Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
Biochem J ; 478(5): 997-1008, 2021 03 12.
Artigo em Inglês | MEDLINE | ID: mdl-33661277

RESUMO

Proteins belonging to the universal ribosomal protein (rp) uS19 family are constituents of small ribosomal subunits, and their conserved globular parts are involved in the formation of the head of these subunits. The eukaryotic rp uS19 (previously known as S15) comprises a C-terminal extension that has no homology in the bacterial counterparts. This extension is directly implicated in the formation of the ribosomal decoding site and thereby affects translational fidelity in a manner that has no analogy in bacterial ribosomes. Another eukaryote-specific feature of rp uS19 is its essential participance in the 40S subunit maturation due to the interactions with the subunit assembly factors required for the nuclear exit of pre-40S particles. Beyond properties related to the translation machinery, eukaryotic rp uS19 has an extra-ribosomal function concerned with its direct involvement in the regulation of the activity of an important tumor suppressor p53 in the Mdm2/Mdmx-p53 pathway. Mutations in the RPS15 gene encoding rp uS19 are linked to diseases (Diamond Blackfan anemia, chronic lymphocytic leukemia and Parkinson's disease) caused either by defects in the ribosome biogenesis or disturbances in the functioning of ribosomes containing mutant rp uS19, likely due to the changed translational fidelity. Here, we review currently available data on the involvement of rp uS19 in the operation of the translational machinery and in the maturation of 40S subunits, on its extra-ribosomal function, and on relationships between mutations in the RPS15 gene and certain human diseases.


Assuntos
Anemia de Diamond-Blackfan/patologia , Leucemia Linfocítica Crônica de Células B/patologia , Doença de Parkinson/patologia , Biossíntese de Proteínas , Proteínas Ribossômicas/metabolismo , Ribossomos/metabolismo , Anemia de Diamond-Blackfan/etiologia , Animais , Humanos , Leucemia Linfocítica Crônica de Células B/etiologia , Doença de Parkinson/etiologia , Proteínas Ribossômicas/genética , Ribossomos/genética
2.
Int J Hematol ; 112(6): 894-899, 2020 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-32772263

RESUMO

Diamond-Blackfan Anemia (DBA) is a congenital pure red cell aplasia caused by heterozygous variants in ribosomal protein genes. The hematological features associated with DBA are highly variable and non-hematological abnormalities are common. We report herein on an affected mother and her daughter presenting with transfusion-dependent anemia. The mother showed mild physical abnormalities and entered spontaneous remission at age 13 years. Her daughter was born with occipital meningocele. Exome sequencing of DNA from the mother revealed a heterozygous novel splice site variant (NM_001011.4:c.508-3T > G) in the Ribosomal Protein S7 gene (RPS7) inherited by the daughter. Functional analysis of the RPS7 variant expressed from a mini-gene construct revealed that the exon 7 acceptor splice site was replaced by a cryptic splice resulting in a transcript missing 64 bp of exon 7 (p.Val170Serfs*8). Our study confirms a pathogenic effect of a novel RPS7 variant in DBA associated with spontaneous remission in the mother and meningocele in her daughter, thus adding to the genotype-phenotype correlations in DBA.


Assuntos
Anemia de Diamond-Blackfan/genética , Aberrações Cromossômicas , Estudos de Associação Genética , Variação Genética/genética , Meningocele/genética , Splicing de RNA/genética , Proteínas Ribossômicas/genética , Adolescente , Adulto , Anemia de Diamond-Blackfan/etiologia , Criança , Éxons/genética , Feminino , Humanos , Meningocele/etiologia , Relações Mãe-Filho , Remissão Espontânea , Análise de Sequência de DNA
3.
Blood Cells Mol Dis ; 62: 1-5, 2016 11.
Artigo em Inglês | MEDLINE | ID: mdl-27732904

RESUMO

Diamond-Blackfan anemia (DBA) is a congenital erythroid aplasia that usually presents as macrocytic anemia during infancy. Ribosomal protein S19 (RPS19) is identified as the first gene associated with DBA. RPS19 is mutated in 25% of DBA patients, but its role in DBA pathogenesis remains to be elucidated. We have identified a novel heterozygous frameshift mutation in RPS19 gene in a DBA child presenting with profound anemia after birth. A single nucleotide heterozygous deletion (C.251delG) results in frameshift in RPS19 gene in exon 4 at codon 84 with possible premature stop codon (p.Arg84LysfsX21). The mutant allele was not detected in her parents, indicating de novo mutation. Both alleles were expressed at the same level. Using an immunofluorescence technique, the mutated-type RPS19 expressions were mostly localized to entire nuclei with little staining for nucleoli and its intracellular localization significantly differed from the wild-type RPS19, which was localized to both nuclei and nucleoli. This type of a mutation could be very helpful in further understanding the role of the RPS19 protein in DBA pathogenesis.


