Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 65
Filtrar
2.
Curr Res Transl Med ; 72(1): 103438, 2024 03.
Artículo en Inglés | MEDLINE | ID: mdl-38244303

RESUMEN

Congenital sideroblastic anemia (CSA) is a group of disorders caused by different genetic mutations that result in low iron utilization and ineffective erythropoiesis. Current treatments are limited, and some patients do not respond to vitamin B6 therapy. Luspatercept is a novel erythropoietic maturation agent approved for adult ß-thalassemia and Myelodysplastic syndromes with ring sideroblasts (MDS-RS) associated with ineffective erythropoiesis. Here we report 2 patients with CSA due to mutations in ALAS2 and SLC25A38 genes who became unresponsive after a period of treatment with vitamin B6 and iron chelators but achieved transfusion independence and a markedly reduced spleen after combination with luspatercept.


Asunto(s)
Receptores de Activinas Tipo II , Anemia Sideroblástica , Enfermedades Genéticas Ligadas al Cromosoma X , Proteínas Recombinantes de Fusión , Adulto , Humanos , 5-Aminolevulinato Sintetasa , Receptores de Activinas Tipo II/efectos adversos , Anemia Sideroblástica/tratamiento farmacológico , Anemia Sideroblástica/genética , Anemia Sideroblástica/congénito , Fragmentos Fc de Inmunoglobulinas/efectos adversos , Proteínas Recombinantes de Fusión/efectos adversos , Vitamina B 6
4.
Exp Clin Transplant ; 21(1): 70-75, 2023 01.
Artículo en Inglés | MEDLINE | ID: mdl-36757170

RESUMEN

Congenital sideroblastic anemia is characterized by anemia and intramitochondrial iron accumulation in erythroid precursors that form ring sideroblasts. The most common recessive forms are caused by sequence variations in the ALAS2 and SLC25A38 genes. In patients with transfusion-dependent and pyridoxine- resistant severe congenital sideroblastic anemia, hematopoietic stem celltransplantis the only curative option. Herein, we described successful implementations of allogeneic hematopoietic stem cell transplant in 4 Iranian children with congenital sideroblastic anemia. The patients had presented with clinical manifestations of anemia early in life, and the diagnoses of congenital sideroblastic anemia were established through blood tests and bone marrow aspiration. Congenital sideroblastic anemia was further confirmed by the identification of pathogenic variants in SLC25A38 in 2 patients. All 4 patients received allogeneic hematopoietic stem cell transplant with myeloablative conditioning regimen that included busulfan, cyclophosphamide, andrabbit antithymocyte globulin. A combination of cyclosporine A and methotrexate or mycophenolate mofetil was used for graft-versus-host disease prophylaxis. Bone marrow and peripheral blood from sibling or related donors with fully matched human leukocyte antigen profiles were applied. The outcomes of hematopoietic stem celltransplantin patients with congenital sideroblastic anemia were favorable. Three patients achieved full donor chimerism (>95%, 98%, and 100%), and the other patient showed mixed chimerism (75%). All patients remained transfusion independent. Hemato- poietic stem celltransplantis a curative treatmentthat can provide long-term survival for patients with congenital sideroblastic anemia, particularly when used in a timely manner. There remain ongoing challenges in various aspects of hematopoietic stem celltransplantin patients with congenital sideroblastic anemia, which remain to be elucidated.


Asunto(s)
Anemia Sideroblástica , Enfermedad Injerto contra Huésped , Trasplante de Células Madre Hematopoyéticas , Niño , Humanos , 5-Aminolevulinato Sintetasa/genética , Anemia Sideroblástica/diagnóstico , Anemia Sideroblástica/genética , Anemia Sideroblástica/congénito , Ciclosporina , Irán , Acondicionamiento Pretrasplante
5.
Hum Mutat ; 42(11): 1367-1383, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34298585

RESUMEN

The congenital sideroblastic anemias (CSAs) are a heterogeneous group of inherited disorders of erythropoiesis characterized by pathologic deposits of iron in the mitochondria of developing erythroblasts. Mutations in the mitochondrial glycine carrier SLC25A38 cause the most common recessive form of CSA. Nonetheless, the disease is still rare, there being fewer than 70 reported families. Here we describe the clinical phenotype and genotypes of 31 individuals from 24 families, including 11 novel mutations. We also review the spectrum of reported mutations and genotypes associated with the disease, describe the unique localization of missense mutations in transmembrane domains and account for the presence of several alleles in different populations.


Asunto(s)
Anemia Sideroblástica/congénito , Genotipo , Proteínas de Transporte de Membrana Mitocondrial/genética , Mutación , Fenotipo , Preescolar , Femenino , Humanos , Lactante , Recién Nacido , Masculino
6.
J Clin Invest ; 130(10): 5245-5256, 2020 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-32634119

RESUMEN

The congenital sideroblastic anemias (CSAs) can be caused by primary defects in mitochondrial iron-sulfur (Fe-S) cluster biogenesis. HSCB (heat shock cognate B), which encodes a mitochondrial cochaperone, also known as HSC20 (heat shock cognate protein 20), is the partner of mitochondrial heat shock protein A9 (HSPA9). Together with glutaredoxin 5 (GLRX5), HSCB and HSPA9 facilitate the transfer of nascent 2-iron, 2-sulfur clusters to recipient mitochondrial proteins. Mutations in both HSPA9 and GLRX5 have previously been associated with CSA. Therefore, we hypothesized that mutations in HSCB could also cause CSA. We screened patients with genetically undefined CSA and identified a frameshift mutation and a rare promoter variant in HSCB in a female patient with non-syndromic CSA. We found that HSCB expression was decreased in patient-derived fibroblasts and K562 erythroleukemia cells engineered to have the patient-specific promoter variant. Furthermore, gene knockdown and deletion experiments performed in K562 cells, zebrafish, and mice demonstrate that loss of HSCB results in impaired Fe-S cluster biogenesis, a defect in RBC hemoglobinization, and the development of siderocytes and more broadly perturbs hematopoiesis in vivo. These results further affirm the involvement of Fe-S cluster biogenesis in erythropoiesis and hematopoiesis and define HSCB as a CSA gene.


Asunto(s)
Anemia Sideroblástica/genética , Chaperonas Moleculares/genética , Mutación , Adolescente , Anemia Sideroblástica/congénito , Anemia Sideroblástica/metabolismo , Animales , Niño , Análisis Mutacional de ADN , Femenino , Mutación del Sistema de Lectura , Técnicas de Silenciamiento del Gen , Humanos , Proteínas Hierro-Azufre/deficiencia , Proteínas Hierro-Azufre/genética , Células K562 , Masculino , Ratones , Ratones Noqueados , Chaperonas Moleculares/metabolismo , Linaje , Polimorfismo de Nucleótido Simple , Regiones Promotoras Genéticas , Adulto Joven , Pez Cebra
7.
Pediatr Hematol Oncol ; 36(1): 46-51, 2019 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-30912988

RESUMEN

Congenital sideroblastic anemia (CSA) is a rare hereditary disease causing disorders of hemoglobin synthesis. Severe, progressive congenital sideroblastic anemia becomes transfusion dependent and results in iron overload. In such cases, patients must undergo stem cell transplantation (SCT) before critical organ dysfunction occurs. However, there has been no consensus on the criteria for SCT for congenital sideroblastic anemia. A 17-year-old Japanese boy was newly diagnosed with congenital sideroblastic anemia manifesting dyspnea on effort. His gene analysis revealed ALAS2 R170L. He gradually become dependent on RBC transfusion. Vitamin B6 (pyridoxal, 30 mg/day) therapy and high-dose alpha-linoleic acid supplementation (150 mg/day) had not been effective. We treated the patient with reduced-intensity SCT from a human leukocyte antigen (HLA) alle 8/8-identical unrelated female donor. The preparation regimen applied consisted of cyclophosphamide, fludarabine, total body irradiation (2 Gy), and anti-thymocyte globulin. We experienced secondary graft failure, nevertheless we used enough immunosuppression. Here we discuss the optimal transplantation regimen for an adult-onset congenital sideroblastic anemia patient with transfusion dependency and mild hemosiderosis. We consider a positive indication for SCT in younger (< 20-year-old) patients with congenital sideroblastic anemia with transfusion dependency. Each case should be individually considered for their suitability for SCT depending on the feasibility and the clinical condition of the patient.


