scholarly journals Silencing of p53 and CDKN1A establishes sustainable immortalized megakaryocyte progenitor cells from human iPSCs

Author(s):  
Masamitsu Sone ◽  
Sou Nakamura ◽  
Sachiko Umeda ◽  
Harumi Ginya ◽  
Motohiko Oshima ◽  
...  
Keyword(s):  
Blood ◽  
2016 ◽  
Vol 128 (22) ◽  
pp. 1474-1474 ◽  
Author(s):  
Michail Zaboikin ◽  
Tatiana Zaboikina ◽  
Carl E. Freter ◽  
Narasimhachar Srinivasakumar

Abstract Gene and cell therapy for hemophilia A requires the use of the appropriate target cell for genetic modification and, given the advances in genome editing, an approach that can be applied universally for the wide variety of genetic mutations in the coagulation factor VIII gene (F8) responsible for hemophilia A. Recent studies using two different conditional knockout mouse models showed that the principal, and possibly exclusive, source for FVIII in the circulation are endothelial cells (Everett LA et al. Blood 2014.123: 3697; Fahs SA et al. Blood 2014.123: 3706). Since endothelial cells are present in all the major organs previously thought to produce coagulation factor VIII (FVIII), these studies provide the basis for the earlier reports indicating different tissues (liver, lung, spleen, lymphatic tissue) as sources of FVIII. ECs conveniently express von Willebrand factor (vWF) that is essential for the stability of FVIII. The precursor of ECs, endothelial progenitor cells (EPCs), have been isolated from adult human peripheral blood and cord blood. EPCs can readily integrate into existing vascular system upon intravenous injection. EPCs are quite rare in peripheral blood (about 20 colony forming cells per 100 mL of blood). Moreover, EPCs derived from adult peripheral blood have lower proliferative potential than those obtained from cord blood (Ingram DA. Blood. 2004. 104: 2752). In contrast, induced pluripotent stem cells (iPSCs), that are more amenable for expansion, can be readily differentiated into EPCs. Studies have also shown that iPSC-derived EPCs when injected intrahepatically in mice integrated into liver sinusoids, resulted in therapeutic levels of FVIII production (Xu D et al. PNAS 2009. 106: 808). Here we describe an optimized in vitro differentiation protocol for derivation of EPCs from iPSCs. We have previously reported the generation and characterization of human iPSCs from lung fibroblasts (Srinivasakumar et al. PeerJ. 2013;1:e224). In this study we used human iPSCs generated from adult dermal fibroblasts using Yamanaka's non-integrating Epstein-Barr based episomal vectors. We used a step-wise differentiation protocol for obtaining EPCs that was a combination of a method for differentiation of iPSCs into hematopoietic progenitors (Fig A, Steps 1 & 2) to generate hemangioblasts (Niwa A et al. PLoS ONE 6(7): e22261), and a protocol for obtaining EPCs from peripheral blood (Step 3) (Mead LE et al. Current Protocols in Stem Cell Biol. 2C.1.1-2C.1.27). A sorting step after differentiation into hemangioblasts followed by a final round of sorting after step 3 yielded >90% pure population of EPC that exhibited the canonical cell surface markers: CD31 and CD34, and absence of CD45 (Fig B). The cells also took up fluorophore-conjugated acetylated LDL (acLDL-A488) that was inhibited with 50x excess of unlabeled acLDL (Fig C). Immunofluorescence staining for vWF revealed characteristic staining reminiscent of Weibel-Palade bodies in the cytoplasm (Fig D). The cells exhibited the typical tube formation ability in Matrigel (Fig E). Additional studies are needed to determine the proliferative potential of these cells and their ability to integrate into vasculature. To address the myriad mutations shown to be responsible for hemophilia A, we have designed high efficiency dimeric guide RNAs (as part of a separate study) (Zaboikin M et al. Manuscript in preparation) for use with the CRISPR/dCas9-Fok1 system (Tsai SQ et al. Nat Biotechnol. 2014. 32:569) for precise modification at the F8 locus downstream of the first coding exon. We also showed in that study the replacement of target sequence at the site with that of a donor template sequence with desired attributes. We hypothesize that using a donor template that encodes the F8 promoter driving a functional F8 cDNA for homology directed repair at the target double stranded break site will provide an universal solution for the large variety of mutations observed in hemophilia A. Results of genome editing of iPSCs using the above mentioned CRISPR/dCas9-Fok1 system (together with the donor template) followed by the differentiation of genetically modified iPSCs into EPCs will be presented. Figure Figure. Disclosures No relevant conflicts of interest to declare.


Leukemia ◽  
2021 ◽  
Author(s):  
Elżbieta Karnas ◽  
Małgorzata Sekuła-Stryjewska ◽  
Katarzyna Kmiotek-Wasylewska ◽  
Sylwia Bobis-Wozowicz ◽  
Damian Ryszawy ◽  
...  

AbstractCord blood (CB) represents a source of hematopoietic stem and progenitor cells (CB-HSPCs) for bone marrow (BM) reconstitution, but clinical CB application is limited in adult patients due to the insufficient number of CB-HSCPCs and the lack of effective ex vivo approaches to increase CB-HSPC functionality. Since human-induced pluripotent stem cells (hiPSCs) have been indicated as donor cells for bioactive extracellular vesicles (EVs) modulating properties of other cells, we are the first to employ hiPSC-derived EVs (hiPSC-EVs) to enhance the hematopoietic potential of CB-derived CD45dimLin-CD34+ cell fraction enriched in CB-HSPCs. We demonstrated that hiPSC-EVs improved functional properties of CB-HSPCs critical for their hematopoietic capacity including metabolic, hematopoietic and clonogenic potential as well as survival, chemotactic response to stromal cell-derived factor 1 and adhesion to the model components of hematopoietic niche in vitro. Moreover, hiPSC-EVs enhanced homing and engraftment of CB-HSPCs in vivo. This phenomenon might be related to activation of signaling pathways in CB-HSPCs following hiPSC-EV treatment, as shown on both gene expression and the protein kinases activity levels. In conclusion, hiPSC-EVs might be used as ex vivo modulators of CB-HSPCs capacity to enhance their functional properties and augment future practical applications of CB-derived cells in BM reconstitution.


Blood ◽  
2016 ◽  
Vol 128 (22) ◽  
pp. 425-425
Author(s):  
Yueying Li ◽  
Chen Jin ◽  
Hao Bai ◽  
Shu Sun ◽  
Paul P. Liu ◽  
...  

Abstract Megakaryocytes (MK), which produce platelets, play important roles in blood coagulation and hemostasis. The master transcription factor RUNX1 regulates lineage-specific transcriptional targets and key signaling pathways, and is known to be essential for megakaryopoiesis. Mono-allelic RUNX1 mutations lead to familial platelet disorder (FPD), which is characterized by thrombocytopenia and abnormal platelet functions. A high percentage (~50%) of these FPD patients later develop myelodysplastic syndromes and acute myeloid leukemia. The exact mechanisms underlying deregulated megakaryopoiesis in FPD remain unclear, partially due to the lack of an adequate experimental model mimicking the human disease. For example, engineered laboratory mice and zebrafish with only one copy of the Runx1 gene do not develop bleeding disorders or leukemia. Using an in vitro hematopoietic differentiation system, we found that megakaryocytic differentiation from FPD-derived induced pluripotent stem cells (iPSCs) were defective (Connelly et al., 2014). Targeted correction of the mutated RUNX1 allele by genome editing restored the MK production and functions, validating the central role of RUNX1 in megakaryopoiesis (Connelly et al., 2014). In this new study, we pursued the hypothesis that direct target genes regulated by RUNX1 play important roles in human megakaryopoiesis. We first performed RNA-Seq analysis on differentiated hematopoietic cells from FPD-iPSCs (harboring a mono-allelic RUNX1 mutation) and RUNX1-corrected isogenic iPSCs. Seventy-nine genes were expressed at a significantly higher level (p<0.01, FDR<0.05) while 93 genes were expressed at a significantly lower level (p<0.01, FDR<0.05) in the RUNX1-corrected cells as compared to the FPD-iPSCs. To determine whether these differentially expressed genes (DEGs) are the direct targets of RUNX1, we additionally performed genome-wide location analysis of RUNX1 by ChIP-Seq using the same hematopoietic cell population differentiated from the RUNX1-corrected isogenic iPSCs. We detected 5266 (FDR<0.05) binding sites in 4526 gene loci. Combined with the DEG data from RNA-Seq analyses, we further identified 37 up-regulated genes (such as ITGB3 and PF4) and 27 down-regulated genes with RUNX1 binding to the gene's proximity. Among the 64 differentially expressed genes with RUNX1 binding, Gene Ontology (GO) analysis revealed that only 13 genes including PF4 have been reported to be relevant to megakaryopoiesis. In order to verify the roles of these RUNX1 target genes in hematopoiesis and megakaryopoiesis, we carried out gene knockout (KO) experiments by CRISPR-Cas9 in normal human iPSCs followed by in vitro hematopoietic differentiation assays. We first focused on the "down-regulated" genes by RUNX1 binding, with the hypothesis that their KO may enhance hematopoiesis and/or megakaryopoiesis from normal iPSCs. One of such genes is NOTCH4, a member of NOTCH receptor family that plays important roles in development and cell fate determination. A previous study showed that NOTCH signaling specifies MK development from mouse hematopoietic progenitor cells (Mercher et al., 2008), while we have not seen publications on the NOTCH4 in human MK development. Using the improved CRISPR technology, we successfully achieved KO of one copy of NOTCH4 in the wildtype iPSCs. We found that heterozygous KO of NOTCH4 increased MK (progenitor) production by 95% (p<0.05), while the production of CD34+ multipotent hematopoietic progenitor cells were not affected. To further verify its function, we inhibited NOTCH4 signaling with a gamma-secretase inhibitor. Notably, inhibition of NOTCH4 signal starting at day 2 of hematopoietic differentiation improved the efficiency of MK progenitor production by 50% (p<0.05) and more mature MK production by 70% (p<0.05). Taken together, we conclude that NOTCH4, a newly discovered RUNX1 target gene, negatively regulates megakaryopoiesis in a developmental-stage specific manner. Unlocking this inhibitory effect by small molecule inhibitors can promote MK production ex vivo. The described approach will enable us to discover additional novel genes that influence human hematopoiesis and megakaryopoiesis, which in turn will help to promote ex vivo generation of MKs from human iPSCs or postnatal hematopoietic stem/progenitor cells. Disclosures No relevant conflicts of interest to declare.


2010 ◽  
Vol 34 (8) ◽  
pp. S41-S41
Author(s):  
Yang Bi ◽  
Yun He ◽  
Tingyu Li ◽  
Tao Feng ◽  
Tongchuan He

2000 ◽  
Vol 111 (1) ◽  
pp. 363-370 ◽  
Author(s):  
Katsuto Takenaka ◽  
Mine Harada ◽  
Tomoaki Fujisaki ◽  
Koji Nagafuji ◽  
Shinichi Mizuno ◽  
...  

2001 ◽  
Vol 120 (5) ◽  
pp. A542-A543
Author(s):  
T HIROSE ◽  
K YASUCHIKA ◽  
T FUJIKAWA ◽  
H FUJII ◽  
S OE ◽  
...  

Sign in / Sign up

Export Citation Format

Share Document