Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 575
Filtrer
1.
Cells ; 13(11)2024 May 27.
Article de Anglais | MEDLINE | ID: mdl-38891052

RÉSUMÉ

Diamond-Blackfan anemia (DBA) is a rare genetic disorder affecting the bone marrow's ability to produce red blood cells, leading to severe anemia and various physical abnormalities. Approximately 75% of DBA cases involve heterozygous mutations in ribosomal protein (RP) genes, classifying it as a ribosomopathy, with RPS19 being the most frequently mutated gene. Non-RP mutations, such as in GATA1, have also been identified. Current treatments include glucocorticosteroids, blood transfusions, and hematopoietic stem cell transplantation (HSCT), with HSCT being the only curative option, albeit with challenges like donor availability and immunological complications. Gene therapy, particularly using lentiviral vectors and CRISPR/Cas9 technology, emerges as a promising alternative. This review explores the potential of gene therapy, focusing on lentiviral vectors and CRISPR/Cas9 technology in combination with non-integrating lentiviral vectors, as a curative solution for DBA. It highlights the transformative advancements in the treatment landscape of DBA, offering hope for individuals affected by this condition.


Sujet(s)
Anémie de Blackfan-Diamond , Thérapie génétique , Anémie de Blackfan-Diamond/génétique , Anémie de Blackfan-Diamond/thérapie , Thérapie génétique/méthodes , Humains , Systèmes CRISPR-Cas/génétique , Vecteurs génétiques , Lentivirus/génétique , Animaux , Protéines ribosomiques/génétique , Mutation/génétique , Édition de gène/méthodes
2.
Leukemia ; 38(6): 1256-1265, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38740980

RÉSUMÉ

Recent advances in in-depth data-independent acquisition proteomic analysis have enabled comprehensive quantitative analysis of >10,000 proteins. Herein, an integrated proteogenomic analysis for inherited bone marrow failure syndrome (IBMFS) was performed to reveal their biological features and to develop a proteomic-based diagnostic assay in the discovery cohort; dyskeratosis congenita (n = 12), Fanconi anemia (n = 11), Diamond-Blackfan anemia (DBA, n = 9), Shwachman-Diamond syndrome (SDS, n = 6), ADH5/ALDH2 deficiency (n = 4), and other IBMFS (n = 18). Unsupervised proteomic clustering identified eight independent clusters (C1-C8), with the ribosomal pathway specifically downregulated in C1 and C2, enriched for DBA and SDS, respectively. Six patients with SDS had significantly decreased SBDS protein expression, with two of these not diagnosed by DNA sequencing alone. Four patients with ADH5/ALDH2 deficiency showed significantly reduced ADH5 protein expression. To perform a large-scale rapid IBMFS screening, targeted proteomic analysis was performed on 417 samples from patients with IBMFS-related hematological disorders (n = 390) and healthy controls (n = 27). SBDS and ADH5 protein expressions were significantly reduced in SDS and ADH5/ALDH2 deficiency, respectively. The clinical application of this first integrated proteogenomic analysis would be useful for the diagnosis and screening of IBMFS, where appropriate clinical screening tests are lacking.


Sujet(s)
Maladies de la moelle osseuse , Aplasies médullaires , Protéogénomique , Humains , Aplasies médullaires/génétique , Aplasies médullaires/anatomopathologie , Protéogénomique/méthodes , Mâle , Femelle , Maladies de la moelle osseuse/génétique , Maladies de la moelle osseuse/anatomopathologie , Enfant , Adulte , Adolescent , Enfant d'âge préscolaire , Anémie de Blackfan-Diamond/génétique , Anémie de Blackfan-Diamond/diagnostic , Jeune adulte , Anémie de Fanconi/génétique , Anémie de Fanconi/diagnostic , Protéomique/méthodes , Nourrisson , Maladie de Shwachman/génétique , Dyskératose congénitale/génétique , Dyskératose congénitale/diagnostic , Dyskératose congénitale/anatomopathologie
3.
Exp Hematol ; 135: 104235, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38740323

RÉSUMÉ

The emergence of multiomic single-cell technologies over the last decade has led to improved insights into both normal hematopoiesis and its perturbation in a variety of hematological disorders. Diamond-Blackfan anemia (DBA) syndrome is one such disorder where single-cell assays have helped to delineate the cellular and molecular defects underlying the disease. DBA is caused by heterozygous loss-of-function germline variants in genes encoding ribosomal proteins (RPs). Despite the widespread role of ribosomes in hematopoiesis, the most frequent and severe cytopenia in DBA is anemia. In this review we discussed how single-cell studies, including clonogenic cell culture assays, fluorescence-activated cell sorting (FACS) and single-cell RNA sequencing (scRNA-seq), have led to insights into the pathogenesis of DBA. The main therapies are regular blood transfusions, glucocorticoids, or hematopoietic stem cell transplantation (HSCT) but all are associated with significant morbidity and mortality. We will therefore outline how single-cell studies can inform new therapies for DBA. Furthermore, we discussed how DBA serves as a useful model for understanding normal erythropoiesis in terms of its cellular hierarchy, molecular regulation during homeostasis, and response to "stress."


Sujet(s)
Anémie de Blackfan-Diamond , Analyse sur cellule unique , Anémie de Blackfan-Diamond/thérapie , Anémie de Blackfan-Diamond/génétique , Humains , Érythropoïèse/génétique , Transplantation de cellules souches hématopoïétiques , Protéines ribosomiques/génétique
4.
Lancet Haematol ; 11(5): e368-e382, 2024 May.
Article de Anglais | MEDLINE | ID: mdl-38697731

RÉSUMÉ

Diamond-Blackfan anaemia (DBA), first described over 80 years ago, is a congenital disorder of erythropoiesis with a predilection for birth defects and cancer. Despite scientific advances, this chronic, debilitating, and life-limiting disorder continues to cause a substantial physical, psychological, and financial toll on patients and their families. The highly complex medical needs of affected patients require specialised expertise and multidisciplinary care. However, gaps remain in effectively bridging scientific discoveries to clinical practice and disseminating the latest knowledge and best practices to providers. Following the publication of the first international consensus in 2008, advances in our understanding of the genetics, natural history, and clinical management of DBA have strongly supported the need for new consensus recommendations. In 2014 in Freiburg, Germany, a panel of 53 experts including clinicians, diagnosticians, and researchers from 27 countries convened. With support from patient advocates, the panel met repeatedly over subsequent years, engaging in ongoing discussions. These meetings led to the development of new consensus recommendations in 2024, replacing the previous guidelines. To account for the diverse phenotypes including presentation without anaemia, the panel agreed to adopt the term DBA syndrome. We propose new simplified diagnostic criteria, describe the genetics of DBA syndrome and its phenocopies, and introduce major changes in therapeutic standards. These changes include lowering the prednisone maintenance dose to maximum 0·3 mg/kg per day, raising the pre-transfusion haemoglobin to 9-10 g/dL independent of age, recommending early aggressive chelation, broadening indications for haematopoietic stem-cell transplantation, and recommending systematic clinical surveillance including early colorectal cancer screening. In summary, the current practice guidelines standardise the diagnostics, treatment, and long-term surveillance of patients with DBA syndrome of all ages worldwide.


Sujet(s)
Anémie de Blackfan-Diamond , Consensus , Humains , Anémie de Blackfan-Diamond/diagnostic , Anémie de Blackfan-Diamond/thérapie , Anémie de Blackfan-Diamond/génétique , Prise en charge de la maladie , Transplantation de cellules souches hématopoïétiques
5.
JCI Insight ; 9(10)2024 May 22.
Article de Anglais | MEDLINE | ID: mdl-38775150

RÉSUMÉ

This study lays the groundwork for future lentivirus-mediated gene therapy in patients with Diamond Blackfan anemia (DBA) caused by mutations in ribosomal protein S19 (RPS19), showing evidence of a new safe and effective therapy. The data show that, unlike patients with Fanconi anemia (FA), the hematopoietic stem cell (HSC) reservoir of patients with DBA was not significantly reduced, suggesting that collection of these cells should not constitute a remarkable restriction for DBA gene therapy. Subsequently, 2 clinically applicable lentiviral vectors were developed. In the former lentiviral vector, PGK.CoRPS19 LV, a codon-optimized version of RPS19 was driven by the phosphoglycerate kinase promoter (PGK) already used in different gene therapy trials, including FA gene therapy. In the latter one, EF1α.CoRPS19 LV, RPS19 expression was driven by the elongation factor alpha short promoter, EF1α(s). Preclinical experiments showed that transduction of DBA patient CD34+ cells with the PGK.CoRPS19 LV restored erythroid differentiation, and demonstrated the long-term repopulating properties of corrected DBA CD34+ cells, providing evidence of improved erythroid maturation. Concomitantly, long-term restoration of ribosomal biogenesis was verified using a potentially novel method applicable to patients' blood cells, based on ribosomal RNA methylation analyses. Finally, in vivo safety studies and proviral insertion site analyses showed that lentivirus-mediated gene therapy was nontoxic.


Sujet(s)
Anémie de Blackfan-Diamond , Thérapie génétique , Vecteurs génétiques , Cellules souches hématopoïétiques , Lentivirus , Protéines ribosomiques , Anémie de Blackfan-Diamond/thérapie , Anémie de Blackfan-Diamond/génétique , Humains , Thérapie génétique/méthodes , Lentivirus/génétique , Protéines ribosomiques/génétique , Vecteurs génétiques/génétique , Cellules souches hématopoïétiques/métabolisme , Animaux , Souris , Mâle , Femelle , Ribosomes/métabolisme , Ribosomes/génétique , Régions promotrices (génétique) , Mutation , Transplantation de cellules souches hématopoïétiques/méthodes
6.
Br J Haematol ; 204(5): 2077-2085, 2024 May.
Article de Anglais | MEDLINE | ID: mdl-38462764

RÉSUMÉ

Diamond-Blackfan anaemia (DBA) is a rare, inherited bone marrow failure syndrome with a ribosomal defect causing slowed globin chain production with normal haem synthesis, causing an overabundance of reactive iron/haem and erythroid-specific cellular toxicity. Eltrombopag, a non-peptide thrombopoietin receptor agonist, is a potent intracellular iron chelator and induced a robust durable response in an RPS19-mutated DBA patient on another trial. We hypothesized eltrombopag would improve RBC production in DBA patients. We conducted a single-centre, single-arm pilot study (NCT04269889) assessing safety and erythroid response of 6 months of daily, fixed-dose eltrombopag for DBA patients. Fifteen transfusion-dependent (every 3-5 weeks) patients (median age 18 [range 2-56]) were treated. One responder had sustained haemoglobin improvement and >50% reduction in RBC transfusion frequency. Of note, 7/15 (41%) patients required dose reductions or sustained discontinuation of eltrombopag due to asymptomatic thrombocytosis. Despite the low response rate, eltrombopag has now improved erythropoiesis in several patients with DBA with a favourable safety profile. Dosing restrictions due to thrombocytosis may cause insufficient iron chelation to decrease haem production and improve anaemia in most patients. Future work will focus on erythropoiesis dynamics in patients and use of haem synthesis inhibitors without an impact on other haematopoietic lineages.


Sujet(s)
Anémie de Blackfan-Diamond , Benzoates , Hydrazines , Pyrazoles , Humains , Anémie de Blackfan-Diamond/traitement médicamenteux , Pyrazoles/usage thérapeutique , Hydrazines/usage thérapeutique , Hydrazines/administration et posologie , Hydrazines/effets indésirables , Benzoates/usage thérapeutique , Benzoates/administration et posologie , Benzoates/effets indésirables , Adulte , Mâle , Femelle , Enfant , Adolescent , Adulte d'âge moyen , Jeune adulte , Enfant d'âge préscolaire , Projets pilotes , Résultat thérapeutique , Récepteurs à la thrombopoïétine/agonistes , Récidive , Érythropoïèse/effets des médicaments et des substances chimiques
7.
Tidsskr Nor Laegeforen ; 144(4)2024 03 19.
Article de Norvégien | MEDLINE | ID: mdl-38506013

RÉSUMÉ

Background: Anemia in children is common and finding the underlying cause is often uncomplicated. However, in some cases, the underlying diagnosis is rare and difficult to diagnose. Case presentation: A toddler presented with severe anemia with normal red cell indices and a low reticulocyte count. The remaining hematological parameters were normal, bar a slight thrombocytosis. At this point a diagnosis of transient erythroblastopenia of childhood (TEC) was made. The child continued to have slight anemia with intermittent macrocytosis and reticulocytopenia throughout childhood. Growth and development was normal, and there were no signs of congenital abnormalities in the heart or kidneys nor any craniofacial or phalangeal defects. Repeated bone marrow examinations showed no significant abnormal findings. As a teenager the patient was diagnosed with Diamond-Blackfan anemia through an exome-based gene panel which revealed a mutation in the RPL11 gene. Interpretation: Congenital bone marrow failure syndromes do not always present in the classical way, leading to a delayed diagnosis. The increasing availability of different gene panels for patients with persistent abnormal hematological laboratory parameters offers the possibility of a more accurate diagnostic pathway, which is important for adequate follow-up and genetic counselling.


Sujet(s)
Anémie de Blackfan-Diamond , Anémie hémolytique congénitale , Anémie , Adolescent , Humains , Anémie/diagnostic , Anémie/étiologie , Anémie de Blackfan-Diamond/diagnostic , Anémie de Blackfan-Diamond/génétique , Mutation
8.
Br J Haematol ; 204(5): 1598-1599, 2024 May.
Article de Anglais | MEDLINE | ID: mdl-38485153

RÉSUMÉ

The rich history surrounding Diamond-Blackfan anaemia (DBA), originally described in 1938 as congenital hypoplastic anaemia2 reflects the evolution of paediatric haematology. In their paper, the authors1 present the results of a clinical trial using the thrombopoietin-mimetic agent eltrombopag to treat red cell failure in DBA. A low response rate belies the importance of this work. Commentary on: Duncan et al. Treatment of refractory/relapsed Diamond-Blackfan anaemia with eltrombopag. Br J Haematol 2024;204:2077-2085.


Sujet(s)
Anémie de Blackfan-Diamond , Benzoates , Hydrazines , Pyrazoles , Anémie de Blackfan-Diamond/traitement médicamenteux , Humains , Pyrazoles/usage thérapeutique , Benzoates/usage thérapeutique , Hydrazines/usage thérapeutique
9.
Blood Cells Mol Dis ; 106: 102838, 2024 May.
Article de Anglais | MEDLINE | ID: mdl-38413287

RÉSUMÉ

Diamond-Blackfan anemia (DBA) was the first ribosomopathy described in humans. DBA is a congenital hypoplastic anemia, characterized by macrocytic aregenerative anemia, manifesting by differentiation blockage between the BFU-e/CFU-e developmental erythroid progenitor stages. In 50 % of the DBA cases, various malformations are noted. Strikingly, for a hematological disease with a relative erythroid tropism, DBA is due to ribosomal haploinsufficiency in 24 different ribosomal protein (RP) genes. A few other genes have been described in DBA-like disorders, but they do not fit into the classical DBA phenotype (Sankaran et al., 2012; van Dooijeweert et al., 2022; Toki et al., 2018; Kim et al., 2017 [1-4]). Haploinsufficiency in a RP gene leads to defective ribosomal RNA (rRNA) maturation, which is a hallmark of DBA. However, the mechanistic understandings of the erythroid tropism defect in DBA are still to be fully defined. Erythroid defect in DBA has been recently been linked in a non-exclusive manner to a number of mechanisms that include: 1) a defect in translation, in particular for the GATA1 erythroid gene; 2) a deficit of HSP70, the GATA1 chaperone, and 3) free heme toxicity. In addition, p53 activation in response to ribosomal stress is involved in DBA pathophysiology. The DBA phenotype may thus result from the combined contributions of various actors, which may explain the heterogenous phenotypes observed in DBA patients, even within the same family.


Sujet(s)
Anémie de Blackfan-Diamond , Anémie dysérythropoïétique congénitale , Anémie macrocytaire , Humains , Anémie de Blackfan-Diamond/génétique , Protéines ribosomiques/génétique , Protéines ribosomiques/métabolisme , Précurseurs érythroïdes/métabolisme , Mutation
10.
J Pediatr Hematol Oncol ; 46(2): e195-e198, 2024 Mar 01.
Article de Anglais | MEDLINE | ID: mdl-38277626

RÉSUMÉ

Diamond-Blackfan anemia (DBA) is a rare, inherited bone marrow failure syndrome that is both genetically and clinically heterogeneous. The diagnosis of DBA has changed over time, with advancements in our understanding of the varied genetic etiologies and phenotypic manifestations of the disease. We present a rare case of a patient who never developed erythroid precursor hypoplasia, adding to the understanding of atypical manifestations of DBA. Our patient had spontaneous remission followed by subsequent relapse, both atypical and poorly understood processes in DBA. We highlight important considerations in diagnostically challenging cases and review major outstanding questions surrounding DBA.


Sujet(s)
Anémie de Blackfan-Diamond , Humains , Anémie de Blackfan-Diamond/complications , Anémie de Blackfan-Diamond/génétique , Anémie de Blackfan-Diamond/diagnostic , Aplasies médullaires , Protéines ribosomiques/génétique
12.
Leukemia ; 38(1): 1-9, 2024 01.
Article de Anglais | MEDLINE | ID: mdl-37973818

RÉSUMÉ

ABSTACT: Diamond-Blackfan anemia (DBA) is a rare congenital bone marrow failure disorder characterized by erythroid hypoplasia. It primarily affects infants and is often caused by heterozygous allelic variations in ribosomal protein (RP) genes. Recent studies also indicated that non-RP genes like GATA1, TSR2, are associated with DBA. P53 activation, translational dysfunction, inflammation, imbalanced globin/heme synthesis, and autophagy dysregulation were shown to contribute to disrupted erythropoiesis and impaired red blood cell production. The main therapeutic option for DBA patients is corticosteroids. However, half of these patients become non-responsive to corticosteroid therapy over prolonged treatment and have to be given blood transfusions. Hematopoietic stem cell transplantation is currently the sole curative option, however, the treatment is limited by the availability of suitable donors and the potential for serious immunological complications. Recent advances in gene therapy using lentiviral vectors have shown promise in treating RPS19-deficient DBA by promoting normal hematopoiesis. With deepening insights into the molecular framework of DBA, emerging therapies like gene therapy hold promise for providing curative solutions and advancing comprehension of the underlying disease mechanisms.


Sujet(s)
Anémie de Blackfan-Diamond , Transplantation de cellules souches hématopoïétiques , Nourrisson , Humains , Anémie de Blackfan-Diamond/génétique , Anémie de Blackfan-Diamond/thérapie , Protéines ribosomiques/génétique , Protéines ribosomiques/métabolisme , Thérapie génétique , Érythropoïèse/génétique , Aplasies médullaires
13.
Am J Med Genet A ; 194(3): e63454, 2024 Mar.
Article de Anglais | MEDLINE | ID: mdl-37897121

RÉSUMÉ

A 26-year-old female proband with a clinical diagnosis and consistent phenotype of Diamond-Blackfan anemia (DBA, OMIM 105650) without an identified genotype was referred to the Undiagnosed Diseases Network. DBA is classically associated with monoallelic variants that have an autosomal-dominant or -recessive mode of inheritance. Intriguingly, her case was solved by a detection of a digenic interaction between non-allelic RPS19 and RPL27 variants. This was confirmed with a machine learning structural model, co-segregation analysis, and RNA sequencing. This is the first report of DBA caused by a digenic effect of two non-allelic variants demonstrated by machine learning structural model. This case suggests that atypical phenotypic presentations of DBA may be caused by digenic inheritance in some individuals. We also conclude that a machine learning structural model can be useful in detecting digenic models of possible interactions between products encoded by alleles of different genes inherited from non-affected carrier parents that can result in DBA with an unrealized 25% recurrence risk.


Sujet(s)
Anémie de Blackfan-Diamond , Humains , Femelle , Adulte , Anémie de Blackfan-Diamond/diagnostic , Anémie de Blackfan-Diamond/génétique , Protéines ribosomiques/génétique , Génotype , Allèles , Phénotype , Séquence nucléotidique , Mutation
15.
Pediatr Blood Cancer ; 71(3): e30834, 2024 Mar.
Article de Anglais | MEDLINE | ID: mdl-38149846

RÉSUMÉ

Diamond-Blackfan anemia (DBA) is a congenital anemia with erythroid cell aplasia. Most of the causative genes are ribosomal proteins. GATA1, a hematopoietic master transcription factor required for erythropoiesis, also causes DBA. GATA1 is located on Xp11.23; therefore, DBA develops only in males in an X-linked inheritance pattern. Here, we report a case of transient erythroblastopenia and moderate anemia in a female newborn infant with a de novo GATA1 variant. In this patient, increased methylation of the GATA1 wild-type allele was observed in erythroid cells. Skewed lyonization of GATA1 may cause mild transient erythroblastopenia in a female patient.


Sujet(s)
Anémie aplasique , Anémie de Blackfan-Diamond , Anémie hémolytique congénitale , Mâle , Nourrisson , Nouveau-né , Humains , Femelle , Protéines ribosomiques/génétique , Anémie de Blackfan-Diamond/génétique , Érythropoïèse , Facteur de transcription GATA-1/génétique
16.
Medicina (Kaunas) ; 59(11)2023 Nov 05.
Article de Anglais | MEDLINE | ID: mdl-38004002

RÉSUMÉ

Diamond-Blackfan anemia (DBA) is a congenital bone marrow failure syndrome associated with malformations. DBA is related to defective ribosome biogenesis, which impairs erythropoiesis, causing hyporegenerative macrocytic anemia. The disease has an autosomal dominant inheritance and is commonly diagnosed in the first year of life, requiring continuous treatment. We present the case of a young woman who, at the age of 21, developed severe symptomatic anemia. Although, due to malformations, a congenital syndrome had been suspected since birth, a confirmation diagnosis was not made until the patient was referred to our center for an evaluation of her anemia. In her neonatal medical history, she presented with anemia that required red blood cell transfusions, but afterwards remained with a stable, mild, asymptomatic anemia throughout her childhood and adolescence. Her family history was otherwise unremarkable. To explain the symptomatic anemia, vitamin deficiencies, autoimmune diseases, bleeding causes, and myeloid and lymphoid neoplasms were investigated and ruled out. A molecular investigation showed the RPL5 gene variant c.392dup, p.(Asn131Lysfs*6), confirming the diagnosis of DBA. All family members have normal blood values and none harbored the mutation. Here, we will discuss the unusual evolution of this case and revisit the literature.


Sujet(s)
Anémie de Blackfan-Diamond , Mutation avec décalage du cadre de lecture , Humains , Jeune adulte , Nouveau-né , Femelle , Adolescent , Enfant , Mutation avec décalage du cadre de lecture/génétique , Protéines ribosomiques/génétique , Mutation , Anémie de Blackfan-Diamond/complications , Anémie de Blackfan-Diamond/diagnostic , Anémie de Blackfan-Diamond/génétique , Phénotype
17.
Int J Mol Sci ; 24(19)2023 Oct 06.
Article de Anglais | MEDLINE | ID: mdl-37834388

RÉSUMÉ

Mice with a constitutive increase in p53 activity exhibited features of dyskeratosis congenita (DC), a bone marrow failure syndrome (BMFS) caused by defective telomere maintenance. Further studies confirmed, in humans and mice, that germline mutations affecting TP53 or its regulator MDM4 may cause short telomeres and alter hematopoiesis, but also revealed features of Diamond-Blackfan anemia (DBA) or Fanconi anemia (FA), two BMFSs, respectively, caused by defects in ribosomal function or DNA repair. p53 downregulates several genes mutated in DC, either by binding to promoter sequences (DKC1) or indirectly via the DREAM repressor complex (RTEL1, DCLRE1B), and the p53-DREAM pathway represses 22 additional telomere-related genes. Interestingly, mutations in any DC-causal gene will cause telomere dysfunction and subsequent p53 activation to further promote the repression of p53-DREAM targets. Similarly, ribosomal dysfunction and DNA lesions cause p53 activation, and p53-DREAM targets include the DBA-causal gene TSR2, at least 9 FA-causal genes, and 38 other genes affecting ribosomes or the FA pathway. Furthermore, patients with BMFSs may exhibit brain abnormalities, and p53-DREAM represses 16 genes mutated in microcephaly or cerebellar hypoplasia. In sum, positive feedback loops and the repertoire of p53-DREAM targets likely contribute to partial phenotypic overlaps between BMFSs of distinct molecular origins.


Sujet(s)
Anémie de Blackfan-Diamond , Dyskératose congénitale , Anémie de Fanconi , Humains , Animaux , Souris , Protéine p53 suppresseur de tumeur/génétique , Aplasies médullaires , Anémie de Fanconi/génétique , Anémie de Blackfan-Diamond/génétique , Dyskératose congénitale/génétique , Télomère/génétique , Protéines nucléaires/génétique , Protéines du cycle cellulaire/génétique , Protéines proto-oncogènes/génétique , Exodeoxyribonucleases/génétique
19.
Int J Lab Hematol ; 45(5): 766-773, 2023 Oct.
Article de Anglais | MEDLINE | ID: mdl-37376976

RÉSUMÉ

INTRODUCTION: Diamond-Blackfan anemia (DBA) is a rare congenital bone marrow failure syndrome characterized by erythroid aplasia, physical malformation, and cancer predisposition. Twenty ribosomal protein genes and three non-ribosomal protein genes have been identified associated with DBA. METHODS: To investigate the presence of novel mutations and gain a deeper understanding of the molecular mechanisms of disease, targeted next-generation sequencing was performed in 12 patients with clinically suspected DBA. Literatures were retrieved with complete clinical information published in English by November 2022. The clinical features, treatment, and RPS10/RPS26 mutations were analyzed. RESULTS: Among the 12 patients, 11 mutations were identified and 5 of them were novel (RPS19, p.W52S; RPS10, p.P106Qfs*11; RPS26, p.R28*; RPL5, p.R35*; RPL11, p.T44Lfs*40). Including 2 patients in this study, 13 patients with RPS10 mutations and 38 patients with RPS26 mutations were reported from 4 and 6 countries, respectively. The incidences of physical malformation in patients with RPS10 and RPS26 mutations (22% and 36%, respectively) were lower than the overall incidence in DBA patients (~50%). Patients with RPS26 mutations had a worse response rate of steroid therapy than RPS10 (47% vs. 87.5%), but preferred RBC transfusions (67% vs. 44%, p = 0.0253). CONCLUSION: Our findings add to the DBA pathogenic variant database and demonstrate the clinical presentations of the DBA patients with RPS10/RPS26 mutations. It shows that next-generation sequencing is a powerful tool for the diagnosis of genetic diseases such as DBA.


Sujet(s)
Anémie de Blackfan-Diamond , Humains , Anémie de Blackfan-Diamond/génétique , Mutation , Génotype
20.
Elife ; 122023 Jun 05.
Article de Anglais | MEDLINE | ID: mdl-37272618

RÉSUMÉ

Ribosomal protein (Rp) gene haploinsufficiency can result in Diamond-Blackfan Anemia (DBA), characterized by defective erythropoiesis and skeletal defects. Some mouse Rp mutations recapitulate DBA phenotypes, although others lack erythropoietic or skeletal defects. We generated a conditional knockout mouse to partially delete Rps12. Homozygous Rps12 deletion resulted in embryonic lethality. Mice inheriting the Rps12KO/+ genotype had growth and morphological defects, pancytopenia, and impaired erythropoiesis. A striking reduction in hematopoietic stem cells (HSCs) and progenitors in the bone marrow (BM) was associated with decreased ability to repopulate the blood system after competitive and non-competitive BM transplantation. Rps12KO/+ lost HSC quiescence, experienced ERK and MTOR activation, and increased global translation in HSC and progenitors. Post-natal heterozygous deletion of Rps12 in hematopoietic cells using Tal1-Cre-ERT also resulted in pancytopenia with decreased HSC numbers. However, post-natal Cre-ERT induction led to reduced translation in HSCs and progenitors, suggesting that this is the most direct consequence of Rps12 haploinsufficiency in hematopoietic cells. Thus, RpS12 has a strong requirement in HSC function, in addition to erythropoiesis.


Sujet(s)
Anémie de Blackfan-Diamond , Pancytopénie , Animaux , Souris , Anémie de Blackfan-Diamond/génétique , Anémie de Blackfan-Diamond/métabolisme , Érythropoïèse/génétique , Gènes essentiels , Haploinsuffisance , Cellules souches hématopoïétiques/métabolisme , Souris knockout , Pancytopénie/génétique , Pancytopénie/métabolisme , Protéines ribosomiques/génétique , Protéines ribosomiques/métabolisme
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...