Generation of improved human cerebral organoids from single copyDYRK1Aknockout induced pluripotent stem cells in trisomy 21: hypothetical solutions for neurodevelopmental models and therapeutic alternatives in down syndrome

2016 ◽  
Vol 40 (12) ◽  
pp. 1256-1270 ◽  
Author(s):  
E. Sacide Çağlayan
Blood ◽  
2010 ◽  
Vol 116 (21) ◽  
pp. 881-881
Author(s):  
Natsumi Nishihama ◽  
Yasuhiro Ebihara ◽  
Feng Ma ◽  
Wenyu Yang ◽  
Daisuke Tomizawa ◽  
...  

Abstract Abstract 881 Trisomy 21, genetic hallmark of Down syndrome, is the most frequent human chromosomal abnormality. Infants and children with Down Syndrome (DS) are known to have some hematological disorders with an increased risk of developing leukemia. Ten to 20% of newborn with DS are diagnosed as neonatal preleukemic status, Transient Myeloproliferative Disorder (TMD), and approximately 30% of TMD patients are predisposed to acute megakaryoblastic leukemia (AMKL). Recently, acquired mutations in the N-terminal activation domain of the GATA1 gene, leading to expression of a shorter GATA1 isoform (GATA1s), have been reported in AMKL and TMD (Wechsler et al., 2002; Mundschau et al., 2003), but neither patients nor mice with germline mutations leading to expression of GATA1s developed AMKL and TMD in the absent of trisomy 21. These findings suggested that trisomy 21 itself directly contributes to the development of AMKL and TMD. However, the role of trisomy 21 in hematopoiesis, particularly in the human fetus remains poorly understood. To better understand the effects of trisomy 21 on hematopoiesis in embryonic stage and leukemogenesis, we employed human induced pluripotent stem cells (hiPSCs) derived from patients with DS (DS-hiPSCs). Six DS-hiPS and 5 hiPS cell lines (control) from healthy donors, which we used here, were all created from skin fibroblasts and reprogrammed by the defined 3 or 4 reprogramming factors (OCT3/4, KLF4, and SOX2, or c-MYC in addition to the 3 factors, respectively). We generated blood cells from DS-hiPSCs and controls with coculture system using murine aorta-gonad-mesonephros (AGM)-derived stromal cell line (Ma et al., 2009). The cells from hiPSCs were harvested at D11 or D12 of coculture and analyzed the presence of hematopoietic markers and the potentials of hematopoietic colony formation. In the experiments using hiPSCs reprogrammed by 3 factors, human CD34 expression in harvested cells from DS-hiPSCs or controls were detected 10.06 ± 4.35% and 3.04%, respectively. CD45 expression of CD34+ cells was small proportion in both DS-hiPSCs and controls. We next examined the hematopoietic colony formation. Both myeloid and erythroid colonies were detected. Number of colonies formed from DS-hiPSCs was 43.7±11.1 to 74.3±11.2 per an iPSC colony. It's approximately 2 to 3.5 folds numbers of control (p-value<0.05). Similar results were obtained in the experiments using hiPSCs reprogrammed by 4 factors. These results indicated that hiPSCs derived from patients with Down syndrome could differentiate into multiple hematopoietic cell lineages and the differentiation into hematopoietic lineage was promoted in DS patients. Further researches are under investigation to identify the responsible genes in trisomy 21 for acceleration of hematopoiesis with microarray analysis. Our study may contribute to understanding of the effects of trisomy 21 on hematopoiesis and effective use of patients derived hiPSCs in research and clinical application. Disclosures: No relevant conflicts of interest to declare.


2013 ◽  
Vol 319 (4) ◽  
pp. 498-505 ◽  
Author(s):  
Huai-En Lu ◽  
Yao-Chen Yang ◽  
Sheng-Mei Chen ◽  
Hong-Lin Su ◽  
Pai-Cheng Huang ◽  
...  

Blood ◽  
2019 ◽  
Vol 134 (Supplement_1) ◽  
pp. 2738-2738
Author(s):  
Sonali Barwe ◽  
E. Anders Kolb ◽  
Anilkumar Gopalakrishnapillai

Down syndrome (DS) is recognized as one of the most important leukemia-predisposing syndromes. Specifically, 1-2% of DS children develop acute myeloid leukemia (AML) prior to age 5. AML in DS children (DS-AML) is characterized by the pathognomonic mutation in the gene encoding the essential hematopoietic transcription factor GATA1, resulting in N-terminally truncated mutant GATA1 (GATA1s). Trisomy 21 and GATA1s together induce a transient myeloproliferative disorder (TMD) exhibiting pre-leukemic characteristics. Approximately thirty percent of these cases progress into DS-AML by acquisition of additional somatic mutations in a step-wise manner. We employed disease modeling in vitro by the use of customizable induced pluripotent stem cells (iPSCs) (7, 8) to generate a TMD model. Isogenic iPSC lines derived from the fibroblasts of a DS patient with trisomy 21 and with disomy 21 were used. We also obtained DS2-iPS10 (iPSCs derived from DS patient fibroblast) from Prof. George Daley, Children's Hospital, Harvard University (Boston, MA). CRISPR (Clustered Regularly Interspaced Short Palindromic Repeats)/Cas9 system with the indicated guide sequence (Fig. 1A) was used to introduce clinically relevant GATA1 mutation in both disomic and trisomic iPSC lines. A representative plot of TIDE (Tracking of Indels by Decomposition) analysis showing 98% allelic mutation frequency of a clone with 2 bp deletion at chromosomal level (Fig. 1B) correlated with sequence analysis using Basic Local Sequence Alignment Tool (BLAST) and Sanger sequencing chromatogram (Fig. 1C). This mutation resulted in the disruption of first initiation codon and thus prevented the production of full length GATA1 protein, while allowing the usage of second initiation codon at 84th position to generate GATA1s. GATA1 and GATA1s are not expressed in iPSCs. To determine the expression of GATA1s, we differentiated these mutant iPSC lines into hematopoietic stem cell progenitors (HSPCs) using hematopoietic differentiation kit (StemCell Technologies) following a protocol depicted in Fig. 1D. The HSPCs derived from two distinct clones of trisomic iPSCs showed expression of full-length GATA1 protein and GATA1 mutant HSPCs lacked the expression of full-length GATA1 as expected (Fig. 1E). These HSPCs expressed GATA1s. Given that trisomy 21 promotes hematopoietic differentiation, an increase in the percentage of erythroid, megakaryoid and myeloid population was observed in trisomy 21 HSPCs with full length GATA1 (Fig. 1F, compare bars 1 and 3 in each category). The expression of GATA1s reduced erythroid lineage cells whereas it augmented megakaryoid and myeloid lineages in both disomy 21 (compare red and blue bars 1 and 2) and trisomy 21 backgrounds (compare bars 3 and 4). HSPCs derived from trisomy 21 iPSCs with GATA1s exhibited more megakaryoid expansion compared to the GATA1s in disomy 21 background (Fig. 1F, compare bars 2 and 4), in agreement with the synergistic function of trisomy 21 and GATA1s in promoting TMD. Transplantation of HSPCs derived from GATA1 mutated trisomic iPSCS into NSG-SGM3 mice showed the presence of human CD45+ cells in peripheral blood at 12 weeks post cell injection (Fig. 1G). In conclusion, we have developed a model system representing TMD, which can be used for step-wise modeling of Down-syndrome AML by introducing additional mutations. Figure 1 Disclosures No relevant conflicts of interest to declare.


Blood ◽  
2020 ◽  
Vol 136 (Supplement 1) ◽  
pp. 1-1
Author(s):  
Brahim Arkoun ◽  
Virginie Dufour ◽  
Aurélie Siret ◽  
Stefania Mazzi ◽  
Yasmine Mammasse ◽  
...  

Introduction Development of Acute megakaryoblastic leukemia in Down syndrome children (DS-AMKL) is a multi-step process. Acquired GATA1s mutation during fetal hematopoiesis is responsible of a transient myeloproliferative disorder (TMD) characterized by an accumulation of megakaryoblasts. Although most of TMD regress around birth, some TMD can progress from the initial GATA1s clone to AMKL through the acquisition of additional mutations, including in (i) the cohesin complex (i.e: SMC3), (ii) the JAK/STAT signaling pathway, such as MPL and (iii) the polycomb repressive complex 2 (EZH2). How these mutations cooperate to deregulate megakaryocyte (MK) differentiation and to induce a full-blown AMKL, along with the precise role of trisomy 21 (T21) during this transformation process remain unclear. Because modeling of DS-AMKL is particularly difficult in mice, we performed a step-wise introduction of GATA1s, a gain of function mutation of MPL (MPLW515K) and a heterozygous loss of function mutation in a cohesin (SMC3), separately or in combination, in T21 and isogenic disomic 21 (Dis21) human induced Pluripotent Stem Cells (iPSCs). Methods Trisomy 21 iPSCs were kindly provided by M. Weiss (Memphis, TN). CRISPR/Cas 9 genome editing of GATA1 or SMC3 allowed the generation of GATA1s T21, SMC3+/- T21 and GATA1s SMC3+/- T21 iPSC clones. CRISPR/Cas9-mediated knock-in of MPLW515K was performed in T21 GATA1s iPSCs. The subsequent T21 GATA1sMPLW515K/W515Kclones were selected as well as a revertant Dis21 GATA1sMPLW515K/W515Kclone. Finally, SMC3 insertion/deletion were obtained in isogenic T21 and Dis21 GATA1s MPLW515K/W515K SMC3+/-iPSCs clones. Hematopoietic differentiation was induced in 2D cultures in presence of a matrix and a cocktail of cytokines followed by a MK differentiation with SCF and TPO. MK differentiation was studied by clonogenic assays, flow cytometry, confocal microscopy and ultrastructural studies. Gene expression analyses were performed by RNA-seq on highly purified MK from all genotypes. Results GATA1s alone blocked MK maturation characterized by a persistent CD34 expression, an accumulation of abnormal large granules, a defect in the development of demarcation membranes (DMS), and a marked decrease in proplatelet formation. The typical GATA1s MK were large megakaryoblasts with numerous large granules and rare DMS. However, GATA1s alone had no effect on the clonogenic activity in CFU-MK assays and MK numbers. The introduction of the MPLW515K mutation did not modify this phenotype either in Dis21 or T21 GATA1s MK, but induced a complete TPO independence. SMC3+/- alone enhanced the MK maturation allowing the generation of a higher number of proplatelets-generating MK. Importantly, the combination of GATA1s and SMC3+/- mutations had a marked cooperative effect that worsened the MK maturation defect, led to the generation of abnormal megakaryoblasts with only a pre-DMS and resulted in enhanced proliferation and ploidization both in Dis21 and T21 iPSCs. Interestingly, the proliferation was markedly higher in T21 clones compared to Dis21 counterparts. RNA-seq and GSEA analyses showed that T21 GATA1s SMC3+/- mutant MK exhibited transcriptional signatures consistent with a dramatic decrease in the expression of maturation genes, including GATA1 target genes, while DNA replication gene markers were increased compared with GATA1s alone. T21 GATA1s MPLW515K/W515K SMC3+/- MK were enriched for AMKL signatures as compared to isogenic Dis21 GATA1sMPLW515K/W515K SMC3+/- MK. Ongoing ATAC-seq analyses will define the consequence of the different mutations on chromatin accessibility. Conclusion Using iPSC modeling, we analyzed in a human cell-context the consequences of the different combination of mutations associated with DS-AMKL that would be difficult to model using human primary cells. Our data demonstrate that GATA1s expression cooperates with SMC3+/- to enhance proliferation of megakaryoblasts from T21 iPSCs and isogenic Dis21 iPSCs hence reproducing the abnormalities observed in DS-AMKL. T21 is not directly involved in the MK differentiation defects but rather give a proliferative advantage supporting its role in leukemia development. Disclosures No relevant conflicts of interest to declare.


2017 ◽  
Vol 56 (2) ◽  
pp. 835-847 ◽  
Author(s):  
Erdem B. Dashinimaev ◽  
Alexander S. Artyuhov ◽  
Alexey P. Bolshakov ◽  
Ekaterina A. Vorotelyak ◽  
Andrey V. Vasiliev

2012 ◽  
Vol 11 (5) ◽  
pp. 615-619 ◽  
Author(s):  
Li B. Li ◽  
Kai-Hsin Chang ◽  
Pei-Rong Wang ◽  
Roli K. Hirata ◽  
Thalia Papayannopoulou ◽  
...  

2012 ◽  
Vol 3 (2) ◽  
pp. 14 ◽  
Author(s):  
Xiaoning Mou ◽  
Yuanbo Wu ◽  
Henghua Cao ◽  
Qingzhang Meng ◽  
Qihui Wang ◽  
...  

2017 ◽  
Vol 6 (6) ◽  
pp. 1465-1476 ◽  
Author(s):  
Young M. Lee ◽  
Bruna L. Zampieri ◽  
Jonah J. Scott-McKean ◽  
Mark W. Johnson ◽  
Alberto C.S. Costa

2014 ◽  
Vol 16 (5) ◽  
pp. 331-344 ◽  
Author(s):  
Caterina Pipino ◽  
Sayandip Mukherjee ◽  
Anna L David ◽  
Michael P. Blundell ◽  
Steven W. Shaw ◽  
...  

Sign in / Sign up

Export Citation Format

Share Document