmacológico , Anemia Sideroblástica/enzimologia , Anemia Sideroblástica/genética , Sequência de Bases , Doenças Genéticas Ligadas ao Cromossomo X , Humanos , Masculino , Mutação , Piridoxina/uso terapêutico , Alinhamento de Sequência
13.
Cell Mol Biol (Noisy-le-grand) ; 55(1): 102-10, 2009 Feb 16.
Artigo em Inglês | MEDLINE | ID: mdl-19268008

RESUMO

5-Aminolevulinate synthase is a homodimeric pyridoxal 5'-phosphate-dependent enzyme that catalyzes the first step of the heme biosynthetic pathway in animals, fungi, and the alpha-subclass of the photosynthetic purple bacteria. The reaction cycle involves condensation of glycine with succinyl-coenzyme A to yield 5-aminolevulinate, carbon dioxide, and CoA. Mutations in the human erythroid-specific aminolevulinate synthase gene are associated with the erythropoietic disorder X-linked sideroblastic anemia. Recent kinetic and crystallographic data have facilitated an unprecedented understanding of how this important enzyme produces 5-aminolevulinate, and suggest possible directions for future research that may lead to treatments not only for X-linked sideroblastic anemia, but also other diseases.


Assuntos
5-Aminolevulinato Sintetase/metabolismo , Heme/biossíntese , 5-Aminolevulinato Sintetase/química , 5-Aminolevulinato Sintetase/genética , Ácido Aminolevulínico/metabolismo , Anemia Sideroblástica/enzimologia , Anemia Sideroblástica/genética , Humanos , Cinética , Modelos Moleculares , Mutação , Relação Estrutura-Atividade
14.
Artigo em Inglês | MEDLINE | ID: mdl-19074058

RESUMO

Acquired sideroblastic anemia with unilineage dysplasia (WHO RARS) is a clonal stem cell disorder characterized by erythroid dysplasia, mitochondrial accumulation of mitochondrial ferritin, defective erythroid maturation and anemia. A fraction of these patients also show elevated platelet counts; since 2001 this has been defined as RARS with marked thrombocytosis (RARS-T). It has recently been described that around half of RARS-T patients, along with a small subset of other MDS and mixed myelodysplastic/ myeloproliferative disorders, carry the JAK2 mutation, and that MPL mutations are found in single patients. Clinically, RARS-T patients show features of both RARS, essential thrombocythmia (ET) and to some extent also myelofibrosis. However, the degree of anemia and overall survival is more similar to RARS than myeloproliferative disorders. The occurrence of JAK2 mutations and features of ET in RARS is too frequent to be the result of chance only, and it is possible that this link may provide a key to an increased understanding of the genetic abnormalities causing ring sideroblast formation.


Assuntos
Anemia Sideroblástica/genética , Cromossomos Humanos Par 5 , Janus Quinase 2/genética , Síndromes Mielodisplásicas/genética , Anemia Sideroblástica/enzimologia , Anemia Sideroblástica/etiologia , Anemia Sideroblástica/patologia , Células da Medula Óssea/patologia , Eritroblastos/patologia , Eritropoese/genética , Humanos , Intoxicação por Chumbo/complicações , Mutação , Síndromes Mielodisplásicas/classificação , Síndromes Mielodisplásicas/enzimologia , Síndromes Mielodisplásicas/patologia , Trombocitose/complicações , Trombocitose/enzimologia , Trombocitose/genética , Organização Mundial da Saúde
17.
J Med Genet ; 44(3): 173-80, 2007 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-17056637

RESUMO

INTRODUCTION: Myopathy, lactic acidosis and sideroblastic anaemia (MLASA) is a rare condition that combines early-onset myopathy with lactic acidosis and sideroblastic anaemia. MLASA has been associated with a missense mutation in pseudouridylate synthase 1 (PUS1), an enzyme located in both nucleus and mitochondria, which converts uridine into pseudouridine in several cytosolic and mitochondrial tRNA positions and increases the efficiency of protein synthesis in both compartments. SUBJECTS AND METHODS: We have identified two Italian brothers, offspring of distantly related parents, both of whom are affected by MLASA. The six exons of the PUS1 gene were analysed by automated sequencing. RESULTS: We found combined defects in mitochondrial respiratory chain complexes in muscle and fibroblast homogenates of both patients, and low levels of mtDNA translation products in fibroblast mitochondria. A novel, homozygous stop mutation was present in PUS1 (E220X). We have investigated the structural and mechanistic aspects of the double localisation of PUS1, demonstrating that the isoform located in the nucleus contains an N-terminal extension which is absent in the mature mitochondrial isoform. CONCLUSIONS: The stop mutation in PUS1 is likely to determine the loss of function of the protein, since it predicts the synthesis of a protein missing 208/427 amino acid residues on the C terminus, and was associated with low mtDNA translation. The structural differences in nuclear versus mitochondrial isoforms of PUS1 may be implicated in the variability of the clinical presentations in MLASA.