Assuntos
Anemia de Diamond-Blackfan/genética , Nucléolo Celular/química , Mutação da Fase de Leitura , Proteínas Ribossômicas/genética , Deleção de Sequência , Anemia de Diamond-Blackfan/etiologia , Povo Asiático , Células Sanguíneas/patologia , Exame de Medula Óssea , Feminino , Heterozigoto , Humanos , Lactente , RNA Mensageiro/análise , Proteínas Ribossômicas/análise
4.
Leukemia ; 29(11): 2221-9, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-25987256

RESUMO

Diamond-Blackfan anemia (DBA) is a congenital erythroid hypoplasia caused by haploinsufficiency of genes encoding ribosomal proteins (RPs). Perturbed ribosome biogenesis in DBA has been shown to induce a p53-mediated ribosomal stress response. However, the mechanisms of p53 activation and its relevance for the erythroid defect remain elusive. Previous studies have indicated that activation of p53 is caused by the inhibition of mouse double minute 2 (Mdm2), the main negative regulator of p53, by the 5S ribonucleoprotein particle (RNP). Meanwhile, it is not clear whether this mechanism solely mediates the p53-dependent component found in DBA. To approach this question, we crossed our mouse model for RPS19-deficient DBA with Mdm2(C305F) knock-in mice that have a disrupted 5S RNP-Mdm2 interaction. Upon induction of the Rps19 deficiency, Mdm2(C305F) reversed the p53 response and improved expansion of hematopoietic progenitors in vitro, and ameliorated the anemia in vivo. Unexpectedly, disruption of the 5S RNP-Mdm2 interaction also led to selective defect in erythropoiesis. Our findings highlight the sensitivity of erythroid progenitor cells to aberrations in p53 homeostasis mediated by the 5S RNP-Mdm2 interaction. Finally, we provide evidence indicating that physiological activation of the 5S RNP-Mdm2-p53 pathway may contribute to functional decline of the hematopoietic system in a cell-autonomous manner over time.


Assuntos
Anemia de Diamond-Blackfan/etiologia , Células Precursoras Eritroides/fisiologia , Proteínas Proto-Oncogênicas c-mdm2/fisiologia , Ribonucleoproteínas/fisiologia , Animais , Modelos Animais de Doenças , Doxiciclina/farmacologia , Eritropoese , Camundongos , Camundongos Endogâmicos C57BL , RNA Ribossômico 5S/fisiologia , Proteínas Ribossômicas/fisiologia , Transdução de Sinais , Proteína Supressora de Tumor p53/fisiologia
5.
Biochim Biophys Acta ; 1842(6): 765-8, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24412987

RESUMO

Diamond Blackfan anemia is a red cell hypoplasia that typically presents within the first year of life. Most cases of Diamond Blackfan anemia are caused by ribosome assembly defects linked to haploinsufficiency for structural proteins of either ribosomal subunit. Nucleolar stress associated with abortive ribosome assembly leads to p53 activation via the interaction of free ribosomal proteins with HDM2, a negative regulator of p53. Significant challenges remain in linking this nucleolar stress signaling pathway to the clinical features of Diamond Blackfan anemia. Defining aspects of disease presentation may relate to developmental and physiological triggers that work in conjunction with nucleolar stress signaling to heighten the p53 response in the developing erythron after birth. The growing number of ribosomopathies provides additional challenges for linking molecular mechanisms with clinical phenotypes. This article is part of a Special Issue entitled: Role of the Nucleolus in Human Disease.


Assuntos
Anemia de Diamond-Blackfan/genética , Eritrócitos/metabolismo , Proteínas Ribossômicas/genética , Ribossomos/genética , Anemia de Diamond-Blackfan/etiologia , Anemia de Diamond-Blackfan/fisiopatologia , Eritrócitos/patologia , Humanos , Mutação , Proteínas Proto-Oncogênicas c-mdm2/genética , Proteínas Ribossômicas/metabolismo , Ribossomos/metabolismo , Transdução de Sinais/genética , Ativação Transcricional , Proteína Supressora de Tumor p53/genética
6.
Georgian Med News ; (216): 46-51, 2013 Mar.
Artigo em Russo | MEDLINE | ID: mdl-23567308

RESUMO

This study was designed to investigate the children with congenital (Diamond-Blackfan Anaemia - DBA) and acquired pure red cell aplasia (PRCA). 4 children, aged 1 month to 3 years with PRCA were enrolled in a trial. Investigations include: detailed history and physical examination, complete blood count with red blood cell indices, reticulocyte count, bone marrow examination, iron metabolism, viral serologies, immunological and urine analysis, anti-erythrocyte antibodies, measurement of hemoglobin F and erythrocyte adenosinedezaminase activity, chest x-ray, liver and renal function tests. Based on clinical and para-clinical data analyses and catamnestic observations two cases were diagnosed with DBA and other two with acquired PRCA among which one was determined by EBV virus and another by transient erythroblastopenia. Nowadays 3 children with PRCA are asymptomatic. In case of PRCA (because of its rare occurrence) a differentiated approach is required to every specific occasion. A series of investigations should be conducted to determine the origin and choose the treatment principles accordingly.


Assuntos
Anemia de Diamond-Blackfan/etiologia , Anemia de Diamond-Blackfan/terapia , Aplasia Pura de Série Vermelha/etiologia , Aplasia Pura de Série Vermelha/terapia , Anemia de Diamond-Blackfan/diagnóstico , Contagem de Células Sanguíneas , Transfusão de Sangue , Exame de Medula Óssea , Pré-Escolar , Feminino , Humanos , Lactente , Recém-Nascido , Prednisolona/efeitos adversos , Prednisolona/uso terapêutico , Gravidez , Complicações Infecciosas na Gravidez , Aplasia Pura de Série Vermelha/diagnóstico
7.
Blood Cells Mol Dis ; 35(2): 189-92, 2005.
Artigo em Inglês | MEDLINE | ID: mdl-15996880

RESUMO

Diamond-Blackfan anemia (DBA) is a rare congenital pure red cell aplasia. Previous studies indicate that mutations of a gene on chromosome 19q13.2, which encodes a ribosomal protein, are responsible for 25% of cases. Recent investigations suggest both the presence of a second candidate region on chromosome 8p and non-19q, non-8p disease. In linkage analysis studies of 28 multiplex DBA families, we identified 8 families with disease linkage to chromosome 1q31. In 4 families, the disease linked exclusively to 1q31. Here, we report that the FLVCR gene on 1q31, which encodes a cytoplasmic heme exporter associated with red cell aplasia in cats, is not involved in DBA in these families.


Assuntos
Anemia de Diamond-Blackfan/genética , Ligação Genética , Proteínas de Membrana Transportadoras/genética , Receptores Virais/genética , Regiões 3' não Traduzidas , Anemia de Diamond-Blackfan/etiologia , Cromossomos Humanos Par 1 , Análise Mutacional de DNA , Saúde da Família , Heme/metabolismo , Humanos , Proteínas de Membrana Transportadoras/metabolismo , Proteínas de Membrana Transportadoras/fisiologia , Linhagem , Mutação Puntual , Receptores Virais/metabolismo , Receptores Virais/fisiologia
8.
Mol Ther ; 11(4): 627-37, 2005 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-15771965

RESUMO

Diamond-Blackfan anemia (DBA) is a congenital red cell aplasia in which 25% of the patients have a mutation in the ribosomal protein S19 (RPS19) gene. No models exist for RPS19-deficient DBA and the molecular pathogenesis is unknown. To establish an in vitro inducible model for DBA, human erythroid leukemic cell lines, TF-1 and UT-7 cells, were cotransduced with a lentiviral vector expressing the green fluorescent protein (GFP) gene and small interfering RNA (siRNA) against RPS19 controlled by a tet operator regulatory element and another transactivator vector containing the red fluorescent protein (RFP) gene and the cDNA encoding a tetracycline-controllable transcriptional repressor. Following transduction, the RFP-positive and GFP-negative cell population was sorted by flow cytometry. Upon incubation with doxycycline (0.5 mug/ml), more than 98% of cells expressed GFP and the siRNA. Significant suppression of erythroid differentiation, cell growth, and colony formation was observed in cells treated with siRNA against RPS19 but not in cells treated with a control vector. These findings show that RPS19 plays an important role in the regulation of hematopoietic cell proliferation and erythroid differentiation. These novel cell lines represent models for RPS19-deficient DBA and can be used to identify the molecular mechanisms in RPS19-deficient DBA.


Assuntos
Anemia de Diamond-Blackfan/etiologia , Linhagem Celular , RNA Interferente Pequeno/genética , Proteínas Ribossômicas/deficiência , Anemia de Diamond-Blackfan/genética , Anemia de Diamond-Blackfan/metabolismo , Sequência de Bases , Diferenciação Celular , Proliferação de Células , Regulação para Baixo , Doxiciclina/farmacologia , Eritrócitos/efeitos dos fármacos , Eritrócitos/metabolismo , Células Eritroides/metabolismo , Expressão Gênica/efeitos dos fármacos , Vetores Genéticos/genética , Hematopoese/genética , Hematopoese/fisiologia , Humanos , Lentivirus/genética , Modelos Biológicos , Dados de Sequência Molecular , Células Mieloides/metabolismo , Proteínas Ribossômicas/genética , Proteínas Ribossômicas/metabolismo , Células-Tronco
9.
Haematologica ; 89(4): 480-9, 2004 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-15075082

RESUMO

BACKGROUND AND OBJECTIVES: Diamond-Blackfan anemia (DBA) is a rare, pure red blood cell aplasia of childhood caused by an intrinsic defect in erythropoietic progenitors. Malformations occur in about 40% of patients. More than half of patients respond to steroids; non-responders need chronic transfusions or stem cell transplantation (SCT). Mutations in the gene encoding ribosomal protein S19 are found in 25% of patients, but the link with erythropoiesis is unclear. A second DBA locus has been found on chromosome 8p22-p23; analysis of genes of the region is in progress. METHODS AND INFORMATION SOURCES: We present clinical and molecular data from 97 Italian DBA patients and a review of the literature. RESULTS AND STATE OF THE ART: We describe five new RPS19 gene mutations: four point mutations and one unbalanced chromosomal translocation. Hematologic findings, malformations and outcome are similar in the RPS19 mutated and the non-mutated groups. No genotype-phenotype correlation has been found so far in RPS19 mutated patients. Our data, however, and a thorough review of literature show a worse outcome (expressed as transfusion dependence) in patients with mutations that completely abolish one allele, i.e. gross chromosomal rearrangements and mutations at the initiation codon. The association of mental retardation with large deletions at the 19q locus points to a contiguous gene syndrome. A recurrent missense mutation (Arg62Trp) is associated with transfusion dependence in eight of the nine reported cases. PERSPECTIVES: Nationwide collaboration and population-based registries recording molecular data are essential for the further dissection of this rare heterogeneous disease and the definition of new therapeutic trials.


Assuntos
Anemia de Diamond-Blackfan/genética , Mutação , Anemia de Diamond-Blackfan/epidemiologia , Anemia de Diamond-Blackfan/etiologia , Códon de Iniciação/genética , Humanos , Itália/epidemiologia , Epidemiologia Molecular , Fenótipo , Translocação Genética
10.
Blood Cells Mol Dis ; 31(1): 93-7, 2003.
Artigo em Inglês | MEDLINE | ID: mdl-12850491

RESUMO

Diamond Blackfan Anemia (DBA) is a congenital disorder characterized by decreased red blood cell production and developmental abnormalities. We herein show that DBA progenitors produced lower numbers of phenotypically normal erythroid colonies in vitro, whereas nonerythroid colonies were normally abundant and developed. To determine whether DBA stem cells are capable of producing early erythroid, monocyto-granulocytic, and lymphoid progenitors in vivo we used a mouse xenotransplantation model. We demonstrate that DBA stem cells poorly repopulated erythroid progeny in NOD/SCID mice, whereas the monocyto-granulocytic and lymphoid progenies were repopulated normally. Therefore, we conclude that disordered DBA erythropoiesis may be a result of defective erythroid-lineage commitment and maintenance of early erythroid progenitors.


Assuntos
Anemia de Diamond-Blackfan/patologia , Eritropoese , Transplante de Células-Tronco Hematopoéticas , Células-Tronco Hematopoéticas/patologia , Anemia de Diamond-Blackfan/etiologia , Animais , Antígenos CD34 , Estudos de Casos e Controles , Criança , Ensaio de Unidades Formadoras de Colônias , Sobrevivência de Enxerto , Humanos , Leucopoese , Camundongos , Camundongos Endogâmicos NOD , Modelos Animais , Transplante Heterólogo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA