scholarly journals Generation of SIV resistant T cells and Macrophages from Nonhuman Primate Induced Pluripotent Stem Cells with Edited CCR5 locus

2021 ◽  
Author(s):  
Saritha S D'Souza ◽  
Akhilesh Kumar ◽  
Jason Weinfurter ◽  
Mi Ae Park ◽  
John Maufort ◽  
...  

Adoptive therapies with genetically modified somatic T cells rendered HIV resistant have shown promise for AIDS therapy. A renewable source of HIV resistant human T cells from induced pluripotent stem cells (iPSCs) would further facilitate and broaden the applicability of these therapies. Here, we report successful targeting of the CCR5 locus in iPSCs generated from peripheral blood T cells (T-iPSCs) or fibroblasts (fib-iPSCs) from Mauritian Cynomolgus macaques (MCM), using CRISPR/Cas9 technology. We found that CCR5 editing does not affect pluripotency or hematopoietic and T cell differentiation potentials of fib-iPSCs. However, deletion of CCR5 in T-iPSCs leads to selective loss of their T cell redifferentiation potential without affecting myeloid development. T cells and macrophages produced from CCR5-edited MCM- iPSCs did not support replication of the CCR5-tropic simian immunodeficiency viruses SIVmac239 (T-cell tropic) and SIVmac316 (macrophage-tropic). Overall, these studies provide a platform for further exploration of AIDS therapies based on gene-edited iPSCs in a nonhuman primate preclinical model.

2021 ◽  
Vol 12 ◽  
Author(s):  
Fei Lu ◽  
Xiao-Jing-Nan Ma ◽  
Wei-Lin Jin ◽  
Yang Luo ◽  
Xun Li

Immunotherapy has become an indispensable part of the comprehensive treatment of hepatocellular carcinoma (HCC). Immunotherapy has proven effective in patients with early HCC, advanced HCC, or HCC recurrence after liver transplantation. Clinically, the most commonly used immunotherapy is immune checkpoint inhibition using monoclonal antibodies, such as CTLA-4 and PD-1. However, it cannot fundamentally solve the problems of a weakened immune system and inactivation of immune cells involved in killing tumor cells. T cells can express tumor antigen-recognizing T cell receptors (TCRs) or chimeric antigen receptors (CARs) on the cell surface through gene editing to improve the specificity and responsiveness of immune cells. According to previous studies, TCR-T cell therapy is significantly better than CAR-T cell therapy in the treatment of solid tumors and is one of the most promising immune cell therapies for solid tumors so far. However, its application in the treatment of HCC is still being researched. Technological advancements in induction and redifferentiation of induced pluripotent stem cells (iPSCs) allow us to use T cells to induce T cell-derived iPSCs (T-iPSCs) and then differentiate them into TCR-T cells. This has allowed a convenient strategy to study HCC models and explore optimal treatment strategies. This review gives an overview of the major advances in the development of protocols to generate neoantigen-specific TCR-T cells from T-iPSCs. We will also discuss their potential and challenges in the treatment of HCC.


2021 ◽  
Author(s):  
Shoichi Iriguchi ◽  
Yutaka Yasui ◽  
Yohei Kawai ◽  
Suguru Arima ◽  
Mihoko Kunitomo ◽  
...  

Abstract Clinical efficacy demonstrated by chimeric antigen receptor T cell therapy call for further development that could broaden their applicability. One such direction is to develop alternate T-cell sources and T cells differentiated from pluripotent stem cells may be an ideal candidate.The present protocol provides a feeder-free and scalable method to generate T lymphocytes from induced pluripotent stem cells.


Blood ◽  
2014 ◽  
Vol 124 (21) ◽  
pp. 3232-3232
Author(s):  
Kazuhisa Chonabayashi ◽  
Masahiro Kawahara ◽  
Keisuke Okita ◽  
Masatoshi Nishizawa ◽  
Norimitsu Kadowaki ◽  
...  

Abstract Myelodysplastic syndromes (MDS) are a heterogeneous group of clonal stem cell diseases characterized by inefficient hematopoiesis and risk of progression to acute myeloid leukemia with poor prognosis. Although massive parallel sequencing studies have revealed a number of genomic alterations associated with MDS, functional consequences of these alterations remain poorly understood, mainly due to a difficulty in the ex vivo culture of primary MDS cells and a lack of good animal models. Induced pluripotent stem cells (iPSCs) from MDS patients are expected to provide a new platform for elucidation of the pathogenesis of MDS. We attempted to generate iPSCs from peripheral blood mononuclear cells of a MDS patient (RAEB-1 by WHO classification) with chromosome 20q deletion, using episomal methods. We successfully established more than 30 iPSC lines derived from Non-T cells as well as 6 iPSC lines derived from T cells at the same time. Karyotyping and SNP-CGH analysis revealed that most of the Non-T-cell-derived iPSC lines (Del20q-iPSC lines) have the isolated 20q deletion at q11.2-13.1 identical to those of the primary MDS cells, whereas all T-cell-derived iPSC lines (NK-T-iPSC lines) have normal karyotype. In order to evaluate chromosome stability, we validated karyotype of 3 randomly selected Del20q-iPSC lines after 30 passages and found no additional chromosomal aberrations other than deletion 20q. Del20q-iPSC lines displayed characteristic morphology and expressed pluripotent stem cell markers at the levels comparable to those in isogenic NK-T-iPSC lines and ES cell lines. Nine randomly selected Del20q-iPSC lines and all 6 NK-T-iPSC lines formed teratomas. Next, we performed microarray analysis in CD34+38-CD43+lineage- hematopoietic progenitor cells (HPCs) re-induced from 6 Del20q-iPSC lines and 3 NK-T-iPSC lines. 315 genes were up-regulated (fold change >2) and 437 genes were down-regulated (fold change <0.5) in Del20q-iPSC-derived HPCs compared to isogenic NK-T-iPSC-derived HPCs. In particular, expression levels of 48 genes located on 20q11.2-13.1 had reduced expression by at least 2 fold (76 genes by 1.5 fold). Finally, we investigated the potential of hematopoietic differentiation in 9 Del20q-iPSC lines and 6 isogenic NK-T-iPSC lines. The efficiency of HPC production assessed by the OP9 co-culture system and the embryoid body differentiation culture system was comparable between Del20q-iPSC lines and NK-T-iPSC lines. However, colony forming capacity of iPSC-derived HPCs in methylcellulose culture and granulocyte and erythroid differentiation of iPSCs were severely impaired in all tested Del20q-iPSC lines (CFU-C numbers: 23±4 vs 114±16 per 2,500 HPCs, p< .001; CD66b+/CD11b+ cells: 3.9±1.2% vs 49.0±6.7%, p< .001; CD235a+ cells: 3.8±2.9% vs 32.2±5.2%, p< .001, in Del20q-iPSC lines vs NK-T-iPSC lines respectively). These results indicate that Del20q-iPSC lines are capable of exhibiting the identical feature of the MDS patient. This iPSC-based system could be useful for studying the precise molecular mechanisms of MDS and may also allow testing new therapeutic compounds under genetically defined conditions. Disclosures Yamanaka: iPS Academia Japan: Consultancy.


2021 ◽  
Vol 37 (2) ◽  
Author(s):  
Mehwish Zehravi ◽  
MOHSIN WAHID ◽  
Junaid Ashraf

Objective: To derive Duchenne muscular dystrophy patient specific induced pluripotent stem cells (iPSCs) from CD3+T cells of DMD patients using episomal reprogramming and characterization of these DMD-iPSCs using immunofluorescence to confirm their pluripotent state. Methods: DMD patients were selected based upon their clinical history and examination. Peripheral blood mononuclear cells were isolated from peripheral blood of DMD patients (n=3) by density gradient centrifugation and were used to generate DMD patient specific T cells (DMD-T cells) using rhIL-2, plate bound anti CD3 antibody and T cell specific media along with specific culture conditions that promote T cell expansion. CD3+ T cells were characterized by flow cytometry and reprogrammed using episomal plasmid to generate DMD-iPSCs. These DMD-iPSCs were characterized using immunofluorescence. The study was carried out at Dow Research Institute of Biotechnology and Biomedical Sciences during August 2017- July 2018 for a period of approximately 12 months. Results: The peripheral blood mononuclear cells (PBMNC) derived T cells appeared as suspended cell clumps morphologically were positive for the expression of CD3 and negative for CD31. The DMD patient specific iPSCs appeared as round, compact and flat colonies with well-defined edges morphologically and were positive for the expression of pluripotency markers OCT4, SSEA-4 and TRA-1-81 on immunofluorescence. Conclusion: CD3+ T cell derived DMD-iPSCs were obtained under feeder free and xeno-free culture conditions using episomal reprogramming. doi: https://doi.org/10.12669/pjms.37.2.3388 How to cite this:Zehravi M, Wahid M, Ashraf J. Episomal reprogramming of Duchenne muscular dystrophy patients derived CD3+ T cells towards induced pluripotent stem cells. Pak J Med Sci. 2021;37(2):---------. doi: https://doi.org/10.12669/pjms.37.2.3388 This is an Open Access article distributed under the terms of the Creative Commons Attribution License (http://creativecommons.org/licenses/by/3.0), which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited.


Sign in / Sign up

Export Citation Format

Share Document