Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 80
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Cell ; 186(11): 2456-2474.e24, 2023 05 25.
Artículo en Inglés | MEDLINE | ID: mdl-37137305

RESUMEN

Systematic evaluation of the impact of genetic variants is critical for the study and treatment of human physiology and disease. While specific mutations can be introduced by genome engineering, we still lack scalable approaches that are applicable to the important setting of primary cells, such as blood and immune cells. Here, we describe the development of massively parallel base-editing screens in human hematopoietic stem and progenitor cells. Such approaches enable functional screens for variant effects across any hematopoietic differentiation state. Moreover, they allow for rich phenotyping through single-cell RNA sequencing readouts and separately for characterization of editing outcomes through pooled single-cell genotyping. We efficiently design improved leukemia immunotherapy approaches, comprehensively identify non-coding variants modulating fetal hemoglobin expression, define mechanisms regulating hematopoietic differentiation, and probe the pathogenicity of uncharacterized disease-associated variants. These strategies will advance effective and high-throughput variant-to-function mapping in human hematopoiesis to identify the causes of diverse diseases.


Asunto(s)
Edición Génica , Células Madre Hematopoyéticas , Humanos , Diferenciación Celular , Sistemas CRISPR-Cas , Genoma , Hematopoyesis , Células Madre Hematopoyéticas/metabolismo , Ingeniería Genética , Análisis de la Célula Individual
2.
Br J Haematol ; 204(2): 595-605, 2024 02.
Artículo en Inglés | MEDLINE | ID: mdl-37945316

RESUMEN

Juvenile myelomonocytic leukaemia (JMML) is characterized by gene variants that deregulate the RAS signalling pathway. Children with neurofibromatosis type 1 (NF-1) carry a defective NF1 allele in the germline and are predisposed to JMML, which presumably requires somatic inactivation of the NF1 wild-type allele. Here we examined the two-hit concept in leukaemic cells of 25 patients with JMML and NF-1. Ten patients with JMML/NF-1 exhibited a NF1 loss-of-function variant in combination with uniparental disomy of the 17q arm. Five had NF1 microdeletions combined with a pathogenic NF1 variant and nine carried two compound-heterozygous NF1 variants. We also examined 16 patients without clinical signs of NF-1 and no variation in the JMML-associated driver genes PTPN11, KRAS, NRAS or CBL (JMML-5neg) and identified eight patients with NF1 variants. Three patients had microdeletions combined with hemizygous NF1 variants, three had compound-heterozygous NF1 variants and two had heterozygous NF1 variants. In addition, we found a high incidence of secondary ASXL1 and/or SETBP1 variants in both groups. We conclude that the clinical diagnosis of JMML/NF-1 reliably indicates a NF1-driven JMML subtype, and that careful NF1 analysis should be included in the genetic workup of JMML even in the absence of clinical evidence of NF-1.


Asunto(s)
Leucemia Mielomonocítica Juvenil , Neurofibromatosis 1 , Niño , Humanos , Leucemia Mielomonocítica Juvenil/genética , Neurofibromatosis 1/genética , Mutación , Transducción de Señal , Genes Supresores de Tumor
3.
Blood ; 139(7): 1039-1051, 2022 02 17.
Artículo en Inglés | MEDLINE | ID: mdl-34767620

RESUMEN

Human telomere biology disorders (TBD)/short telomere syndromes (STS) are heterogeneous disorders caused by inherited loss-of-function mutations in telomere-associated genes. Here, we identify 3 germline heterozygous missense variants in the RPA1 gene in 4 unrelated probands presenting with short telomeres and varying clinical features of TBD/STS, including bone marrow failure, myelodysplastic syndrome, T- and B-cell lymphopenia, pulmonary fibrosis, or skin manifestations. All variants cluster to DNA-binding domain A of RPA1 protein. RPA1 is a single-strand DNA-binding protein required for DNA replication and repair and involved in telomere maintenance. We showed that RPA1E240K and RPA1V227A proteins exhibit increased binding to single-strand and telomeric DNA, implying a gain in DNA-binding function, whereas RPA1T270A has binding properties similar to wild-type protein. To study the mutational effect in a cellular system, CRISPR/Cas9 was used to knock-in the RPA1E240K mutation into healthy inducible pluripotent stem cells. This resulted in severe telomere shortening and impaired hematopoietic differentiation. Furthermore, in patients with RPA1E240K, we discovered somatic genetic rescue in hematopoietic cells due to an acquired truncating cis RPA1 mutation or a uniparental isodisomy 17p with loss of mutant allele, coinciding with stabilized blood counts. Using single-cell sequencing, the 2 somatic genetic rescue events were proven to be independently acquired in hematopoietic stem cells. In summary, we describe the first human disease caused by germline RPA1 variants in individuals with TBD/STS.


