Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
Add more filters










Database
Language
Publication year range
1.
Gene ; 545(2): 282-9, 2014 Jul 25.
Article in English | MEDLINE | ID: mdl-24835311

ABSTRACT

Defects in genes encoding ribosomal proteins cause Diamond Blackfan Anemia (DBA), a red cell aplasia often associated with physical abnormalities. Other bone marrow failure syndromes have been attributed to defects in ribosomal components but the link between erythropoiesis and the ribosome remains to be fully defined. Several lines of evidence suggest that defects in ribosome synthesis lead to "ribosomal stress" with p53 activation and either cell cycle arrest or induction of apoptosis. Pathways independent of p53 have also been proposed to play a role in DBA pathogenesis. We took an unbiased approach to identify p53-independent pathways activated by defects in ribosome synthesis by analyzing global gene expression in various cellular models of DBA. Ranking-Principal Component Analysis (Ranking-PCA) was applied to the identified datasets to determine whether there are common sets of genes whose expression is altered in these different cellular models. We observed consistent changes in the expression of genes involved in cellular amino acid metabolic process, negative regulation of cell proliferation and cell redox homeostasis. These data indicate that cells respond to defects in ribosome synthesis by changing the level of expression of a limited subset of genes involved in critical cellular processes. Moreover, our data support a role for p53-independent pathways in the pathophysiology of DBA.


Subject(s)
Anemia, Diamond-Blackfan/genetics , Anemia, Diamond-Blackfan/metabolism , Phenotype , Ribosomal Proteins/genetics , Ribosomal Proteins/metabolism , Transcription, Genetic , Alternative Splicing , Cell Line , DNA Mutational Analysis , Gene Expression Regulation , Gene Order , Humans , Molecular Sequence Annotation , Mutation , Reproducibility of Results , Ribosomal Proteins/deficiency , Transcriptome , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism
2.
BMC Genomics ; 10: 442, 2009 Sep 18.
Article in English | MEDLINE | ID: mdl-19765279

ABSTRACT

BACKGROUND: Diamond-Blackfan anaemia (DBA) is a rare inherited red cell hypoplasia characterised by a defect in the maturation of erythroid progenitors and in some cases associated with malformations. Patients have an increased risk of solid tumors. Mutations have been found in several ribosomal protein (RP) genes, i.e RPS19, RPS24, RPS17, RPL5, RPL11, RPL35A. Studies in haematopoietic progenitors from patients show that haplo-insufficiency of an RP impairs rRNA processing and ribosome biogenesis. DBA lymphocytes show reduced protein synthesis and fibroblasts display abnormal rRNA processing and impaired proliferation. RESULTS: To evaluate the involvement of non-haematopoietic tissues in DBA, we have analysed global gene expression in fibroblasts from DBA patients compared to healthy controls. Microarray expression profiling using Affymetrix GeneChip Human Genome U133A 2.0 Arrays revealed that 421 genes are differentially expressed in DBA patient fibroblasts. These genes include a large cluster of ribosomal proteins and factors involved in protein synthesis and amino acid metabolism, as well as genes associated to cell death, cancer and tissue development. CONCLUSION: This analysis reports for the first time an abnormal gene expression profile in a non-haematopoietic cell type in DBA. These data support the hypothesis that DBA may be due to a defect in general or specific protein synthesis.


Subject(s)
Anemia, Diamond-Blackfan/genetics , Fibroblasts/metabolism , Gene Expression Profiling , Ribosomal Proteins/genetics , Base Sequence , Case-Control Studies , DNA Mutational Analysis , Female , Humans , Male , Molecular Sequence Data , Mutation , Oligonucleotide Array Sequence Analysis , Protein Biosynthesis , Reverse Transcriptase Polymerase Chain Reaction
3.
Pediatr Blood Cancer ; 53(3): 411-6, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19489057

ABSTRACT

BACKGROUND: The determination of telomere length is useful for the characterization of dyskeratosis congenita (DC) and of aplastic anemias (AA) as well as hematological malignancies. Short telomeres result from a specific defect of telomere maintenance in DC and likely from higher cellular turnover in AA and hematological malignancies. Data are not conclusive for Diamond-Blackfan anemia (DBA), a pure erythroid aplasia due to defects of ribosomal proteins. Our aim was to evaluate the utility of a qPCR method for telomere length assessment to evaluate the diagnostic contribution of telomere measurement in bone marrow failure syndromes (BMFS). PROCEDURE: Telomere length was evaluated by qPCR in peripheral blood cells from 95 normal individuals and 62 patients with BMFS, including 45 patients with DBA. RESULTS: Results obtained with qPCR are comparable with other quantitative methods, such as flow-FISH and Southern blotting. Our data show that only one DBA patient and a minority of other BMFS patients have very short telomeres, defined as less than the 1st percentile of controls. CONCLUSIONS: The qPCR method for telomere length evaluation is an easy alternative to other methods and may thus be valuable in a clinical hematological laboratory setting. Telomere maintenance does not seem to be involved in the pathogenesis of DBA unlike in other BMFSs.


Subject(s)
Anemia, Aplastic/genetics , Anemia, Diamond-Blackfan/genetics , Dyskeratosis Congenita/genetics , Fanconi Anemia/genetics , Telomere , Adolescent , Adult , Aged , Aged, 80 and over , Anemia, Aplastic/blood , Anemia, Diamond-Blackfan/blood , Cell Cycle Proteins/genetics , Child , Child, Preschool , Fanconi Anemia/blood , Humans , Infant , Middle Aged , Nuclear Proteins/genetics , Polymerase Chain Reaction
4.
Hum Mutat ; 29(11): E263-70, 2008 Nov.
Article in English | MEDLINE | ID: mdl-18781615

ABSTRACT

Mutations in ribosomal proteins RPS19, RPS24 and RPS17 have been reported in Diamond-Blackfan Anemia (DBA), an autosomal dominant disease characterised by pure red cell aplasia. DBA is the prototype of ribosomapathies: a protein synthesis defect in a tissue with a high cellular turnover is considered the cause of the erythroid progenitor failure. We have created the Diamond-Blackfan Anemia mutation database to curate and record DBA gene mutations, together with their functional consequences and clinical phenotypes. This locus-specific resource is open to future submissions and is available online (http://www.dbagenes.unito.it). It is founded on the Leiden Open (source) Variation Database (LOVD) system and includes data from sequence and structure analysis tools, genomic database resources and published reports. It lists all identified variants and background genomic information. Phenotypic data are accessed by selecting a particular mutation. The database includes 219 unique variants of which 86 are disease-causing mutations. The database will be supplemented with other DBA genes as soon as they are reported and their mutations are identified and it should be of assistance to clinicians and investigators involved in DBA research and care.


Subject(s)
Anemia, Diamond-Blackfan/genetics , Databases, Nucleic Acid , Ribosomal Proteins/genetics , Anemia, Diamond-Blackfan/complications , DNA Mutational Analysis , Genome, Human , Humans , Mutation , User-Computer Interface
5.
Hum Mutat ; 29(7): 911-20, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18412286

ABSTRACT

Diamond-Blackfan anemia (DBA) is an inherited disease characterized by pure erythroid aplasia. Thirty percent (30%) of patients display malformations, especially of the hands, face, heart, and urogenital tract. DBA has an autosomal dominant pattern of inheritance. De novo mutations are common and familial cases display wide clinical heterogeneity. Twenty-five percent (25%) of patients carry a mutation in the ribosomal protein (RP) S19 gene, whereas mutations in RPS24, RPS17, RPL35A, RPL11, and RPL5 are rare. These genes encode for structural proteins of the ribosome. A link between ribosomal functions and erythroid aplasia is apparent in DBA, but its etiology is not clear. Most authors agree that a defect in protein synthesis in a rapidly proliferating tissue, such as the erythroid bone marrow, may explain the defective erythropoiesis. A total of 77 RPS19 mutations have been described. Most are whole gene deletions, translocations, or truncating mutations (nonsense or frameshift), suggesting that haploinsufficiency is the basis of DBA pathology. A total of 22 missense mutations have also been described and several works have provided in vitro functional data for the mutant proteins. This review looks at the data on all these mutations, proposes a functional classification, and describes six new mutations. It is shown that patients with RPS19 mutations display a poorer response to steroids and a worse long-term prognosis compared to other DBA patients.


Subject(s)
Anemia, Diamond-Blackfan/genetics , Mutation , Ribosomal Proteins/genetics , Humans , Phenotype , Polymorphism, Genetic
SELECTION OF CITATIONS
SEARCH DETAIL
...