Assuntos
Acidose Láctica/genética , Anemia Sideroblástica/genética , Códon sem Sentido , Hidroliases/genética , Miopatias Mitocondriais/genética , Acidose Láctica/enzimologia , Sequência de Aminoácidos , Anemia Sideroblástica/enzimologia , Núcleo Celular/enzimologia , Consanguinidade , Deficiência de Citocromo-c Oxidase/genética , Complexo I de Transporte de Elétrons/deficiência , Evolução Fatal , Fibroblastos/enzimologia , Fibroblastos/patologia , Células HeLa , Hormônio do Crescimento Humano/deficiência , Humanos , Hidroliases/química , Hidroliases/deficiência , Recém-Nascido , Deficiência Intelectual/genética , Masculino , Mitocôndrias Musculares/enzimologia , Miopatias Mitocondriais/enzimologia , Dados de Sequência Molecular , Músculo Esquelético/enzimologia , Músculo Esquelético/patologia , Fenótipo , Isoformas de Proteínas/análise , Isoformas de Proteínas/genética , Precursores de Proteínas/metabolismo , Processamento de Proteína Pós-Traducional , Transporte Proteico/efeitos dos fármacos , Síndrome , Transcrição Gênica , Transfecção , Valinomicina/farmacologia
18.
Antioxid Redox Signal ; 8(7-8): 1217-25, 2006.
Artigo em Inglês | MEDLINE | ID: mdl-16910769

RESUMO

Iron overload is a feature of an array of human disorders such as sideroblastic anemias, a heterogeneous group of erythropoietic disorders without identified cause in most cases. However, sideroblastic anemias appear to result from a disturbance at the interface between mitochondrial function and iron metabolism. A defining feature is excessive iron deposition within mitochondria of developing red cells, the consequences of which are an increase in cellular free radicals production, increased damage to proteins, and reduced cell survival. Because of its mitochondrial location, superoxide dismutase (SOD2) is the principal defense against the toxicity of superoxide anions generated by the oxidative phosphorylation. We have used hematopoietic stem cell transplantation to study blood cells lacking SOD2. We became interested in the role SOD2 plays in the metabolism of superoxide anions during erythroid development, as anemia is the major phenotype in transplanted animals. Our exploration of this model suggests that oxidative stress-and in particular, mitochondrial- derived oxidants-plays an important role in the pathogenesis of the human disorder, sideroblastic anemia. Here we review the relation between mitochondrial dysfunction and sideroblastic anemia, describe several methods for assessing oxidative damage to mature or developing red cells, present data on, and discuss the potential of antioxidant therapy for this disorder.


Assuntos
Anemia Sideroblástica/metabolismo , Anemia Sideroblástica/patologia , Antioxidantes/metabolismo , Estresse Oxidativo , Superóxido Dismutase/deficiência , Anemia Sideroblástica/enzimologia , Anemia Sideroblástica/etiologia , Anemia Sideroblástica/genética , Animais , Antioxidantes/farmacologia , Eritrócitos/efeitos dos fármacos , Eritrócitos/metabolismo , Eritrócitos/patologia , Humanos , Manganês/administração & dosagem , Manganês/uso terapêutico , Manganês/toxicidade , Camundongos , Camundongos Knockout , Mitocôndrias/metabolismo , Mitocôndrias/patologia , Superóxido Dismutase/genética , Superóxido Dismutase/metabolismo , Superóxidos/metabolismo
19.
Biochem Biophys Res Commun ; 348(3): 799-806, 2006 Sep 29.
Artigo em Inglês | MEDLINE | ID: mdl-16904069

RESUMO

Erythroid-specific 5-aminolevulinate synthase (ALAS2) catalyzes the rate-limiting step in heme biosynthesis of erythroid cells. Here, we show that treatment of erythroid K562 cells with HDAC inhibitors sodium butyrate or Trichostatin A gave rise to a significant increase in ALAS2 gene transcripts, with a concurrent increase in acetylation level of histone H4 at the ALAS2 gene promoter. Histone acetyltransferase p300 bound withALAS2 promoter and overexpression of p300 increased both the promoter reporter expression and endogenous mRNA level of ALAS2. Additionally, two functional Sp1 sites located in ALAS2 promoter were identified. Both of the GATA-1 sites and all the Sp1 sites at the ALAS2 promoter contributed to the transcription synergistic action with p300. These data implicated a close relationship between the acetylation modification of histone at the ALAS2 promoter and the regulation of this gene. Meanwhile, this work identified that ALAS2 is a novel target gene for p300/CBP action as histone acetyltransferases.


Assuntos
5-Aminolevulinato Sintetase/genética , Proteínas de Ciclo Celular/fisiologia , Eritropoese/genética , Histona Acetiltransferases/fisiologia , Fatores de Transcrição/fisiologia , Transcrição Gênica , 5-Aminolevulinato Sintetase/metabolismo , Acetilação , Anemia Sideroblástica/enzimologia , Anemia Sideroblástica/genética , Proteínas de Ciclo Celular/antagonistas & inibidores , Proteínas de Ciclo Celular/biossíntese , Proteínas de Ciclo Celular/genética , Linhagem Celular , Globinas/biossíntese , Globinas/genética , Histona Acetiltransferases/antagonistas & inibidores , Histona Acetiltransferases/biossíntese , Histona Acetiltransferases/genética , Histonas/metabolismo , Humanos , Células K562 , RNA Mensageiro/biossíntese , Fatores de Transcrição/antagonistas & inibidores , Fatores de Transcrição/biossíntese , Fatores de Transcrição/genética , Fatores de Transcrição de p300-CBP
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...