Asunto(s)
Trastornos de Fallo de la Médula Ósea/patología , Mutación con Ganancia de Función , Heterocigoto , Síndromes Mielodisplásicos/patología , Proteína de Replicación A/genética , Acortamiento del Telómero , Telómero/genética , Adolescente , Adulto , Trastornos de Fallo de la Médula Ósea/etiología , Trastornos de Fallo de la Médula Ósea/metabolismo , Diferenciación Celular , Niño , Femenino , Humanos , Recién Nacido , Masculino , Persona de Mediana Edad , Síndromes Mielodisplásicos/etiología , Síndromes Mielodisplásicos/metabolismo , Adulto Joven
5.
Haematologica ; 109(2): 422-430, 2024 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-37584291

RESUMEN

Monosomy 7 is the most common cytogenetic abnormality in pediatric myelodysplastic syndrome (MDS) and associated with a high risk of disease progression. However, in young children, spontaneous loss of monosomy 7 with concomitant hematologic recovery has been described, especially in the presence of germline mutations in SAMD9 and SAMD9L genes. Here, we report on our experience of close surveillance instead of upfront hematopoietic stem cell transplantation (HSCT) in seven patients diagnosed with SAMD9L syndrome and monosomy 7 at a median age of 0.6 years (range, 0.4-2.9). Within 14 months from diagnosis, three children experienced spontaneous hematological remission accompanied by a decrease in monosomy 7 clone size. Subclones with somatic SAMD9L mutations in cis were identified in five patients, three of whom attained hematological remission. Two patients acquired RUNX1 and EZH2 mutations during the observation period, of whom one progressed to myelodysplastic syndrome with excess of blasts (MDS-EB). Four patients underwent allogeneic HSCT at a median time of 26 months (range, 14-40) from diagnosis for MDSEB, necrotizing granulomatous lymphadenitis, persistent monosomy 7, and severe neutropenia. At last follow-up, six patients were alive, while one passed away due to transplant-related causes. These data confirm previous observations that monosomy 7 can be transient in young children with SAMD9L syndrome. However, they also indicate that delaying HSCT poses a substantial risk of severe infection and disease progression. Finally, surveillance of patients with SAMD9L syndrome and monosomy 7 is critical to define the evolving genetic landscape and to determine the appropriate timing of HSCT (clinicaltrials gov. Identifier: NCT00662090).


Asunto(s)
Deleción Cromosómica , Síndromes Mielodisplásicos , Humanos , Niño , Preescolar , Lactante , Remisión Espontánea , Síndromes Mielodisplásicos/diagnóstico , Síndromes Mielodisplásicos/genética , Síndromes Mielodisplásicos/terapia , Progresión de la Enfermedad , Factores de Transcripción/genética , Monosomía , Cromosomas Humanos Par 7/genética , Péptidos y Proteínas de Señalización Intracelular/genética
6.
Am J Hum Genet ; 104(6): 1040-1059, 2019 06 06.
Artículo en Inglés | MEDLINE | ID: mdl-31079900

RESUMEN

The heterogeneous nuclear ribonucleoprotein (HNRNP) genes code for a set of RNA-binding proteins that function primarily in the spliceosome C complex. Pathogenic variants in these genes can drive neurodegeneration, through a mechanism involving excessive stress-granule formation, or developmental defects, through mechanisms that are not known. Here, we report four unrelated individuals who have truncating or missense variants in the same C-terminal region of hnRNPR and who have multisystem developmental defects including abnormalities of the brain and skeleton, dysmorphic facies, brachydactyly, seizures, and hypoplastic external genitalia. We further identified in the literature a fifth individual with a truncating variant. RNA sequencing of primary fibroblasts reveals that these HNRNPR variants drive significant changes in the expression of several homeobox genes, as well as other transcription factors, such as LHX9, TBX1, and multiple HOX genes, that are considered fundamental regulators of embryonic and gonad development. Higher levels of retained intronic HOX sequences and lost splicing events in the HOX cluster are observed in cells carrying HNRNPR variants, suggesting that impaired splicing is at least partially driving HOX deregulation. At basal levels, stress-granule formation appears normal in primary and transfected cells expressing HNRNPR variants. However, these cells reveal profound recovery defects, where stress granules fail to disassemble properly, after exposure to oxidative stress. This study establishes an essential role for HNRNPR in human development and points to a mechanism that may unify other "spliceosomopathies" linked to variants that drive multi-system congenital defects and are found in hnRNPs.


Asunto(s)
Discapacidades del Desarrollo/etiología , Fibroblastos/patología , Regulación de la Expresión Génica , Genes Homeobox/genética , Ribonucleoproteínas Nucleares Heterogéneas/genética , Mutación , Empalme del ARN/genética , Niño , Preescolar , Discapacidades del Desarrollo/patología , Femenino , Fibroblastos/metabolismo , Humanos , Lactante , Masculino , Estrés Oxidativo , Fenotipo , Secuenciación del Exoma
7.
Br J Haematol ; 199(4): 482-495, 2022 11.
Artículo en Inglés | MEDLINE | ID: mdl-35753998

RESUMEN

The importance of predisposition to leukaemia in clinical practice is being increasingly recognized. This is emphasized by the establishment of a novel WHO disease category in 2016 called "myeloid neoplasms with germline predisposition". A major syndrome within this group is GATA2 deficiency, a heterogeneous immunodeficiency syndrome with a very high lifetime risk to develop myelodysplastic syndrome (MDS) and acute myeloid leukaemia (AML). GATA2 deficiency has been identified as the most common hereditary cause of MDS in adolescents with monosomy 7. Allogenic haematopoietic stem cell transplantation is the only curative option; however, chances of survival decrease with progression of immunodeficiency and MDS evolution. Penetrance and expressivity within families carrying GATA2 mutations is often variable, suggesting that co-operating extrinsic events are required to trigger the disease. Predictive tools are lacking, and intrafamilial heterogeneity is poorly understood; hence there is a clear unmet medical need. On behalf of the ERAPerMed GATA2 HuMo consortium, in this review we describe the genetic, clinical, and biological aspects of familial GATA2-related MDS, highlighting the importance of developing robust disease preclinical models to improve early detection and clinical decision-making of GATA2 carriers.


Asunto(s)
Deficiencia GATA2 , Trasplante de Células Madre Hematopoyéticas , Síndromes de Inmunodeficiencia , Leucemia Mieloide Aguda , Síndromes Mielodisplásicos , Trastornos Mieloproliferativos , Humanos , Susceptibilidad a Enfermedades , Deficiencia GATA2/genética , Deficiencia GATA2/terapia , Factor de Transcripción GATA2/genética , Síndromes de Inmunodeficiencia/genética , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/terapia , Síndromes Mielodisplásicos/genética , Síndromes Mielodisplásicos/terapia , Trastornos Mieloproliferativos/complicaciones
8.
Nucleic Acids Res ; 48(2): 770-787, 2020 01 24.
Artículo en Inglés | MEDLINE | ID: mdl-31799629

RESUMEN

Variants in ribosomal protein (RP) genes drive Diamond-Blackfan anemia (DBA), a bone marrow failure syndrome that can also predispose individuals to cancer. Inherited and sporadic RP gene variants are also linked to a variety of phenotypes, including malignancy, in individuals with no anemia. Here we report an individual diagnosed with DBA carrying a variant in the 5'UTR of RPL9 (uL6). Additionally, we report two individuals from a family with multiple cancer incidences carrying a RPL9 missense variant. Analysis of cells from these individuals reveals that despite the variants both driving pre-rRNA processing defects and 80S monosome reduction, the downstream effects are remarkably different. Cells carrying the 5'UTR variant stabilize TP53 and impair the growth and differentiation of erythroid cells. In contrast, ribosomes incorporating the missense variant erroneously read through UAG and UGA stop codons of mRNAs. Metabolic profiles of cells carrying the 5'UTR variant reveal an increased metabolism of amino acids and a switch from glycolysis to gluconeogenesis while those of cells carrying the missense variant reveal a depletion of nucleotide pools. These findings indicate that variants in the same RP gene can drive similar ribosome biogenesis defects yet still have markedly different downstream consequences and clinical impacts.


Asunto(s)
Anemia de Diamond-Blackfan/genética , Procesamiento Postranscripcional del ARN/genética , Proteínas Ribosómicas/genética , Ribosomas/genética , Regiones no Traducidas 5'/genética , Adolescente , Adulto , Anemia de Diamond-Blackfan/patología , Carcinoma de Células Escamosas/genética , Carcinoma de Células Escamosas/patología , Niño , Células Eritroides , Femenino , Humanos , Masculino , Mutación/genética , Precursores del ARN/genética , ARN Mensajero/genética , Secuenciación del Exoma
9.
Br J Haematol ; 193(3): 669-673, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-32744739

RESUMEN

Dyskeratosis Congenita (DKC) is a systemic disorder caused by mutations resulting in impaired telomere maintenance. Clinical features include bone marrow failure and an increased risk of developing hematological malignancies. There are conflicting data whether androgen derivatives (AD) can elongate telomeres in vivo and whether AD treatment enhances the risk of gaining myelodysplastic syndrome-related mutations. Seven TERC or TERT-mutated DKC patients underwent AD treatment. All patients revealed hematological response. Telomere length of lymphocytes and granulocytes increased significantly and no MDS-related mutations were detected. Pending longer follow-up, treatment with AD seems to represent an efficient and safe therapy for DKC patients.


Asunto(s)
Andrógenos/farmacología , Disqueratosis Congénita/sangre , Homeostasis del Telómero/efectos de los fármacos , Telómero/metabolismo , Adulto , Recuento de Células Sanguíneas , Disqueratosis Congénita/tratamiento farmacológico , Disqueratosis Congénita/genética , Femenino , Estudios de Seguimiento , Humanos , Masculino , Persona de Mediana Edad , Mutación , Síndromes Mielodisplásicos/inducido químicamente , Síndromes Mielodisplásicos/genética , Síndromes Mielodisplásicos/metabolismo , ARN/genética , ARN/metabolismo , Telomerasa/genética , Telomerasa/metabolismo , Telómero/genética
10.
Haematologica ; 106(5): 1303-1310, 2021 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-32241839

RESUMEN

Diamond Blackfan anemia (DBA) is predominantly an autosomal dominant inherited red cell aplasia primarily caused by pathogenic germline variants in ribosomal protein genes. DBA due to pathogenic RPL35A variants has been associated with large 3q29 deletions and phenotypes not common in DBA. We conducted a multi-institutional genotype-phenotype study of 45 patients with DBA associated with pathogenic RPL35A germline variants and curated the variant data on 21 additional cases from the literature. Genotype-phenotype analyses were conducted comparing patients with large deletions versus all other pathogenic variants in RPL35A. Twenty-two of the 45 cases had large deletions in RPL35A. After adjusting for multiple tests, a statistically significant association was observed between patients with a large deletion and steroid-resistant anemia, neutropenia, craniofacial abnormalities, chronic gastrointestinal problems, and intellectual disabilities (p<0.01) compared with all other pathogenic variants. Non-large deletion pathogenic variants were spread across RPL35A with no apparent hot spot and 56% of the individual family variants were observed more than once. In this, the largest known study of DBA patients with pathogenic RPL35A variants, we determined that patients with large deletions have a more severe phenotype that is clinically different from those with non-large deletion variants. Genes of interest also deleted in the 3q29 region that could be associated with some of these phenotypic features include LMLN and IQCG. Management of DBA due to large RPL35A deletions may be challenging due to complex problems and require comprehensive assessments by multiple specialists including immunologic, gastrointestinal, and developmental evaluations to provide optimal multidisciplinary care.


Asunto(s)
Anemia de Diamond-Blackfan , Anemia de Diamond-Blackfan/genética , Estudios de Asociación Genética , Humanos , Mutación , Fenotipo , Proteínas Ribosómicas/genética
11.
Am J Hematol ; 96(11): 1491-1504, 2021 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-34342889

RESUMEN

The North American Pediatric Aplastic Anemia Consortium (NAPAAC) is a group of pediatric hematologist-oncologists, hematopathologists, and bone marrow transplant physicians from 46 institutions in North America with interest and expertise in aplastic anemia, inherited bone marrow failure syndromes, and myelodysplastic syndromes. The NAPAAC Bone Marrow Failure Diagnosis and Care Guidelines Working Group was established with the charge of harmonizing the approach to the diagnostic workup of aplastic anemia in an effort to standardize best practices in the field. This document outlines the rationale for initial evaluations in pediatric patients presenting with signs and symptoms concerning for severe aplastic anemia.


