17 research outputs found

    Ribosomal protein gene RPL9 variants can differentially impair ribosome function and cellular metabolism

    Get PDF
    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

    Role of HSP70 protein in Blacfan-Diamond anemia

    No full text
    L’anĂ©mie de Blackfan-Diamond (ABD) est une Ă©rythroblastopĂ©nie congĂ©nitale rare, secondaire Ă  un blocage de la maturation Ă©rythroĂŻde entre les stades BFU-e et CFU-e. L’ABD est le plus souvent la consĂ©quence d’une mutation germinale affectant un gĂšne codant pour une protĂ©ine ribosomique (RP) de la petite ou de la grande sous-unitĂ© du ribosome. Quatorze gĂšnes distincts ont Ă©tĂ© identifiĂ©s. Les gĂšnes les plus frĂ©quemment mutĂ©s sont les gĂšnes RPL5, RPL11 et RPS19 (37% des patients). Plus rarement, l’ABD est la consĂ©quence de mutations dans le gĂšne TSR2 ou dans le gĂšne GATA-1. Ce dernier code pour un facteur de transcription majeur de l’érythropoĂŻĂšse. Chez les patients ABD, les mutations de GATA-1 induisent une perte quasi-totale de la forme longue de GATA-1 qui est nĂ©cessaire Ă  la diffĂ©renciation de la cellule Ă©rythroĂŻde. Notre groupe a identifiĂ© deux phĂ©notypes de l’ABD in vitro en fonction du gĂšne mutĂ©. En cas d’haploinsuffisance RPS19, la prolifĂ©ration Ă©rythroĂŻde est moins rĂ©duite qu’en cas d’haploinsuffisance RPL5 ou de RPL11. Une haploinsuffisance RPS19 n’altĂšre pas la diffĂ©renciation Ă©rythroĂŻde et n’induit pas d’apoptose contrairement Ă  l’haploinsuffisance RPL5 ou RPL11 oĂč il existe un retard de diffĂ©renciation Ă©rythroĂŻde et un excĂšs net d’apoptose responsable au moins en partie de la diminution drastique de la prolifĂ©ration Ă©rythroĂŻde dans ces phĂ©notypes.HSP70 est impliquĂ©e dans la survie cellulaire et la diffĂ©renciation Ă©rythroĂŻde en protĂ©geant GATA-1 du clivage par la caspase-3, une protĂ©ase activĂ©e lors de la diffĂ©renciation Ă©rythroĂŻde terminale. Comme la diffĂ©rence entre les deux phĂ©notypes d’ABD in vitro concernait la diffĂ©renciation Ă©rythroĂŻde et la survie cellulaire, nous avons Ă©mis l’hypothĂšse selon laquelle la mutation de certains gĂšnes RP provoque un dĂ©faut d’expression d’HSP70 conduisant au blocage de la diffĂ©renciation Ă©rythroĂŻde et Ă  l’excĂšs d’apoptose retrouvĂ©s dans les phĂ©notypes sĂ©vĂšres d’ABD.Nous avons Ă©tudiĂ© diffĂ©rents patients atteints d’ABD, porteurs de mutations dans les gĂšnes RPS19, RPL5 ou RPL11 et gĂ©nĂ©rĂ© un modĂšle in vitro d’ABD en exprimant, dans des cellules CD34+ humaines issues de sang de cordon, des ARN interfĂ©rents ciblant RPL5, RPL11 ou RPS19. Chez les patients comme dans le modĂšle reproduisant l’ABD, l’haploinsuffisance RPL5 ou RPL11 diminue drastiquement l’expression protĂ©ique de HSP70 et de GATA-1 (Western blot, microscopie confocale et en cytomĂ©trie couplĂ©e Ă  des techniques d’imagerie, (technologie ImageStream) Ă  la diffĂ©rence de 1’haploinsuffisance RPS19. Dans tous les cas, HSP70 est normalement transcrite et traduite. Les inhibiteurs du protĂ©asome (MG132, lactacystine, bortezomib) restaurent l’expression10de HSP70. La diminution d’expression de HSP70 est donc liĂ©e Ă  une dĂ©gradation protĂ©asomale. L’invalidation de RPL11 induit une polyubiquitinylation importante de HSP70. La transduction lentivirale de l’ADN complĂ©mentaire d’HSP70 dans les cellules primitives invalidĂ©es pour RPL11 permet de restaurer l’expression de HSP70 et de GATA-1 Ă  un niveau similaire aux contrĂŽles et de rĂ©tablir la prolifĂ©ration cellulaire et la diffĂ©renciation Ă©rythroĂŻde, confirmant le rĂŽle clĂ© de HSP70 dans le phĂ©notype sĂ©vĂšre RPL5+/Mut ou RPL11+/Mut. Les formes les plus sĂ©vĂšres de l’ABD sont associĂ©es Ă  la dĂ©gradation de HSP70 par le protĂ©asome. La perte de la protĂ©ine chaperone de GATA-1 induit la perte de GATA-1, facteur de transcription majeur de la diffĂ©renciation Ă©rythroĂŻde. Une augmentation de l’expression de HSP70 pourrait ainsi constituer une nouvelle approche thĂ©rapeutique dans l’ABD.Diamond-Blackfan anemia (DBA) is the first ribosomopathy identified and is characterized by a moderate to severe, usually macrocytic aregenerative anemia associated with congenital malformations in 50% of the DBA cases. This congenital rare erythroblastopenia is due to a blockade in erythroid differentiation between the BFU-e and CFU-e stages. The link between a haploinsufficiency in a ribosomal protein (RP) gene that now encompass 15 different RP genes and the erythroid defect is still to be fully defined. Recently, mutations in TSR2 and GATA-1 genes have been identified in a few DBA families. The GATA-1 gene encodes for the major transcription factor critical for erythropoiesis and mutation in this gene that lead to loss of expression of the long form of the protein, necessary for the erythroid differentiation accounts for erythroblastopenia of DBA phenotype. Our group and others (Dutt et al., Blood 2011) have shown previously that p53 plays an important role in the DBA erythroblastopenia, inducing cell cycle arrest in G0/G1 and depending on the nature of RP gene mutation, a delayed erythroid differentiation and an increased apoptosis. Indeed, we identified two distinct DBA phenotypes (H. Moniz, M. Gastou, Cell Death Dis, 2012): a haploinsufficiency in RPL5 or RPL11 reduced dramatically the erythroid proliferation, delayed the erythroid differentiation, and markedly increased apoptosis, while RPS19 haploinsufficiency while reduced the extent of erythroid proliferation without inducing significant apoptosis. While p53 pathway has been found to be activated in RP haploinsufficient erythroid cells in DBA patients or shRNA-RPS19, -RPL5, or -RPL11 infected CD34+ erythroid cells, the intensity of the p53 activation pathway (p21, BAX, NOXA) is different depending on the mutated RP gene. Since the differences between the two phenotypes involved the eytrhoid differentiation and the degree of apoptosis we hypothesized that HSP70, a chaperone protein of GATA-1 may play a key role in the erythroid defect of DBA. Indeed, HSP70 protects GATA-1 from the cleavage by the caspase 3, a protease activated during erythroid differentiation. As such reduced levels of HSP70 related to a RP haploinsufficiency could account for increased apoptosis and delayed erythroid differentiation of erythroid cells in DBA. Indeed, a defect in RPL5 or RPL11 decreased dramatically the expression level of HSP70 and GATA-1 in primary human erythroid cells from DBA patients and following in vitro knockdown of the proteins in CD34+ cells by RPL5 or RPL11 shRNA. Importantly, RPS19 haploinsufficiency did not exhibit this effect in conjunction with normal levels of HSP70 expression. Furthermore, we found that the decreased expression level of12HSP70 was independent on the p53 activation. Strikingly, HSP70 was noted to be degraded by the proteasome since the bortezomib, the MG132, or the lactacystin were able to restore both the HSP70 expression level and intracellular localization in the cell. The lentiviral infection of depleted RPL11 cord blood CD34+ cells with a wild type HSP70 cDNA restored both the erythroid proliferation and differentiation, and reduced apoptosis, confirming a critical role for HSP70 in the erythroid defect in the RPL11+/Mut DBA phenotypes. The loss of HSP70 may explain the loss of GATA-1 in DBA and also the erythroid tropism of the DBA disease. Restoration of the HSP70 expression level may be a viable and novel therapeutic option for management of this debilitating and difficult to manage erythroid disorder

    XPO1 regulates erythroid differentiation and is a new target for the treatment of ÎČ-thalassemia.

    Full text link
    ÎČ-thalassemia major (ÎČ-TM) is an inherited hemoglobinopathy caused by a quantitative defect in the synthesis of ÎČ-globin chains of hemoglobin, leading to the accumulation of free a-globin chains that aggregate and cause ineffective erythropoiesis. We have previously demonstrated that terminal erythroid maturation requires a transient activation of caspase-3 and that the chaperone Heat Shock Protein 70 (HSP70) accumulates in the nucleus to protect GATA-1 transcription factor from caspase-3 cleavage. This nuclear accumulation of HSP70 is inhibited in human ÎČ-TM erythroblasts due to HSP70 sequestration in the cytoplasm by free a-globin chains, resulting in maturation arrest and apoptosis. Likewise, terminal maturation can be restored by transduction of a nuclear-targeted HSP70 mutant. Here we demonstrate that in normal erythroid progenitors, HSP70 localization is regulated by the exportin-1 (XPO1), and that treatment of ÎČ-thalassemic erythroblasts with an XPO1 inhibitor increased the amount of nuclear HSP70, rescued GATA-1 expression and improved terminal differentiation, thus representing a new therapeutic option to ameliorate ineffective erythropoiesis of ÎČ-TM

    Recurring mutations in RPL15 are linked to hydrops fetalis and treatment independence in diamond-blackfan anemia

    Get PDF
    Diamond-Blackfan anemia (DBA) is a rare inherited bone marrow failure disorder linked predominantly to ribosomal protein gene mutations. Here the European DBA consortium reports novel mutations identified in the RPL15 gene in 6 unrelated individuals diagnosed with DBA. Although point mutations have not been previously reported for RPL15, we identified 4 individuals with truncating mutations p.Tyr81* (in 3 of 4) and p.Gln29*, and 2 with missense variants p.Leu10Pro and p.Lys153Thr. Notably, 75% (3 of 4) of truncating mutation carriers manifested with severe hydrops fetalis and required intrauterine transfusions. Even more remarkable is the observation that the 3 carriers of p.Tyr81* mutation became treatment-independent between four and 16 months of life and maintained normal blood counts until their last follow up. Genetic reversion at the DNA level as a potential mechanism of remission was not observed in our patients. In vitro studies revealed that cells carrying RPL15 mutations have pre-rRNA processing defects, reduced 60S ribosomal subunit formation, and severe proliferation defects. Red cell culture assays of RPL15-mutated primary erythroblast cells also showed a severe reduction in cell proliferation, delayed erythroid differentiation, elevated TP53 activity, and increased apoptosis. This study identifies a novel subgroup of DBA with mutations in the RPL15 gene with an unexpected high rate of hydrops fetalis and spontaneous, long-lasting remission

    S6 kinase phosphorylation and autophagy is induced by reactive oxygen species (ROS).

    No full text
    <p>(<b>A</b>) Confocal microscopy analysis of GFP-LC3 HEK cells transfected with siScr or si<i>RPS19</i> and either untreated or treated with 10 mM Trolox overnight. Size bars = 10 ”M. (<b>B</b>) Quantification of the average number of GFP-LC3 puncta per cell in (<b>A</b>). (<b>C</b>) Western blot analysis of phosphorylated S6 kinase expression in GFP-LC3 HEK transfected with siScr or si<i>RPS19</i> and either untreated or treated with 10 mM Trolox overnight. (<b>D</b>) Western blot analysis of phosphorylated S6 kinase and phosphorylated AKT substrate expression in 2 dpf zebrafish embryos untreated or treated with 10 mM Trolox overnight. (<b>E</b>) Survival rates of embryos treated overnight with 10 mM Trolox. The results are the compilation of three independent experiments with N = 25 embryos representing each mutation. The statistics shown are compared to wild type. (<b>F</b>) Analysis of p53 stabilization in 2 dpf wild type, rpS7-deficient embryos, <i>p53<sup>M214K/M214K</sup></i> mutant embryos, or double mutants (<i>rpS7;p53</i>) by western blotting. (<b>G</b>) Analysis of p53 stabilization by western blotting of zebrafish mutants treated overnight with 100 ”M Trolox.</p

    Insulin abolishes autophagy in cells with RPS19 loss.

    No full text
    <p>(<b>A</b>) Confocal microscopy analysis of GFP-LC3 HEK cells transfected with siScr or si<i>RPS19</i> and either untreated or stimulated with 350 nM insulin for 6 hours. Size bars = 10 ”M. (<b>B</b>) Quantification of the average number of GFP-LC3 puncta per cell in (<b>A</b>). (<b>C</b>) Western blot analysis of LC3 expression in cells from (<b>A</b>). Densitometer analysis used to calculate the ratio of LC3II/actin.</p
    corecore