scholarly journals Chromosomal translocation t(11;14) and p53 deletion induced by the CRISPR/Cas9 system in normal B cell-derived iPS cells

2021 ◽  
Vol 11 (1) ◽  
Author(s):  
Yusuke Azami ◽  
Naohiro Tsuyama ◽  
Yu Abe ◽  
Misaki Sugai-Takahashi ◽  
Ken-ichi Kudo ◽  
...  

AbstractMultiple myeloma (MM) cells are derived from mature B cells based on immunoglobulin heavy chain (IgH) gene analysis. The onset of MM is often caused by a reciprocal chromosomal translocation (cTr) between chr 14 with IgH and chr 11 with CCND1. We propose that mature B cells gain potential to transform by reprograming, and then chromosomal aberrations cause the development of abnormal B cells as a myeloma-initiating cell during B cell redifferentiation. To study myeloma-initiating cells, we have already established normal B cell-derived induced pluripotent stem cells (BiPSCs). Here we established two BiPSCs with reciprocal cTr t(11;14) using the CRISPR/Cas9 system; the cleavage site were located in the IgH Eμ region of either the VDJ rearranged allele or non-rearranged allele of IgH and the 5′-upsteam region of the CCND1 (two types of BiPSC13 with t(11;14) and MIB2-6 with t(11;14)). Furthermore, p53 was deleted using the CRISPR/Cas9 system in BiPSC13 with t(11;14). These BiPSCs differentiated into hematopoietic progenitor cells (HPCs). However, unlike cord blood, those HPCs did not differentiated into B lymphocytes by co-culture with BM stromal cell. Therefore, further ingenuity is required to differentiate those BiPSCs-derived HPCs into B lymphocytes.

Blood ◽  
2018 ◽  
Vol 132 (Supplement 1) ◽  
pp. 1896-1896
Author(s):  
Aki Yanagi ◽  
Naohiro Tsuyama ◽  
Yukari Yanai ◽  
Yu Abe ◽  
Misaki Sugai ◽  
...  

Abstract We previously established normal B cell-derived induced pluripotent stem cells (BiPSCs; BiPSC13 and MIB2-6). BiPSCs are known to maintain VDJ rearrangement of the IgH gene, and they can be induced by the tet-off system to express activation-induced cytidine deaminase (AID; BiPSC13-AID and MIB2-6-AID) and differentiate into hematopoietic progenitor cells (HPCs) (Scientific Rep, 2017). Using these BiPSCs, we attempted to prove the existence of abnormal B cells, which are thought to be myeloma-initiating cells, originating from mature B cells transformed by reprogramming. We speculated that BiPSCs could develop into myeloma-initiating cells that undergo chromosomal translocation or gain genetic abnormalities during redifferentiation into mature B cells. First, using the comet assay, we confirmed the DNA-damaging effect of AID in BiPSCs-AID. Secondly, we differentiated BiPSC13-AID into CD34+/CD38-/CD43-/CD45- cells by co-culture with stromal cells (mouse embryo cell line: 10T1/2), and we subsequently transplanted the cells into the bone marrow (BM) of immunodeficient NRG mice. The presence of CD34+ cells was still observed in mouse BM 4 months after transplantation; however, no differentiation into B cells was detected. Next, using the CRISPR/Cas9 system, we attempted to make BiPSCs with chromosomal translocation t(11;14); we succeeded in establishing a 293T cell line with t(11;14), then confirmed t(11;14) in MIB2-6-AID, and the clone is now being established. Furthermore, we established BiPSC13-Pax5, which can be induced by the tet-off system to express Pax5, and we then differentiated BiPSC13-Pax5 into CD34+/Pax5+/CD38-/CD43-/CD45- cells by co-culture with stromal cells (10T1/2). We expect that the HPCs or hematopoietic stem cells (HSCs) derived from BiPSCs will further differentiate into B cells due to the expression of Pax5 in the BM of NRG mouse. We also established BiPSC13-AID-p53-/-, in which p53 was deleted using the CRISPR/Cas9 system, and the cells differentiated into HPCs. Interestingly, we detected some CD43+/CD45+ cells among CD34+/CD38- cells after the co-culture of BiPSC13-AID with aorta-gonad-mesonephros-derived stromal cell (AGM-S3) instead of 10T1/2. Therefore, AGM-S3 may promote the differentiation of BiPSCs into cells that are more similar to HSCs. These CD34+ cells differentiated from BiPSCs will be transplanted into the BM of NRG mouse. Disclosures Hanamura: CHUGAI PHARMACEUTICAL CO., LTD.: Research Funding; Kyowa Hakko Kirin Company, Limited: Research Funding; Fujimoto Pharmaceutical Corporation: Research Funding; Takeda Pharmaceutical Company Limited.: Other: Lecture fee; Bristol-Myers Squibb: Other: Lecture fee, Research Funding; Celgene: Other: Lecture fee.


Blood ◽  
2019 ◽  
Vol 134 (Supplement_1) ◽  
pp. 2736-2736
Author(s):  
Tina Nilsson ◽  
Ahmed Waraky ◽  
Anders Östlund ◽  
Laleh Arabanian ◽  
Julia Asp ◽  
...  

Introduction Acute myeloid leukemia (AML) is the result of aberrant hematopoietic processes, such as enhanced proliferation, blocked differentiation, and dysregulated apoptosis of hematopoietic stem and progenitor cells, and frequently these changes are initiated by chromosomal translocations in leukemia. Efficient methods for modelling leukemia and to recreate leukemia-associated genetic aberrations, such as chromosome translocations, are therefore crucial for investigating how leukemia is initiated. Today, most such models are murine and usually based on introduction of fusion gene transcripts of interest under the control of a constitutive active promoter using lenti- or retroviral transduction rather than the chromosomal translocation itself. The aim of the current project was to create a human cellular model of a chromosomal translocation that is typically found in AML in children under 24 months of age, the translocation t(7;12)(q36;p13). This translocation has been associated with poor prognosis, and leads to a gene fusion MNX1-ETV6 but also aberrant MNX1 gene expression. Its mechanism for leukemia initiation is so far unknown, mainly due to lack of a suitable experimental model. Material and methods CRISPR/Cas9 was used to reconstruct the genetics of the t(7;12)(q36;p13) rearrangement in human induced pluripotent stem cells (iPSC) while maintaining the genomic architecture and regulatory elements. Ribonucleoprotein (RNP) complex was delivered by lipofection (Nucleofection, Amaxa 4D system) into undifferentiated iPSC (ChiPSC 22, Cellartis). An ATTO550 tag on tracrRNA/RNP complex was used to sort out positive cells by flow cytometry and then seeded as single-cells in 96-well plates. Genomic DNA from the single-cell derived iPSC clones were screened by PCR for the presence of the translocation and positive clones were verified with a FISH probe specific for t(7;12)(q36;p13) (Double Fusion Break Apart probe, Metasystem). RT-qPCR was used to detect and quantify the expression of MNX1-ETV6 fusion and MNX1 transcripts. Differentiation potential was tested with the Trilineage Differentiation and Hematopoietic Kits (STEMdiff, STEMCELL Technologies). Results Using CRISPR/Cas9, we could successfully generate iPSC with the t(7;12)(q36;p13) translocation. The translocation was confirmed using conventional karyotyping and FISH and the mRNA expression of the fusion was confirmed with RT-qPCR. No additional chromosomal aberrations were seen. The t(7;12)(q36;p13) iPSC showed similar growth and differentiation properties as the parental iPSC. They showed propensity to differentiate into all three germ layers, confirming their pluripotent stem cell properties. The potential for differentiation into hematopoietic progenitor cells was shown by expression of CD34+, CD43+ and CD45+. In AML with t(7;12)(q36;p13), MNX1 mRNA expression is increased and this may play a role for leukemia development. In the t(7;12)(q36;p13) iPSC, RT-qPCR indeed showed increased expression of MNX1 expression compared with iPSC without the translocation. This increase of MNX1 was not seen in murine adult bone marrow or fetal liver cells transduced with retrovirus expressing the MNX1-ETV6 fusion. Further characterization of the t(7;12)(q36;p13) iPSC, e.g. whole exome and transcriptome sequencing and engraftment potential in immunocompromised mice (NSG-SGM), is ongoing. Conclusion In summary, we have using CRISPR/Cas9 successfully created a t(7;12)(q36;p13) iPSC line with potential to differentiate into hematopoietic progenitor cells and with gene expression pattern similar to what is seen in human AML samples with the t(7;12)(q36;p13). The introduction of the MNX1-ETV6 fusion in its correct genomic context could recapitulate local gene regulation, making it superior to models based on lenti- or retroviral introduction of fusion genes transcripts. In conclusion, this created cell line will be a valuable tool to study the mechanisms behind t(7;12)(q36;p13) AML, a severe form of AML associated with poor prognosis. Disclosures No relevant conflicts of interest to declare.


Blood ◽  
2020 ◽  
Vol 136 (Supplement 1) ◽  
pp. 18-19
Author(s):  
Misaki Sugai ◽  
Naohiro Tsuyama ◽  
Yu Abe ◽  
Yusuke Azami ◽  
Kenichi Kudo ◽  
...  

The cellular origin of multiple myeloma (MM) has not yet been identified. Based on immunoglobulin heavy chain (IgH) gene analysis, myeloma cells are derived from mature B cells. Chromosomal aberrations such as trisomy and chromosomal translocation (cTr) play a critical role in the early tumorigenesis of MM. We hypothesized that the abnormal cells from which myeloma cells originate might be mature B lymphocytes with chromosomal or genetic changes in the reprogrammed state that enable them to acquire the potential to become tumors in the process of redifferentiation into B lymphocytes. We established induced pluripotent stem cells (iPSs) from normal B lymphocytes (BiPSCs: BiPSC13 & MIB2-6); these BiPSCs have the same VDJ rearrangement of IgH as the original B lymphocytes and differentiate into CD34+/CD38- hematopoietic progenitor cells co-culture with stromal cells, AGM-S3 (Sci Rep, 2017). We then established a method to induce reciprocal cTr t(11;14), which is a reciprocal cTr between IgH and CCND1 and the most frequent cTr in MM, using the CRISPR/Cas9 system; cTr was induced by infection of IgH-CCND1 lentiCRISPRv2 lentivirus, which targets the human IgH Eµ region and 13kb upstream of the CCND1 coding sequence, to BiPSCs (Oncol Lett, 2019). Subsequently, we established cell lines carrying reciprocal cTr t(11;14) between CCND1 and either an allele in which VDJ rearrangement of IgH had been completed or an allele in which VDJ rearrangement had not been completed (stopped at DJ joining) in BiPSC13 t(11;14) (AZ & AX) and MIB2-6 t(11;14) (BC & BG), respectively. These BiPSCs differentiated into CD34+/CD38-/CD45+/-/CD43+/- hematopoietic progenitors cells in co-culture with AGM-S3 or in stem cell differentiation medium; this was subsequently confirmed by the differentiation into granulocytes, macrophages, and erythroblasts in a colony-formation assay. We are now trying to produce BiPSCs in which cTr t(11;14) is induced when they differentiate into mature B cells expressing CD27. First, we used the Cre-loxP recombination system to induce cTr t(11;14) in BiPSCs. BiPSCs were transfected with IgH loxP-Neo-loxP knock-in vector and IgH lentiCRISPRv2 vector. Subsequently, G418-resistant BiPSCs carrying loxP-Neo-loxP in IgH were transfected with iCre-EGFP. After removing the loxP-Neo site from EGFP-positive cells, BiPSCs carrying IgH-loxP were transfected with CCND1 loxP-FRT3-Neo-FRT3 knock-in vector and CCND1 lentiCRISPRv2 vector. Subsequently, G418-resistant BiPSCs carrying IgH-loxP in IgH and loxP-FRT3-Neo-FRT3 in CCND1 were transfected with Flpo-EGFP. After removing the FRT3-Neo site from EGFP-positive cells, BiPSCs carrying IgH-loxP in IgH and CCND1-loxP-FRT3 in CCND1 were transfected with iCre-HygR. Hygromycin B-resistant cells were picked, the reciprocal cTr t(11;14) was confirmed by polymerase chain reaction, and we established BiPSCs with der(11)t(11;14) and BiPSCs with der(14)t(11;14). We also developed a system in which Cre is expressed along with CD27 expression in the B cell lymphoma cell line Raji. These BiPSCs could be useful for the study of myeloma-initiating cells, but whether they would be able to be redifferentiated into B lymphocyte is important. Disclosures Hanamura: Mundipharma K.K.: Honoraria, Membership on an entity's Board of Directors or advisory committees, Speakers Bureau; CSL Behring: Honoraria, Membership on an entity's Board of Directors or advisory committees, Speakers Bureau; MSD K.K.: Honoraria, Membership on an entity's Board of Directors or advisory committees, Research Funding, Speakers Bureau; Sanofi K.K.: Research Funding; Otsuka Pharmaceutical Co., Ltd.: Honoraria, Membership on an entity's Board of Directors or advisory committees, Speakers Bureau; SHIONOGI Co., Ltd.: Honoraria, Membership on an entity's Board of Directors or advisory committees, Speakers Bureau; Novartis Pharma K.K.: Honoraria, Membership on an entity's Board of Directors or advisory committees, Research Funding, Speakers Bureau; DAIICHI SANKYO COMPANY, LIMITED: Honoraria, Membership on an entity's Board of Directors or advisory committees, Speakers Bureau; Kyowa Kirin Co., Ltd.: Honoraria, Membership on an entity's Board of Directors or advisory committees, Speakers Bureau; Eisai Co., Ltd.: Honoraria, Membership on an entity's Board of Directors or advisory committees, Speakers Bureau; NIPPON SHINYAKU CO.,LTD.: Honoraria, Membership on an entity's Board of Directors or advisory committees, Speakers Bureau; Pfizer Japan Inc.: Honoraria, Membership on an entity's Board of Directors or advisory committees, Speakers Bureau; AbbVie Inc.: Honoraria, Membership on an entity's Board of Directors or advisory committees, Speakers Bureau; Takeda Pharmaceutical Company Limited: Honoraria, Membership on an entity's Board of Directors or advisory committees, Research Funding, Speakers Bureau; Janssen Pharmaceutical K.K.: Honoraria, Membership on an entity's Board of Directors or advisory committees, Speakers Bureau; Celgene Corporation: Honoraria, Membership on an entity's Board of Directors or advisory committees, Research Funding, Speakers Bureau; Ono Pharmaceutical Co., Ltd.: Honoraria, Membership on an entity's Board of Directors or advisory committees, Research Funding, Speakers Bureau; Bristol-Myers Squibb: Honoraria, Membership on an entity's Board of Directors or advisory committees, Research Funding, Speakers Bureau.


2011 ◽  
Vol 2 (6) ◽  
pp. 46 ◽  
Author(s):  
Seiga Ohmine ◽  
Allan B Dietz ◽  
Michael C Deeds ◽  
Katherine A Hartjes ◽  
David R Miller ◽  
...  

Blood ◽  
2019 ◽  
Vol 134 (Supplement_1) ◽  
pp. 3057-3057
Author(s):  
Aki Yanagi ◽  
Naohiro Tsuyama ◽  
Misaki Sugai ◽  
Yu Abe ◽  
Yusuke Azami ◽  
...  

The cellular origin of multiple myeloma (MM) has not ben identified. Based on the results of transplantation experiments using bone marrow samples from MM patients in immunodeficient mice, so-called myeloma stem cells have been inferred to be present in CD19-/CD38++/CD138+ or CD138- plasma cells populations; however, it is possible that the results indicated the presence of plasma cell populations with cell proliferation ability rather than the cellular origin of myeloma cells. On the other hand, based on immunoglobulin heavy chain (IgH) gene analysis, myeloma cells are derived from mature B cells. Chromosomal aberrations such as trisomy and chromosomal translocation play a critical role in early tumorigenesis of MM. We hypothesize that the reprograming of mature B cells, in which IgH gene rearrangements have maintained, are the origin of MM. We propose that mature B cells gain potential by reprograming, and then chromosomal aberrations cause the development of abnormal B cells as a myeloma-initiating cell during B cell redifferentiation. To identify myeloma-initiating cells, we established normal B cell-derived induced pluripotent stem cells (BiPSCs; BiPSC13 and MIB2-6). These BiPSCs maintain VDJ rearrangement of the IgH gene, and they can be induced by the tet-off system to express activation-induced cytidine deaminase (AID; BiPSC13-AID and MIB2-6-AID) and differentiate into hematopoietic progenitor cells (HPCs) (Scientific Rep, 2017). Subsequently, we used the CRISPR/Cas9 system (Oncology Letters, 2019) to establish two BiPSCs with chromosomal translocation t(11;14); the cleavage site were located in the IgH Eμ region of the VDJ non-rearranged allele of the IgH gene and the CCND1 5'-upsteam region of the CCND1 gene (BiPSC13 with t(11;14) and MIB2-6 with t(11;14)). Furthermore, p53 was deleted using the CRISPR/Cas9 system (Ota A, J Cell Sci, 2017) in BiPSC13 with t(11;14) and BiPSC13-AID with t(11;14). These BiPSCs differentiated into CD34+/CD38-/CD45+/-/CD43- HPCs in co-culture with stromal cell, AGM-S3, and their ability to subsequently differentiate into granulocytes, macrophages, and erythroblasts was confirmed by colony-formation assay. Until now, the co-culture of BiPSC13, MIB2-6, and MIB2-6 with t(11;14) with AGM-S3 followed by co-culture with stromal cell, MS-5, showed the appearance of CD34-/CD45+ and CD34-/CD10+ cell population. Also of interest, by RT-PCR, the expression levels of E2F, EBF, GATA3 and CD10 were higher in CD34+ HPCs differentiated by the co-culture of BiPSC13 and MIB2-6 with AGM-S3 when compared to the levels in cord blood-derived CD34+ cells. Overall, the results suggest that these BiPSCs have the ability to differentiate into HPCs, and that they may further differentiate into B cells. If these BiPSCs could be differentiated into mature B cells, they may be useful for the elucidation and study of myeloma-initiating cells derived from mature B cells. Disclosures Hanamura: Mundi: Honoraria; Yamada Yohojo: Research Funding; Celgene: Consultancy, Honoraria, Research Funding; Nihon Shinyaku: Honoraria, Research Funding; Fukuyu Hospital: Research Funding; Fujimoto: Research Funding; Taiho: Research Funding; Ono: Consultancy, Honoraria, Research Funding; Otsuka: Honoraria, Research Funding; Bristol-Myers Squibb: Consultancy, Honoraria, Research Funding; Novartis: Consultancy, Honoraria, Research Funding; Chugai: Research Funding; Eisai: Consultancy, Honoraria; Janssen: Consultancy, Honoraria; Eli Lilly: Research Funding; Asai Clinic: Research Funding; AbbVie: Honoraria; MSD: Research Funding; Daiichi Sankyo: Consultancy, Honoraria, Research Funding; Zenyaku: Research Funding; Sanofi: Research Funding; Shionogi: Honoraria, Research Funding; Kyowa Kirin: Consultancy, Honoraria, Research Funding; Takeda: Consultancy, Honoraria, Research Funding; Astellas: Research Funding; Pfizer: Honoraria, Research Funding.


Sign in / Sign up

Export Citation Format

Share Document