Asunto(s)
Anemia Aplásica/diagnóstico , Anemia Aplásica/patología , Médula Ósea/patología , Niño , Diagnóstico Diferencial , Hemoglobina Fetal/análisis , Antígenos HLA/análisis , Humanos , América del Norte , Índice de Severidad de la Enfermedad
12.
Am J Hum Genet ; 100(3): 506-522, 2017 Mar 02.
Artículo en Inglés | MEDLINE | ID: mdl-28257692

RESUMEN

Ribosomal protein (RP) gene mutations, mostly associated with inherited or acquired bone marrow failure, are believed to drive disease by slowing the rate of protein synthesis. Here de novo missense mutations in the RPS23 gene, which codes for uS12, are reported in two unrelated individuals with microcephaly, hearing loss, and overlapping dysmorphic features. One individual additionally presents with intellectual disability and autism spectrum disorder. The amino acid substitutions lie in two highly conserved loop regions of uS12 with known roles in maintaining the accuracy of mRNA codon translation. Primary cells revealed one substitution severely impaired OGFOD1-dependent hydroxylation of a neighboring proline residue resulting in 40S ribosomal subunits that were blocked from polysome formation. The other disrupted a predicted pi-pi stacking interaction between two phenylalanine residues leading to a destabilized uS12 that was poorly tolerated in 40S subunit biogenesis. Despite no evidence of a reduction in the rate of mRNA translation, these uS12 variants impaired the accuracy of mRNA translation and rendered cells highly sensitive to oxidative stress. These discoveries describe a ribosomopathy linked to uS12 and reveal mechanistic distinctions between RP gene mutations driving hematopoietic disease and those resulting in developmental disorders.


Asunto(s)
Proteínas Ribosómicas/genética , Ribosomas/genética , Trastorno del Espectro Autista/genética , Proteínas Portadoras/genética , Células Cultivadas , Niño , Preescolar , Codón/genética , Discapacidades del Desarrollo/genética , Exoma , Femenino , Fibroblastos/citología , Fibroblastos/metabolismo , Variación Genética , Pérdida Auditiva/genética , Humanos , Discapacidad Intelectual/genética , Masculino , Microcefalia/genética , Mutación , Mutación Missense , Proteínas Nucleares/genética , Estrés Oxidativo , Biosíntesis de Proteínas/genética , Alineación de Secuencia , Análisis de Secuencia de ADN
14.
Blood ; 131(20): 2183-2192, 2018 05 17.
Artículo en Inglés | MEDLINE | ID: mdl-29549173

RESUMEN

An international, multicenter registry was established to collect retrospective and prospective clinical data on patients with pyruvate kinase (PK) deficiency, the most common glycolytic defect causing congenital nonspherocytic hemolytic anemia. Medical history and laboratory and radiologic data were retrospectively collected at enrollment for 254 patients with molecularly confirmed PK deficiency. Perinatal complications were common, including anemia that required transfusions, hyperbilirubinemia, hydrops, and prematurity. Nearly all newborns were treated with phototherapy (93%), and many were treated with exchange transfusions (46%). Children age 5 years and younger were often transfused until splenectomy. Splenectomy (150 [59%] of 254 patients) was associated with a median increase in hemoglobin of 1.6 g/dL and a decreased transfusion burden in 90% of patients. Predictors of a response to splenectomy included higher presplenectomy hemoglobin (P = .007), lower indirect bilirubin (P = .005), and missense PKLR mutations (P = .0017). Postsplenectomy thrombosis was reported in 11% of patients. The most frequent complications included iron overload (48%) and gallstones (45%), but other complications such as aplastic crises, osteopenia/bone fragility, extramedullary hematopoiesis, postsplenectomy sepsis, pulmonary hypertension, and leg ulcers were not uncommon. Overall, 87 (34%) of 254 patients had both a splenectomy and cholecystectomy. In those who had a splenectomy without simultaneous cholecystectomy, 48% later required a cholecystectomy. Although the risk of complications increases with severity of anemia and a genotype-phenotype relationship was observed, complications were common in all patients with PK deficiency. Diagnostic testing for PK deficiency should be considered in patients with apparent congenital hemolytic anemia and close monitoring for iron overload, gallstones, and other complications is needed regardless of baseline hemoglobin. This trial was registered at www.clinicaltrials.gov as #NCT02053480.