Asunto(s)
5-Aminolevulinato Sintetasa/genética , Anemia Sideroblástica/congénito , Anemia Sideroblástica/terapia , Rechazo de Injerto/genética , Mutación Missense , Trasplante de Células Madre , Adolescente , Aloinjertos , Sustitución de Aminoácidos , Humanos , Masculino
11.
Gene ; 668: 182-189, 2018 Aug 20.
Artículo en Inglés | MEDLINE | ID: mdl-29787825

RESUMEN

Congenital sideroblastic anemia (CSA) is a series of rare, heterogeneous disorders, characterized by iron overload in the mitochondria of erythroblasts and ringed sideroblasts in bone marrow. In recent years, rapid development of next-generation sequencing technology brings great advance in understanding of genetic and pathophysiologic features of CSA. Based on the pathophysiology of mitochondrial iron metabolism, causative genes of CSA can be divided into three subtypes: heme biosynthesis related; iron­sulfur cluster biosynthesis and transportation related; and mitochondrial respiratory chain synthesis related. Patients with CSA present various clinical manifestation due to relevant mutation gene and require different treatment strategies. The recognition of the causative genes and evolution of pathogenicity is critical. In this review, we summarize the recent progress in mutation genes of CSA, and its potential role in the pathogenesis, diagnosis and treatment.


Asunto(s)
Anemia Sideroblástica/genética , Mutación , Anemia Sideroblástica/congénito , Anemia Sideroblástica/metabolismo , Hemo/biosíntesis , Humanos , Hierro/metabolismo , Proteínas Mitocondriales/biosíntesis
12.
Int J Hematol ; 107(1): 44-54, 2018 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-29139060

RESUMEN

Sideroblastic anemias are anemic disorders characterized by the presence of ring sideroblasts in a patient's bone marrow. These disorders are typically divided into two types, congenital or acquired sideroblastic anemia. Recently, several genes were reported as responsible for congenital sideroblastic anemia; however, the relationship between the function of the gene products and ring sideroblasts is largely unclear. In this review article, we will focus on the iron metabolism in erythroid cells as well as in patients with congenital sideroblastic anemia.


Asunto(s)
Anemia Sideroblástica/congénito , Anemia Sideroblástica/genética , Células Eritroides/metabolismo , Hierro/metabolismo , Acil-CoA Deshidrogenasa de Cadena Larga/deficiencia , Anemia Sideroblástica/sangre , Anemia Sideroblástica/metabolismo , Ataxia Cerebelosa , Cromosomas Humanos X/genética , Síndromes Congénitos de Insuficiencia de la Médula Ósea , Complejo I de Transporte de Electrón/deficiencia , Complejo I de Transporte de Electrón/genética , Femenino , Glutarredoxinas/genética , Proteínas HSP70 de Choque Térmico/genética , Hemo/biosíntesis , Humanos , Errores Innatos del Metabolismo Lipídico , Síndrome MELAS , Masculino , Enfermedades Mitocondriales , Proteínas de Transporte de Membrana Mitocondrial/genética , Proteínas Mitocondriales/genética , Enfermedades Musculares , Mutación
13.
Blood Cells Mol Dis ; 66: 11-18, 2017 07.
Artículo en Inglés | MEDLINE | ID: mdl-28772256

RESUMEN

The most frequent germline mutations responsible for non syndromic congenital sideroblastic anemia are identified in ALAS2 and SLC25A38 genes. Iron overload is a key issue and optimal chelation therapy should be used to limit its adverse effects on the development of children. Our multicentre retrospective descriptive study compared the strategies for diagnosis and management of congenital sideroblastic anemia during the follow-up of six patients with an ALAS2 mutation and seven patients with an SLC25A38 mutation. We described in depth the clinical, biological and radiological phenotype of these patients at diagnosis and during follow-up and highlighted our results with a review of available evidence and data on the management strategies for congenital sideroblastic anemia. This report confirms the considerable variability in manifestations among patients with ALAS2 or SLC25A38 mutations and draws attention to differences in the assessment and the monitoring of iron overload and its complications. The use of an international registry would certainly help defining recommendations for the management of these rare disorders to improve patient outcome.


Asunto(s)
5-Aminolevulinato Sintetasa/genética , Anemia Sideroblástica/congénito , Proteínas de Transporte de Membrana Mitocondrial/genética , Anemia Sideroblástica/genética , Niño , Humanos , Sobrecarga de Hierro , Fenotipo , Estudios Retrospectivos
14.
Rinsho Ketsueki ; 58(4): 347-352, 2017.
Artículo en Japonés | MEDLINE | ID: mdl-28484165

RESUMEN

Sideroblastic anemia is characterized by anemia with ring sideroblasts produced by the bone marrow. Sideroblasts are formed by disutilization and deposit of iron in the mitochondoria. There are two forms of sideroblastic anemia: congenital and acquired. Congenital sideroblastic anemia is caused by mutations in genes involved in heme biosynthesis, iron-sulfur (Fe-S) cluster biogenesis, or mitochondrial metabolism. Although there is a variation in the mutated genes among races, the most common congenital sideroblastic anemia is X-linked sideroblastic anemia caused by mutations in the erythroid-specific δ-aminolevulinate synthase gene, which is the first enzyme of heme biosynthesis in erythroid cells. The most commonly acquired sideroblastic anemia is myelodysplastic syndrome with ring sideroblasts (MDS-RS). It has been shown that the splicing factor 3b subunit 1 (SF3B1) gene, which is a core component of the RNA splicing complex, is highly mutated in MDS-RS, although the underlying mechanism of the onset of the disease by the mutation of the SF3B1 gene remains unclear. Molecular analysis will contribute to the development of effective treatment for congenital and acquired sideroblastic anemia, which are intractable diseases.


Asunto(s)
Anemia Sideroblástica , 5-Aminolevulinato Sintetasa/genética , Anemia Sideroblástica/congénito , Anemia Sideroblástica/diagnóstico , Anemia Sideroblástica/epidemiología , Anemia Sideroblástica/genética , Animales , Humanos , Mutación
16.
Blood ; 124(18): 2867-71, 2014 Oct 30.
Artículo en Inglés | MEDLINE | ID: mdl-25193871

RESUMEN

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.


Asunto(s)
Anemia Sideroblástica/congénito , Anemia Sideroblástica/genética , Discapacidades del Desarrollo/complicaciones , Fiebre/complicaciones , Enfermedades Genéticas Ligadas al Cromosoma X/genética , Síndromes de Inmunodeficiencia/complicaciones , Mutación/genética , ARN Nucleotidiltransferasas/genética , Alelos , Anemia Sideroblástica/complicaciones , Anemia Sideroblástica/enzimología , Discapacidades del Desarrollo/genética , Fiebre/genética , Enfermedades Genéticas Ligadas al Cromosoma X/complicaciones , Enfermedades Genéticas Ligadas al Cromosoma X/enzimología , Células HEK293 , Humanos , Síndromes de Inmunodeficiencia/genética
17.
Haematologica ; 99(2): 252-61, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23935018

RESUMEN

Erythroid-specific 5-aminolevulinate synthase (ALAS2) is the rate-limiting enzyme for heme biosynthesis in erythroid cells, and a missense mutation of the ALAS2 gene is associated with congenital sideroblastic anemia. However, the gene responsible for this form of anemia remains unclear in about 40% of patients. Here, we identify a novel erythroid-specific enhancer of 130 base pairs in the first intron of the ALAS2 gene. The newly identified enhancer contains a cis-acting element that is bound by the erythroid-specific transcription factor GATA1, as confirmed by chromatin immunoprecipitation analysis in vivo and by electrophoretic mobility shift assay in vitro. A promoter activity assay in K562 human erythroleukemia cells revealed that the presence of this 130-base pair region increased the promoter activity of the ALAS2 gene by 10-15-fold. Importantly, two mutations, each of which disrupts the GATA-binding site in the enhancer, were identified in unrelated male patients with congenital sideroblastic anemia, and the lower expression level of ALAS2 mRNA in bone marrow erythroblasts was confirmed in one of these patients. Moreover, GATA1 failed to bind to each mutant sequence at the GATA-binding site, and each mutation abolished the enhancer function on ALAS2 promoter activity in K562 cells. Thus, a mutation at the GATA-binding site in this enhancer may cause congenital sideroblastic anemia. These results suggest that the newly identified intronic enhancer is essential for the expression of the ALAS2 gene in erythroid cells. We propose that the 130-base pair enhancer region located in the first intron of the ALAS2 gene should be examined in patients with congenital sideroblastic anemia in whom the gene responsible is unknown.


Asunto(s)
5-Aminolevulinato Sintetasa/genética , Anemia Sideroblástica/genética , Elementos de Facilitación Genéticos , Factor de Transcripción GATA1/genética , Mutación , Elementos de Respuesta , 5-Aminolevulinato Sintetasa/metabolismo , Anemia Sideroblástica/congénito , Anemia Sideroblástica/metabolismo , Factor de Transcripción GATA1/metabolismo , Humanos , Células K562 , Masculino
18.
Pediatr Int ; 55(6): 675-9, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-24003969

RESUMEN

Sideroblastic anemias are heterogeneous congenital and acquired disorders characterized by anemia and the presence of ringed sideroblasts in the bone marrow. Congenital sideroblastic anemia (CSA) is a rare disease caused by mutations of genes involved in heme biosynthesis, iron-sulfur [Fe-S] cluster biosynthesis, and mitochondrial protein synthesis. The most common form is X-linked sideroblastic anemia, due to mutations in the erythroid-specific δ-aminolevulinate synthase (ALAS2), which is the first enzyme of the heme biosynthesis pathway in erythroid cells. Other known etiologies include mutations in the erythroid specific mitochondrial transporter (SLC25A38), adenosine triphosphate (ATP) binding cassette B7 (ABCB7), glutaredoxin 5 (GLRX5), thiamine transporter SLC19A2, the RNA-modifying enzyme pseudouridine synthase (PUS1), and mitochondrial tyrosyl-tRNA synthase (YARS2), as well as mitochondrial DNA deletions. Due to its rarity, however, there have been few systematic pathophysiological and genetic investigations focusing on sideroblastic anemia. Therefore, a nationwide survey of sideroblastic anemia was conducted in Japan to investigate the epidemiology and pathogenesis of this disease. This review will cover the findings of this recent survey and summarize the current understanding of the pathophysiology and genetic mutations involved in CSA.


Asunto(s)
Anemia Sideroblástica/genética , Anemia Sideroblástica/fisiopatología , Enfermedades Genéticas Ligadas al Cromosoma X/genética , Enfermedades Genéticas Ligadas al Cromosoma X/fisiopatología , Mutación , Anemia Sideroblástica/congénito , Niño , Humanos
19.
Am J Physiol Gastrointest Liver Physiol ; 304(12): G1095-102, 2013 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-23599042

RESUMEN

The iron-diabetes hypothesis proposes an association between iron overload and glucose metabolism that is supported by a number of epidemiological studies. The prevalence of type 2 diabetes in patients with hereditary hemochromatosis and iron-loading thalassemia supports this hypothesis. The Belgrade rat carries a mutation in the iron transporter divalent metal transporter 1 (DMT1) resulting in iron-loading anemia. In this study, we characterized the glycometabolic status of the Belgrade rat. Belgrade rats displayed normal glycemic control. Insulin signaling and secretion were not impaired, and pancreatic tissue did not incur damage despite high levels of nonheme iron. These findings suggest that loss of DMT1 protects against oxidative damage to the pancreas and helps to maintain insulin sensitivity despite iron overload. Belgrade rats had lower body weight but increased food consumption compared with heterozygous littermates. The unexpected energy balance was associated with increased urinary glucose output. Increased urinary excretion of electrolytes, including iron, was also observed. Histopathological evidence suggests that altered renal function is secondary to changes in kidney morphology, including glomerulosclerosis. Thus, loss of DMT1 appears to protect the pancreas from injury but damages the integrity of kidney structure and function.


Asunto(s)
Anemia Sideroblástica/congénito , Glucemia/metabolismo , Glucosuria/metabolismo , Anemia Sideroblástica/genética , Anemia Sideroblástica/metabolismo , Animales , Proteínas de Transporte de Catión/genética , Modelos Animales de Enfermedad , Intolerancia a la Glucosa , Insulina/sangre , Insulina/metabolismo , Resistencia a la Insulina , Secreción de Insulina , Hierro/metabolismo , Sobrecarga de Hierro/metabolismo , Riñón/patología , Riñón/fisiopatología , Músculo Esquelético/metabolismo , Estrés Oxidativo , Páncreas/metabolismo , Páncreas/patología , Páncreas/fisiopatología , Ratas , Ratas Mutantes , Transducción de Señal
20.
Ann Hematol ; 92(1): 1-9, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-22983749

RESUMEN

Sideroblastic anemia is characterized by anemia with the emergence of ring sideroblasts in the bone marrow. There are two forms of sideroblastic anemia, i.e., congenital sideroblastic anemia (CSA) and acquired sideroblastic anemia. In order to clarify the pathophysiology of sideroblastic anemia, a nationwide survey consisting of clinical and molecular genetic analysis was performed in Japan. As of January 31, 2012, data of 137 cases of sideroblastic anemia, including 72 cases of myelodysplastic syndrome (MDS)-refractory cytopenia with multilineage dysplasia (RCMD), 47 cases of MDS-refractory anemia with ring sideroblasts (RARS), and 18 cases of CSA, have been collected. Hemoglobin and MCV level in CSA are significantly lower than those of MDS, whereas serum iron level in CSA is significantly higher than those of MDS. Of 14 CSA for which DNA was available for genetic analysis, 10 cases were diagnosed as X-linked sideroblastic anemia due to ALAS2 gene mutation. The mutation of SF3B1 gene, which was frequently mutated in MDS-RS, was not detected in CSA patients. Together with the difference of clinical data, it is suggested that genetic background, which is responsible for the development of CSA, is different from that of MDS-RS.


Asunto(s)
Anemia Sideroblástica/congénito , 5-Aminolevulinato Sintetasa/deficiencia , 5-Aminolevulinato Sintetasa/genética , 5-Aminolevulinato Sintetasa/metabolismo , Transportadoras de Casetes de Unión a ATP/deficiencia , Transportadoras de Casetes de Unión a ATP/genética , Adolescente , Adulto , Edad de Inicio , Anciano , Anemia Sideroblástica/sangre , Anemia Sideroblástica/clasificación , Anemia Sideroblástica/epidemiología , Anemia Sideroblástica/genética , Niño , Preescolar , Aberraciones Cromosómicas , Femenino , Frecuencia de los Genes , Genes Ligados a X , Enfermedades Genéticas Ligadas al Cromosoma X/sangre , Enfermedades Genéticas Ligadas al Cromosoma X/genética , Glutarredoxinas/deficiencia , Glutarredoxinas/genética , Encuestas Epidemiológicas , Humanos , Hidroliasas/deficiencia , Hidroliasas/genética , Lactante , Recién Nacido , Japón/epidemiología , Masculino , Proteínas de Transporte de Membrana/deficiencia , Proteínas de Transporte de Membrana/genética , Persona de Mediana Edad , Proteínas de Transporte de Membrana Mitocondrial/deficiencia , Proteínas de Transporte de Membrana Mitocondrial/genética , Síndromes Mielodisplásicos/sangre , Síndromes Mielodisplásicos/tratamiento farmacológico , Síndromes Mielodisplásicos/epidemiología , Síndromes Mielodisplásicos/genética , Fosfoproteínas/deficiencia , Fosfoproteínas/genética , Factores de Empalme de ARN , Proteínas Recombinantes de Fusión/metabolismo , Ribonucleoproteína Nuclear Pequeña U2/deficiencia , Ribonucleoproteína Nuclear Pequeña U2/genética , Resultado del Tratamiento , Vitamina B 6/uso terapéutico , Adulto Joven
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA