scholarly journals Ribosomal and hematopoietic defects in induced pluripotent stem cells derived from Diamond Blackfan anemia patients

Blood ◽  
2013 ◽  
Vol 122 (6) ◽  
pp. 912-921 ◽  
Author(s):  
Loïc Garçon ◽  
Jingping Ge ◽  
Shwetha H. Manjunath ◽  
Jason A. Mills ◽  
Marisa Apicella ◽  
...  

Key PointsRibosome biogenesis and hematopoiesis are impaired in iPSCs from DBA patients. The abnormalities of DBA iPSCs are ameliorated by genetic restoration of the defective ribosomal protein genes.

Blood ◽  
2014 ◽  
Vol 124 (21) ◽  
pp. 254-254
Author(s):  
Jingping Ge ◽  
Marisa Apicella ◽  
Jason A. Mills ◽  
Loic Garcon ◽  
Deborah L. French ◽  
...  

Abstract DBA is an inherited bone marrow failure syndrome that usually develops in the first year of life with clinical features of red cell aplasia and sometimes variable developmental abnormalities. Most affected patients have heterozygous loss of function mutations of one of the 11 ribosomal proteins (RPs) or mutations in the GATA1 gene which encodes an erythroid specific transcription factor. We have previously demonstrated that induced pluripotent stem cells (iPSCs) from fibroblast of DBA patients with RPS19 or RPL5 mutations recapitulate the pathogenesis of DBA, with the mutant lines showing abnormal ribosome biogenesis and altered erythropoiesis. The mechanism whereby haploinsufficiency for RPs causes failure of erythropoiesis and the other DBA features is still unknown. We investigated the pathways that are affected in these DBA iPSCs using an Affymetrix human exon array, and we observed the striking dysregulation of the TGF β pathway in DBA lines. The TGF β downstream target genes, such as DKK1, BAMBI, FN1, COL3A1, COLA1A1 and PAI-1 significantly increased in the DBA iPSCs. The TGF β signaling is complex and can occur via a canonical pathway or by a number of non-canonical pathways. We measured levels of a number of intermediates in these pathways by western blot, and observed a significant increase in the levels of p-JNK, a mediator of a non-canonical pathway, in the DBA iPSCs. Moreover, when the mutant cells were corrected by ectopic expression of WT RPS19 or RPL5, levels of p-JNK returned to normal. We also investigated the SMAD family, which are mediators of the TGF β canonical pathway and are known to negatively regulate the regeneration of hematopoietic stem cells. We observed a drastic decrease in SMAD4, but no change in p-SMAD2. Again corrected lines showed normal expression levels of SMAD4. Our data suggests that the activation of a non-canonical TGF β pathway in the DBA iPSCs may lead increased expression of the downstream genes; and the decrease of anti-proliferative factor SMAD4 may explain how DBA iPSCs maintain their growth. We conclude that the mutations of RPS19 or RPL5 both affect ribosome biogenesis and TGF β signaling, which can cause the failure of erythropoiesis at the stem cell stage. We further suggest that the suppression of SMAD4 may be used as a therapeutic target for DBA treatment. Disclosures No relevant conflicts of interest to declare.


Blood ◽  
2013 ◽  
Vol 121 (24) ◽  
pp. 4925-4929 ◽  
Author(s):  
Shilpa Gandre-Babbe ◽  
Prasuna Paluru ◽  
Chiaka Aribeana ◽  
Stella T. Chou ◽  
Silvia Bresolin ◽  
...  

Key Points Patient-derived iPSCs recapitulate juvenile myelomonocytic leukemia. MEK inhibition normalizes GM-CSF independence and hypersensitivity in myeloid precursors from JMML iPSCs.


Blood ◽  
2014 ◽  
Vol 124 (12) ◽  
pp. 1926-1930 ◽  
Author(s):  
Jon P. Connelly ◽  
Erika M. Kwon ◽  
Yongxing Gao ◽  
Niraj S. Trivedi ◽  
Abdel G. Elkahloun ◽  
...  

Key Points Successful generation of iPSC lines from FPD patient fibroblasts. Restoration of megakaryopoiesis after RUNX1 mutation correction.


Blood ◽  
2013 ◽  
Vol 122 (21) ◽  
pp. 1224-1224
Author(s):  
Jingping Ge ◽  
Marisa Apicella ◽  
Shwetha Manjunath ◽  
Jason A. Mills ◽  
Loïc Garçon ◽  
...  

Abstract DBA is an inherited bone marrow failure syndrome that usually presents in the first year of life with red cell aplasia and variable developmental abnormalities. Most affected patients have heterozygous loss of function mutations in one of 11 genes encoding small or large subunit ribosomal proteins (RPs). It is not known how RP haploinsufficiency causes failed erythropoiesis. The penetrance of DBA varies widely, even between members of the same family. This is presumably due to modifying factors but the nature of these remains enigmatic. We have previously derived induced pluripotent stem cells (iPSCs) from fibroblasts of a DBA patient with a RPS19 mutation and the mutant line showed abnormal ribosome biogenesis and altered erythropoiesis, thus recapitulating the DBA phenotype. To investigate the mechanisms underlying variable penetrance in DBA, we generated induced pluripotent stem cells (iPSCs) from members of a family with DBA caused by a nonsense mutation in RPS19 (p.Q126X). We analyzed iPSCs from two DBA patients and an asymptomatic individual, all of whom carry the RPS19 mutation. iPSC lines from the DBA affected individuals showed faulty 40S (small) ribosomal subunit assembly, reduced processing of 18S rRNA, and impaired hematopoiesis, with about 2.5 fold decreased expansion of mutant erythroblasts compared to those derived from iPSCs from normal individuals or the asymptomatic carrier. In contrast, iPSCs from the asymptomatic carrier showed milder defects in ribosome biogenesis and hematopoiesis. Genetic correction of ribosomal protein deficiency via cDNA transfer into the “safe harbor” AAVS1 locus alleviated abnormalities in ribosome biogenesis and hematopoiesis in the DBA iPSCs. Using an Affymetrix human exon array to investigate the pathways that are affected in these iPSCs, we observed striking over-activation of the TGF beta pathway in iPSCs of the DBA affected individuals compared to the normal control lines. For example, TGF beta downstream genes, such as DKK1, BAMBI, IGFBP5, COL3A1, COLA1A1, SERPINE1, TGFBI and RGS5 were significantly increased in the DBA iPSCs compared to wild type control cells (Table 1Table 1TGF beta signaling that are upregulated in DBA iPSCs with RPS19 (p.Q126X) mutation.GENERPS19+/p.Q126XpCommentsFN13.6770.002Initiation and progression of matrix assemblyRGS52.0310.002Regulator of vascular remodelingTGFBI4.4410.000Interacts with Extracellular Matrix (ECM)IGFBP55.1720.001Regulator of IGF, interacts with ECM, regulator cell growth/differentiation/ apoptosisBAMBI1.7540.017TGF beta negative regulatorSERPINE12.620.001Inhibitor of fibrinolysis; Inhibits malignant cell invasionCOL3A11.9890.007Component of ECMCOL1A12.1320.001Component of ECMTGFB11.3710.025Regulates cell proliferation, differentiation and apoptosisDKK15.3810.001Negative regulator of Wnt signaling). We conclude that the variable penetrance of the RPS19 mutation in this family is due to modulation of its effect on ribosome biogenesis and that TGF beta signaling may be important in DBA pathogenesis. Whether the genetic factors responsible for the variable penetrance are inherited or acquired remains an important question. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2013 ◽  
Vol 122 (7) ◽  
pp. 1162-1173 ◽  
Author(s):  
Jung Bok Lee ◽  
Tamra E. Werbowetski-Ogilvie ◽  
Jong-Hee Lee ◽  
Brendan A. S. McIntyre ◽  
Angelique Schnerch ◽  
...  

Key Points Notch signaling controls hematopoiesis of human pluripotent stem cells.


2010 ◽  
Vol 34 (8) ◽  
pp. S36-S36
Author(s):  
Ping Duan ◽  
Xuelin Ren ◽  
Wenhai Yan ◽  
Xuefei Han ◽  
Xu Yan ◽  
...  

Sign in / Sign up

Export Citation Format

Share Document