Asunto(s)
Anemia Hemolítica Congénita no Esferocítica/diagnóstico , Estudios de Asociación Genética , Piruvato Quinasa/deficiencia , Errores Innatos del Metabolismo del Piruvato/diagnóstico , Adolescente , Adulto , Anemia Hemolítica Congénita no Esferocítica/etiología , Anemia Hemolítica Congénita no Esferocítica/metabolismo , Anemia Hemolítica Congénita no Esferocítica/terapia , Transfusión Sanguínea , Niño , Preescolar , Colecistectomía/efectos adversos , Colecistectomía/métodos , Terapia Combinada , Activación Enzimática , Femenino , Genotipo , Humanos , Lactante , Recién Nacido , Masculino , Persona de Mediana Edad , Mutación , Fenotipo , Piruvato Quinasa/metabolismo , Errores Innatos del Metabolismo del Piruvato/etiología , Errores Innatos del Metabolismo del Piruvato/metabolismo , Errores Innatos del Metabolismo del Piruvato/terapia , Esplenectomía/efectos adversos , Esplenectomía/métodos , Evaluación de Síntomas , Resultado del Tratamiento , Adulto Joven
15.
Am J Hematol ; 95(5): 472-482, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-32043619

RESUMEN

Pyruvate kinase (PK) deficiency is a rare recessive congenital hemolytic anemia caused by mutations in the PKLR gene. This study reports the molecular features of 257 patients enrolled in the PKD Natural History Study. Of the 127 different pathogenic variants detected, 84 were missense and 43 non-missense, including 20 stop-gain, 11 affecting splicing, five large deletions, four in-frame indels, and three promoter variants. Within the 177 unrelated patients, 35 were homozygous and 142 compound heterozygous (77 for two missense, 48 for one missense and one non-missense, and 17 for two non-missense variants); the two most frequent mutations were p.R510Q in 23% and p.R486W in 9% of mutated alleles. Fifty-five (21%) patients were found to have at least one previously unreported variant with 45 newly described mutations. Patients with two non-missense mutations had lower hemoglobin levels, higher numbers of lifetime transfusions, and higher rates of complications including iron overload, extramedullary hematopoiesis, and pulmonary hypertension. Rare severe complications, including lower extremity ulcerations and hepatic failure, were seen more frequently in patients with non-missense mutations or with missense mutations characterized by severe protein instability. The PKLR genotype did not correlate with the frequency of complications in utero or in the newborn period. With ICCs ranging from 0.4 to 0.61, about the same degree of clinical similarity exists within siblings as it does between siblings, in terms of hemoglobin, total bilirubin, splenectomy status, and cholecystectomy status. Pregnancy outcomes were similar across genotypes in PK deficient women. This report confirms the wide genetic heterogeneity of PK deficiency.


Asunto(s)
Anemia Hemolítica Congénita no Esferocítica/genética , Estudios de Asociación Genética/métodos , Piruvato Quinasa/deficiencia , Errores Innatos del Metabolismo del Piruvato/genética , Adolescente , Adulto , Niño , Preescolar , Femenino , Humanos , Lactante , Recién Nacido , Masculino , Persona de Mediana Edad , Piruvato Quinasa/genética , Adulto Joven
16.
J Pediatr Hematol Oncol ; 42(4): e235-e237, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-30933022

RESUMEN

Diamond-Blackfan Anemia (DBA) is a rare inherited form of pure red cell aplasia that usually manifests in infancy or early childhood, and is characterized by normochromic macrocytic anemia and bone marrow erythroblastopenia. The majority of DBA cases are associated with mutations in ribosomal protein genes. Here, we describe a Lebanese girl with RPL5-mutated DBA unresponsive to steroid treatment who died from complications following late hematopoietic stem cell transplantation performed at the age of 15 years.


Asunto(s)
Anemia de Diamond-Blackfan/genética , Secuencia de Bases , Resistencia a Medicamentos/genética , Mutación del Sistema de Lectura , Proteínas Ribosómicas/genética , Eliminación de Secuencia , Adolescente , Aloinjertos , Anemia de Diamond-Blackfan/terapia , Niño , Preescolar , Resultado Fatal , Femenino , Trasplante de Células Madre Hematopoyéticas , Humanos , Recién Nacido , Líbano , Esteroides
19.
Hum Mutat ; 39(8): 1102-1111, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-29766597

RESUMEN

Diamond-Blackfan anemia (DBA) is a rare genetic hypoplasia of erythroid progenitors characterized by mild to severe anemia and associated with congenital malformations. Clinical manifestations in DBA patients are quite variable and genetic testing has become a critical factor in establishing a diagnosis of DBA. The majority of DBA cases are due to heterozygous loss-of-function mutations in ribosomal protein (RP) genes. Causative mutations are fairly straightforward to identify in the case of large deletions and frameshift and nonsense mutations found early in a protein coding sequence, but diagnosis becomes more challenging in the case of missense mutations and small in-frame indels. Our group recently characterized the phenotype of lymphoblastoid cell lines established from DBA patients with pathogenic lesions in RPS19 and observed that defective pre-rRNA processing, a hallmark of the disease, was rescued by lentiviral vectors expressing wild-type RPS19. Here, we use this complementation assay to determine whether RPS19 variants of unknown significance are capable of rescuing pre-rRNA processing defects in these lymphoblastoid cells as a means of assessing the effects of these sequence changes on the function of the RPS19 protein. This approach will be useful in differentiating pathogenic mutations from benign polymorphisms in identifying causative genes in DBA patients.


Asunto(s)
Anemia de Diamond-Blackfan/genética , Proteínas Ribosómicas/genética , Línea Celular , Codón sin Sentido/genética , Biología Computacional , ADN Complementario/genética , Mutación del Sistema de Lectura/genética , Humanos , Mutación/genética , Fenotipo
20.
Blood ; 127(11): 1387-97; quiz 1518, 2016 Mar 17.
Artículo en Inglés | MEDLINE | ID: mdl-26702063

RESUMEN

Germline GATA2 mutations cause cellular deficiencies with high propensity for myeloid disease. We investigated 426 children and adolescents with primary myelodysplastic syndrome (MDS) and 82 cases with secondary MDS enrolled in 2 consecutive prospective studies of the European Working Group of MDS in Childhood (EWOG-MDS) conducted in Germany over a period of 15 years. Germline GATA2 mutations accounted for 15% of advanced and 7% of all primary MDS cases, but were absent in children with MDS secondary to therapy or acquired aplastic anemia. Mutation carriers were older at diagnosis and more likely to present with monosomy 7 and advanced disease compared with wild-type cases. For stratified analysis according to karyotype, 108 additional primary MDS patients registered with EWOG-MDS were studied. Overall, we identified 57 MDS patients with germline GATA2 mutations. GATA2 mutations were highly prevalent among patients with monosomy 7 (37%, all ages) reaching its peak in adolescence (72% of adolescents with monosomy 7). Unexpectedly, monocytosis was more frequent in GATA2-mutated patients. However, when adjusted for the selection bias from monosomy 7, mutational status had no effect on the hematologic phenotype. Finally, overall survival and outcome of hematopoietic stem cell transplantation (HSCT) were not influenced by mutational status. This study identifies GATA2 mutations as the most common germline defect predisposing to pediatric MDS with a very high prevalence in adolescents with monosomy 7. GATA2 mutations do not confer poor prognosis in childhood MDS. However, the high risk for progression to advanced disease must guide decision-making toward timely HSCT.


Asunto(s)
Factor de Transcripción GATA2/deficiencia , Síndromes Mielodisplásicos/genética , Adolescente , Edad de Inicio , Niño , Preescolar , Aberraciones Cromosómicas , Cromosomas Humanos Par 1/genética , Cromosomas Humanos Par 7/genética , Cromosomas Humanos Par 8/genética , Ensayos Clínicos Fase III como Asunto , Análisis Mutacional de ADN , Sordera/genética , Femenino , Factor de Transcripción GATA2/genética , Predisposición Genética a la Enfermedad , Mutación de Línea Germinal , Humanos , Síndromes de Inmunodeficiencia/genética , Estimación de Kaplan-Meier , Masculino , Síndromes Mielodisplásicos/epidemiología , Síndromes Mielodisplásicos/etiología , Síndromes Mielodisplásicos/patología , Fenotipo , Prevalencia , Pronóstico , Estudios Prospectivos , Sesgo de Selección , Adulto Joven
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA