Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 31
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Am J Hum Genet ; 103(6): 930-947, 2018 12 06.
Artículo en Inglés | MEDLINE | ID: mdl-30503522

RESUMEN

Diamond-Blackfan anemia (DBA) is a rare bone marrow failure disorder that affects 7 out of 1,000,000 live births and has been associated with mutations in components of the ribosome. In order to characterize the genetic landscape of this heterogeneous disorder, we recruited a cohort of 472 individuals with a clinical diagnosis of DBA and performed whole-exome sequencing (WES). We identified relevant rare and predicted damaging mutations for 78% of individuals. The majority of mutations were singletons, absent from population databases, predicted to cause loss of function, and located in 1 of 19 previously reported ribosomal protein (RP)-encoding genes. Using exon coverage estimates, we identified and validated 31 deletions in RP genes. We also observed an enrichment for extended splice site mutations and validated their diverse effects using RNA sequencing in cell lines obtained from individuals with DBA. Leveraging the size of our cohort, we observed robust genotype-phenotype associations with congenital abnormalities and treatment outcomes. We further identified rare mutations in seven previously unreported RP genes that may cause DBA, as well as several distinct disorders that appear to phenocopy DBA, including nine individuals with biallelic CECR1 mutations that result in deficiency of ADA2. However, no new genes were identified at exome-wide significance, suggesting that there are no unidentified genes containing mutations readily identified by WES that explain >5% of DBA-affected case subjects. Overall, this report should inform not only clinical practice for DBA-affected individuals, but also the design and analysis of rare variant studies for heterogeneous Mendelian disorders.


Asunto(s)
Anemia de Diamond-Blackfan/genética , Adolescente , Niño , Preescolar , Estudios de Cohortes , Exoma/genética , Exones/genética , Femenino , Eliminación de Gen , Estudios de Asociación Genética/métodos , Humanos , Péptidos y Proteínas de Señalización Intercelular/genética , Masculino , Mutación/genética , Fenotipo , Proteínas Ribosómicas/genética , Ribosomas/genética , Análisis de Secuencia de ARN/métodos , Secuenciación del Exoma/métodos
2.
Pediatr Blood Cancer ; 67(12): e28748, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-33025707

RESUMEN

BACKGROUND: Diamond-Blackfan anemia (DBA) is an inherited bone marrow failure syndrome characterized by anemia, short stature, congenital anomalies, and cancer predisposition. Most cases are due to mutations in genes encoding ribosomal proteins (RP) leading to RP haploinsufficiency. Effective treatments for the anemia of DBA include chronic red cell transfusions, long-term corticosteroid therapy, or hematopoietic stem cell transplantation. In a small patient series and in animal models, there have been hematologic responses to L-leucine with amelioration of anemia. The study objectives of this clinical trial were to determine feasibility, safety, and efficacy of L-leucine in transfusion-dependent patients with DBA. PROCEDURE: Patients ≥2 years of age received L-leucine 700 mg/m2 orally three times daily for nine months to determine a hematologic response and any improvement in growth (NCT01362595). RESULTS: This multicenter, phase I/II study enrolled 55 subjects; 43 were evaluable. There were 21 males; the median age at enrollment was 10.4 years (range, 2.5-46.1 years). No significant adverse events were attributable to L-leucine. Two subjects had a complete erythroid response and five had a partial response. Nine of 25, and 11 of 25, subjects experienced a positive weight and height percentile change, respectively, at the end of therapy. CONCLUSIONS: L-leucine is safe, resulted in an erythroid response in 16% of subjects with DBA, and led to an increase in weight and linear growth velocity in 36% and 44% of evaluable subjects, respectively. Further studies will be critical to understand the role of L-leucine in the management of patients with DBA.


Asunto(s)
Anemia de Diamond-Blackfan/terapia , Transfusión Sanguínea/métodos , Leucina/uso terapéutico , Adolescente , Adulto , Anemia de Diamond-Blackfan/patología , Niño , Preescolar , Terapia Combinada , Estudios de Factibilidad , Femenino , Estudios de Seguimiento , Humanos , Masculino , Persona de Mediana Edad , Proyectos Piloto , Pronóstico , Adulto Joven
3.
Haematologica ; 103(12): 2008-2015, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-30026338

RESUMEN

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.


Asunto(s)
Acidosis Láctica/genética , Anemia Sideroblástica/genética , Enfermedades Genéticas Ligadas al Cromosoma X/genética , Mutación de Línea Germinal , Síndrome MELAS/genética , Proteínas Mitocondriales/genética , Tirosina-ARNt Ligasa/genética , Acidosis Láctica/enzimología , Adolescente , Anemia Sideroblástica/enzimología , Femenino , Estudios de Asociación Genética , Enfermedades Genéticas Ligadas al Cromosoma X/enzimología , Humanos , Lactante , Síndrome MELAS/enzimología , Masculino , Persona de Mediana Edad , Mutación Missense , Adulto Joven
6.
Pediatr Blood Cancer ; 61(5): 869-74, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24285674

RESUMEN

BACKGROUND: Randomized clinical trials in pediatric aplastic anemia (AA) are rare and data to guide standards of care are scarce. PROCEDURE: Eighteen pediatric institutions formed the North American Pediatric Aplastic Anemia Consortium to foster collaborative studies in AA. The initial goal of NAPAAC was to survey the diagnostic studies and therapies utilized in AA. RESULTS: Our survey indicates considerable variability among institutions in the diagnosis and treatment of AA. There were areas of general consensus, including the need for a bone marrow evaluation, cytogenetic and specific fluorescent in situ hybridization assays to establish diagnosis and exclude genetic etiologies with many institutions requiring results prior to initiation of immunosuppressive therapy (IST); uniform referral for hematopoietic stem cell transplantation as first line therapy if an HLA-identical sibling is identified; the use of first-line IST containing horse anti-thymocyte globulin and cyclosporine A (CSA) if an HLA-identical sibling donor is not identified; supportive care measures; and slow taper of CSA after response. Areas of controversy included the need for telomere length results prior to IST, the time after IST initiation defining a treatment failure; use of hematopoietic growth factors; the preferred rescue therapy after failure of IST; the use of specific hemoglobin and platelet levels as triggers for transfusion support; the use of prophylactic antibiotics; and follow-up monitoring after completion of treatment. CONCLUSIONS: These initial survey results reflect heterogeneity in diagnosis and care amongst pediatric centers and emphasize the need to develop evidence-based diagnosis and treatment approaches in this rare disease.


Asunto(s)
Anemia Aplásica/diagnóstico , Anemia Aplásica/tratamiento farmacológico , Encuestas de Atención de la Salud , Inmunosupresores/uso terapéutico , Pautas de la Práctica en Medicina , Adolescente , Anemia Aplásica/epidemiología , Suero Antilinfocítico/uso terapéutico , Niño , Preescolar , Ciclosporina/uso terapéutico , Femenino , Estudios de Seguimiento , Humanos , Lactante , Masculino , América del Norte/epidemiología , Pronóstico , Hermanos
7.
Hum Genet ; 132(11): 1265-74, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-23812780

RESUMEN

Diamond-Blackfan anemia (DBA) is an inherited red blood cell aplasia that usually presents during the first year of life. The main features of the disease are normochromic and macrocytic anemia, reticulocytopenia, and nearly absent erythroid progenitors in the bone marrow. The patients also present with growth retardation and craniofacial, upper limb, heart and urinary system congenital malformations in ~30-50 % of cases. The disease has been associated with point mutations and large deletions in ten ribosomal protein (RP) genes RPS19, RPS24, RPS17, RPL35A, RPL5, RPL11, RPS7, RPS10, RPS26, and RPL26 and GATA1 in about 60-65 % of patients. Here, we report a novel large deletion in RPL15, a gene not previously implicated to be causative in DBA. Like RPL26, RPL15 presents the distinctive feature of being required both for 60S subunit formation and for efficient cleavage of the internal transcribed spacer 1. In addition, we detected five deletions in RP genes in which mutations have been previously shown to cause DBA: one each in RPS19, RPS24, and RPS26, and two in RPS17. Pre-ribosomal RNA processing was affected in cells established from the patients bearing these deletions, suggesting a possible molecular basis for their pathological effect. These data identify RPL15 as a new gene involved in DBA and further support the presence of large deletions in RP genes in DBA patients.


Asunto(s)
Anemia de Diamond-Blackfan/genética , Eliminación de Gen , Proteínas Ribosómicas/genética , Hibridación Genómica Comparativa , Variaciones en el Número de Copia de ADN , Técnicas de Silenciamiento del Gen , Células HeLa , Humanos , Mutación , ARN Ribosómico/análisis , ARN Ribosómico/genética , ARN Interferente Pequeño , Proteínas Ribosómicas/metabolismo
8.
Am J Hum Genet ; 86(2): 222-8, 2010 Feb 12.
Artículo en Inglés | MEDLINE | ID: mdl-20116044

RESUMEN

Diamond-Blackfan anemia (DBA), an inherited bone marrow failure syndrome characterized by anemia that usually presents before the first birthday or in early childhood, is associated with birth defects and an increased risk of cancer. Although anemia is the most prominent feature of DBA, the disease is also characterized by growth retardation and congenital malformations, in particular craniofacial, upper limb, heart, and urinary system defects that are present in approximately 30%-50% of patients. DBA has been associated with mutations in seven ribosomal protein (RP) genes, RPS19, RPS24, RPS17, RPL35A, RPL5, RPL11, and RPS7, in about 43% of patients. To continue our large-scale screen of RP genes in a DBA population, we sequenced 35 ribosomal protein genes, RPL15, RPL24, RPL29, RPL32, RPL34, RPL9, RPL37, RPS14, RPS23, RPL10A, RPS10, RPS12, RPS18, RPL30, RPS20, RPL12, RPL7A, RPS6, RPL27A, RPLP2, RPS25, RPS3, RPL41, RPL6, RPLP0, RPS26, RPL21, RPL36AL, RPS29, RPL4, RPLP1, RPL13, RPS15A, RPS2, and RPL38, in our DBA patient cohort of 117 probands. We identified three distinct mutations of RPS10 in five probands and nine distinct mutations of RPS26 in 12 probands. Pre-rRNA analysis in lymphoblastoid cells from patients bearing mutations in RPS10 and RPS26 showed elevated levels of 18S-E pre-rRNA. This accumulation is consistent with the phenotype observed in HeLa cells after knockdown of RPS10 or RPS26 expression with siRNAs, which indicates that mutations in the RPS10 and RPS26 genes in DBA patients affect the function of the proteins in rRNA processing.


Asunto(s)
Anemia de Diamond-Blackfan/genética , Mutación/genética , Proteínas Ribosómicas/genética , Secuencia de Bases , Humanos , Procesamiento Postranscripcional del ARN
10.
Hum Mutat ; 33(7): 1037-44, 2012 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-22431104

RESUMEN

Diamond-Blackfan anemia (DBA) is an inherited form of pure red cell aplasia that usually presents in infancy or early childhood and is associated with congenital malformations in ∼30-50% of patients. DBA has been associated with mutations in nine ribosomal protein (RP) genes in about 53% of patients. We completed a large-scale screen of 79 RP genes by sequencing 16 RP genes (RPL3, RPL7, RPL8, RPL10, RPL14, RPL17, RPL19, RPL23A, RPL26, RPL27, RPL35, RPL36A, RPL39, RPS4X, RPS4Y1, and RPS21) in 96 DBA probands. We identified a de novo two-nucleotide deletion in RPL26 in one proband associated with multiple severe physical abnormalities. This mutation gives rise to a remarkable ribosome biogenesis defect that affects maturation of both the small and the large subunits. We also found a deletion in RPL19 and missense mutations in RPL3 and RPL23A, which may be variants of unknown significance. Together with RPL5, RPL11, and RPS7, RPL26 is the fourth RP regulating p53 activity that is linked to DBA.


Asunto(s)
Anomalías Múltiples/genética , Anemia de Diamond-Blackfan/genética , Anemia de Diamond-Blackfan/metabolismo , Mutación del Sistema de Lectura/genética , ARN Ribosómico/genética , Proteínas Ribosómicas/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Northern Blotting , Western Blotting , Células HeLa , Humanos , ARN Interferente Pequeño , Proteína Ribosomal L3 , Proteínas Ribosómicas/genética , Proteína p53 Supresora de Tumor/genética
11.
Am J Hum Genet ; 83(6): 769-80, 2008 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-19061985

RESUMEN

Diamond-Blackfan anemia (DBA), a congenital bone-marrow-failure syndrome, is characterized by red blood cell aplasia, macrocytic anemia, clinical heterogeneity, and increased risk of malignancy. Although anemia is the most prominent feature of DBA, the disease is also characterized by growth retardation and congenital anomalies that are present in approximately 30%-50% of patients. The disease has been associated with mutations in four ribosomal protein (RP) genes, RPS19, RPS24, RPS17, and RPL35A, in about 30% of patients. However, the genetic basis of the remaining 70% of cases is still unknown. Here, we report the second known mutation in RPS17 and probable pathogenic mutations in three more RP genes, RPL5, RPL11, and RPS7. In addition, we identified rare variants of unknown significance in three other genes, RPL36, RPS15, and RPS27A. Remarkably, careful review of the clinical data showed that mutations in RPL5 are associated with multiple physical abnormalities, including craniofacial, thumb, and heart anomalies, whereas isolated thumb malformations are predominantly present in patients carrying mutations in RPL11. We also demonstrate that mutations of RPL5, RPL11, or RPS7 in DBA cells is associated with diverse defects in the maturation of ribosomal RNAs in the large or the small ribosomal subunit production pathway, expanding the repertoire of ribosomal RNA processing defects associated with DBA.


Asunto(s)
Anemia de Diamond-Blackfan/genética , Fisura del Paladar/genética , Mutación , Proteínas Ribosómicas/genética , Pulgar/anomalías , Humanos , Subunidades Ribosómicas Grandes/genética , Subunidades Ribosómicas Pequeñas/genética
12.
Br J Haematol ; 148(4): 611-22, 2010 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-19958353

RESUMEN

Diamond Blackfan anaemia (DBA) is a severe congenital failure of erythropoiesis. Despite mutations in one of several ribosome protein genes, including RPS19, the cause of the erythroid specificity is still a mystery. We hypothesized that, because the chromatin of late erythroid cells becomes condensed and transcriptionally inactive prior to enucleation, the rapidly proliferating immature cells require very high ribosome synthetic rates. RNA biogenesis was measured in primary mouse fetal liver erythroid progenitor cells; during the first 24 h, cell number increased three to fourfold while, remarkably, RNA content increased sixfold, suggesting an accumulation of an excess of ribosomes during early erythropoiesis. Retrovirus infected siRNA RPS19 knockdown cells showed reduced proliferation but normal differentiation, and cell cycle analysis showed a G1/S phase delay. p53 protein was increased in the knockdown cells, and the mRNA level for p21, a transcriptional target of p53, was increased. Furthermore, we show that RPS19 knockdown decreased MYB protein, and Kit mRNA was reduced, as was the amount of cell surface KIT protein. Thus, in this small hairpin RNA murine model of DBA, RPS19 insufficient erythroid cells may proliferate poorly because of p53-mediated cell cycle arrest, and also because of decreased expression of the key erythroid signalling protein KIT.


Asunto(s)
Anemia de Diamond-Blackfan/genética , Eritropoyesis/genética , Anemia de Diamond-Blackfan/patología , Anemia de Diamond-Blackfan/fisiopatología , Diferenciación Celular/genética , Proliferación Celular , Células Cultivadas , Fase G1/genética , Técnicas de Silenciamiento del Gen , Genes myb , Humanos , Hígado/citología , Hígado/embriología , ARN/biosíntesis , ARN Interferente Pequeño/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa/métodos , Proteínas Ribosómicas/genética , Proteína p53 Supresora de Tumor/metabolismo
13.
Blood ; 112(5): 1582-92, 2008 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-18535205

RESUMEN

Diamond-Blackfan anemia (DBA) is an inherited bone marrow failure syndrome characterized by anemia, congenital abnormalities, and cancer predisposition. Small ribosomal subunit genes RPS19, RPS24, and RPS17 are mutated in approximately one-third of patients. We used a candidate gene strategy combining high-resolution genomic mapping and gene expression microarray in the analysis of 2 DBA patients with chromosome 3q deletions to identify RPL35A as a potential DBA gene. Sequence analysis of a cohort of DBA probands confirmed involvement RPL35A in DBA. shRNA inhibition shows that Rpl35a is essential for maturation of 28S and 5.8S rRNAs, 60S subunit biogenesis, normal proliferation, and cell survival. Analysis of pre-rRNA processing in primary DBA lymphoblastoid cell lines demonstrated similar alterations of large ribosomal subunit rRNA in both RPL35A-mutated and some RPL35A wild-type patients, suggesting additional large ribosomal subunit gene defects are likely present in some cases of DBA. These data demonstrate that alterations of large ribosomal subunit proteins cause DBA and support the hypothesis that DBA is primarily the result of altered ribosomal function. The results also establish that haploinsufficiency of large ribosomal subunit proteins contributes to bone marrow failure and potentially cancer predisposition.


Asunto(s)
Anemia de Diamond-Blackfan/genética , Mutación , Proteínas Ribosómicas/genética , Secuencia de Aminoácidos , Anemia de Diamond-Blackfan/metabolismo , Anemia de Diamond-Blackfan/patología , Apoptosis/genética , Secuencia de Bases , Estudios de Casos y Controles , Línea Celular , Proliferación Celular , Deleción Cromosómica , Mapeo Cromosómico , Cromosomas Humanos Par 3/genética , Estudios de Cohortes , ADN/genética , Femenino , Humanos , Lactante , Masculino , Datos de Secuencia Molecular , Análisis de Secuencia por Matrices de Oligonucleótidos , Linaje , Procesamiento Postranscripcional del ARN , ARN Interferente Pequeño/genética , Proteínas Ribosómicas/metabolismo , Ribosomas/metabolismo
14.
Am J Hematol ; 85(2): 111-6, 2010 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-20054847

RESUMEN

Mutations in the RPS19 gene have been identified in 25% of individuals affected by Diamond-Blackfan anemia (DBA), a congenital erythroblastopenia characterized by an aregenerative anemia and a variety of malformations. More than 60 mutations in the five coding exons of RPS19 have been described to date. We previously reported a mutation (c.-1 + 26G>T) and an insertion at -631 upstream of ATG (c.-147_-146insGCCA) in the noncoding region. Because DBA phenotype is extremely heterogeneous from silent to severe and because haploinsufficiency seems to play a role in this process, it is likely that genetic variations in the noncoding regions affecting translation of RPS19 can modulate the phenotypic expression of DBA. However, to date, very few studies have addressed this question comprehensively. In this study, we performed detailed sequence analysis of the RPS19 gene in 239 patients with DBA and 110 of their relatives. We found that 6.2% of the patients with DBA carried allelic variations upstream of ATG: 3.3% with c.-1 + 26G>T; 2.5% with c.-147_-146insGCCA; and 0.4% with c.-174G>A. Interestingly, the c.-147_-146insGCCA, which has been found in a black American and French Caribbean control population, was not found in 500 Caucasian control chromosomes we studied. However, it was found in association with the same haplotype distribution of four intronic polymorphisms in our patients with DBA. Although a polymorphism, the frequency of this variant in the patients with DBA and its association with the same haplotype raises the possibility that this polymorphism and the other genetic variations in the noncoding region could play a role in DBA pathogenesis.


Asunto(s)
Anemia de Diamond-Blackfan/genética , Exones/genética , Mutación , Polimorfismo Genético , Proteínas Ribosómicas/genética , Femenino , Humanos , Masculino , Fenotipo
15.
JBJS Case Connect ; 9(2): e0376, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31085938

RESUMEN

CASE: We report on a patient with heterozygous prothrombin G20210A mutation (PTM) and a history of venous thromboembolism (VTE) after knee arthroscopy, who was undergoing hip arthroscopy. This common mutation has an overall prevalence of 2.0% and results in a 280% to 420% likelihood of thrombosis compared to patients without the mutation. CONCLUSIONS: Hip arthroscopy is associated with a low risk of VTE with currently no guidelines for thromboprophylaxis. Patients should be managed in relation to their risk factors utilizing a collaborative, individualized approach. Treatment with short-term low-molecular-weight heparin resulted in no thromboembolism at 18-month follow-up for this patient with PTM.


Asunto(s)
Artroscopía/efectos adversos , Articulación de la Cadera/diagnóstico por imagen , Atención Perioperativa/métodos , Trombofilia/genética , Adolescente , Cuidados Posteriores , Anticoagulantes/uso terapéutico , Artroscopía/métodos , Femenino , Heparina de Bajo-Peso-Molecular/uso terapéutico , Heterocigoto , Articulación de la Cadera/patología , Articulación de la Cadera/cirugía , Humanos , Prácticas Interdisciplinarias/métodos , Mutación/genética , Factores de Riesgo , Trombofilia/tratamiento farmacológico , Trombofilia/epidemiología , Resultado del Tratamiento , Tromboembolia Venosa/etiología , Tromboembolia Venosa/prevención & control
16.
J Clin Invest ; 129(9): 3821-3826, 2019 06 18.
Artículo en Inglés | MEDLINE | ID: mdl-31211692

RESUMEN

Shwachman-Diamond Syndrome (SDS) is a rare and clinically-heterogeneous bone marrow (BM) failure syndrome caused by mutations in the Shwachman-Bodian-Diamond Syndrome (SBDS) gene. Although SDS was described over 50 years ago, the molecular pathogenesis is poorly understood due, in part, to the rarity and heterogeneity of the affected hematopoietic progenitors. To address this, we used single cell RNA sequencing to profile scant hematopoietic stem and progenitor cells from SDS patients. We generated a single cell map of early lineage commitment and found that SDS hematopoiesis was left-shifted with selective loss of granulocyte-monocyte progenitors. Transcriptional targets of transforming growth factor-beta (TGFß) were dysregulated in SDS hematopoietic stem cells and multipotent progenitors, but not in lineage-committed progenitors. TGFß inhibitors (AVID200 and SD208) increased hematopoietic colony formation of SDS patient BM. Finally, TGFß3 and other TGFß pathway members were elevated in SDS patient blood plasma. These data establish the TGFß pathway as a novel candidate biomarker and therapeutic target in SDS and translate insights from single cell biology into a potential therapy.


Asunto(s)
Médula Ósea/fisiopatología , Células Madre Hematopoyéticas/patología , Síndrome de Shwachman-Diamond/fisiopatología , Factor de Crecimiento Transformador beta1/metabolismo , Adolescente , Adulto , Antígenos CD34/metabolismo , Diferenciación Celular , Linaje de la Célula , Niño , Granulocitos/citología , Hematopoyesis , Humanos , Inflamación , Monocitos/citología , Mutación , Fosforilación , Análisis de Secuencia de ARN , Transducción de Señal , Factor de Crecimiento Transformador beta/metabolismo , Adulto Joven
17.
J Clin Invest ; 129(7): 2878-2887, 2019 04 30.
Artículo en Inglés | MEDLINE | ID: mdl-31038472

RESUMEN

The etiology of severe hemolytic anemia in most patients with recessive hereditary spherocytosis (rHS) and the related disorder hereditary pyropoikilocytosis (HPP) is unknown. Whole exome sequencing of DNA from probands of 24 rHS or HPP kindreds identified numerous mutations in erythrocyte membrane α-spectrin (SPTA1). Twenty-eight mutations were novel, with null alleles frequently found in trans to missense mutations. No mutations were identified in a third of SPTA1 alleles (17/48). Whole genome sequencing revealed linkage disequilibrium between the common rHS-linked α-spectrinBug Hill polymorphism and a rare intron 30 variant in all 17 mutation-negative alleles. In vitro minigene studies and in vivo splicing analyses revealed the intron 30 variant changes a weak alternate branch point (BP) to a strong BP. This change leads to increased utilization of an alternate 3' splice acceptor site, perturbing normal α-spectrin mRNA splicing and creating an elongated mRNA transcript. In vivo mRNA stability studies revealed the newly created termination codon in the elongated transcript activates nonsense mediated decay leading to spectrin deficiency. These results demonstrate a unique mechanism of human genetic disease contributes to the etiology of a third of cases of rHS, facilitating diagnosis and treatment of severe anemia, and identifying a new target for therapeutic manipulation.


Asunto(s)
Anemia Hemolítica Congénita , Membrana Eritrocítica , Mutación Missense , Sitios de Empalme de ARN , Empalme del ARN/genética , Espectrina , Anemia Hemolítica Congénita/genética , Anemia Hemolítica Congénita/metabolismo , Anemia Hemolítica Congénita/patología , Membrana Eritrocítica/genética , Membrana Eritrocítica/metabolismo , Membrana Eritrocítica/patología , Femenino , Humanos , Masculino , Espectrina/biosíntesis , Espectrina/genética
18.
Hematol Oncol Clin North Am ; 32(4): 569-580, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-30047411

RESUMEN

Acquired aplastic anemia and inherited bone marrow failure syndromes both present with pancytopenia and must be distinguished because they have differences in treatment decisions and continued monitoring requirements. Advances in the genetic interrogation of patient samples have led to identification of inherited germline diseases and appreciation that patients with inherited bone marrow failure disorders may be normal in appearance with few expected clinical clues. Somatic mutations in aplastic anemia may have prognostic value. Hematopoietic stem cells from inherited marrow failure diseases can correct the proliferative defect and may develop further somatic mutations that progress to myelodysplastic syndrome or acute myeloid leukemia.


Asunto(s)
Anemia Aplásica , Enfermedades de la Médula Ósea , Médula Ósea , Enfermedades Genéticas Congénitas , Hemoglobinuria Paroxística , Leucemia Mieloide Aguda , Mutación , Síndromes Mielodisplásicos , Anemia Aplásica/genética , Anemia Aplásica/metabolismo , Anemia Aplásica/patología , Médula Ósea/metabolismo , Médula Ósea/patología , Enfermedades de la Médula Ósea/genética , Enfermedades de la Médula Ósea/metabolismo , Enfermedades de la Médula Ósea/patología , Trastornos de Fallo de la Médula Ósea , Enfermedades Genéticas Congénitas/genética , Enfermedades Genéticas Congénitas/metabolismo , Enfermedades Genéticas Congénitas/patología , Hemoglobinuria Paroxística/genética , Hemoglobinuria Paroxística/metabolismo , Hemoglobinuria Paroxística/patología , Humanos , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/metabolismo , Leucemia Mieloide Aguda/patología , Síndromes Mielodisplásicos/genética , Síndromes Mielodisplásicos/metabolismo , Síndromes Mielodisplásicos/patología
19.
Hematol Oncol Clin North Am ; 32(4): 701-712, 2018 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-30047421

RESUMEN

Diamond-Blackfan anemia (DBA) is a severe congenital hypoplastic anemia caused by mutation in a ribosomal protein gene. Major clinical issues concern the optimal management of patients resistant to steroids, the first-line therapy. Hematopoietic stem cell transplantation is indicated in young patients with an HLA-matched unaffected sibling donor, and recent results with matched unrelated donor transplants indicate that these patients also do well. When neither steroids nor a transplant is possible red cell transfusions are required, and iron loading is rapid in some DBA patients, so effective chelation is vital. Also discussed are novel treatments under investigation for DBA.


Asunto(s)
Anemia de Diamond-Blackfan , Transfusión de Eritrocitos , Trasplante de Células Madre Hematopoyéticas , Mutación , Donantes de Tejidos , Aloinjertos , Anemia de Diamond-Blackfan/genética , Anemia de Diamond-Blackfan/metabolismo , Anemia de Diamond-Blackfan/patología , Anemia de Diamond-Blackfan/terapia , Humanos , Hermanos
20.
Sci Transl Med ; 9(376)2017 02 08.
Artículo en Inglés | MEDLINE | ID: mdl-28179501

RESUMEN

Diamond-Blackfan anemia (DBA) is a congenital disorder characterized by the failure of erythroid progenitor differentiation, severely curtailing red blood cell production. Because many DBA patients fail to respond to corticosteroid therapy, there is considerable need for therapeutics for this disorder. Identifying therapeutics for DBA requires circumventing the paucity of primary patient blood stem and progenitor cells. To this end, we adopted a reprogramming strategy to generate expandable hematopoietic progenitor cells from induced pluripotent stem cells (iPSCs) from DBA patients. Reprogrammed DBA progenitors recapitulate defects in erythroid differentiation, which were rescued by gene complementation. Unbiased chemical screens identified SMER28, a small-molecule inducer of autophagy, which enhanced erythropoiesis in a range of in vitro and in vivo models of DBA. SMER28 acted through autophagy factor ATG5 to stimulate erythropoiesis and up-regulate expression of globin genes. These findings present an unbiased drug screen for hematological disease using iPSCs and identify autophagy as a therapeutic pathway in DBA.


Asunto(s)
Anemia de Diamond-Blackfan/tratamiento farmacológico , Descubrimiento de Drogas , Células Madre Hematopoyéticas/metabolismo , Compuestos Alílicos/farmacología , Anemia de Diamond-Blackfan/patología , Antígenos CD34/metabolismo , Autofagia/efectos de los fármacos , Proteína 5 Relacionada con la Autofagia/metabolismo , Diferenciación Celular/efectos de los fármacos , Reprogramación Celular , Células Eritroides/efectos de los fármacos , Células Eritroides/patología , Eritropoyesis/efectos de los fármacos , Prueba de Complementación Genética , Globinas/metabolismo , Trasplante de Células Madre Hematopoyéticas , Células Madre Hematopoyéticas/efectos de los fármacos , Humanos , Células Madre Pluripotentes Inducidas/efectos de los fármacos , Células Madre Pluripotentes Inducidas/metabolismo , Quinazolinas/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA