Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 41
Filtrar
2.
Stem Cell Res ; 71: 103138, 2023 09.
Artigo em Inglês | MEDLINE | ID: mdl-37343428

RESUMO

Cell-based therapy offers great promise for treating degenerative diseases. Although autologous cell-based therapy is ideal, it may be impractical due to the high manufacturing cost and long production time. Allogeneic cell-based therapy offers a more cost-effective alternative; however, the risk of graft rejection is a major concern. Here, we generated HLA class-I and -II null iPSC line by knocking out CIITA gene in the B2M-knockout MUSIi001-A-1 cell line using CRISPR/Cas9 system. The MUSIi001-A-2 line provides a valuable model for studying immunological responses against allogeneic T cells and serves as a prototype for developing specific cell types for future cell-based therapy.


Assuntos
Células-Tronco Pluripotentes Induzidas , Humanos , Sistemas CRISPR-Cas/genética , Rejeição de Enxerto , Linhagem Celular
3.
Stem Cell Res ; 67: 103043, 2023 03.
Artigo em Inglês | MEDLINE | ID: mdl-36791635

RESUMO

Distal renal tubular acidosis (dRTA), a disease characterized by the failure of the distal nephron to secrete acid into the urine, can be caused by mutations in SLC4A1 gene encoding erythroid and kidney anion exchanger 1 (AE1). Here, an induced pluripotent stem cell (iPSC) line was generated from a patient with dRTA and hemolytic anemia carrying compound heterozygous SLC4A1 mutations containing c.1199_1225del (p.Ala400_Ala408del), resulting in Southeast Asian ovalocytosis (SAO), and c.1331C>A (p.Thr444Asn). Peripheral blood mononuclear cells (PBMCs) were reprogrammed using Sendai viral reprogramming. The established iPSC line, MUSIi019-A, exhibited pluripotent property and retained the same mutations observed in the patients.


Assuntos
Acidose Tubular Renal , Células-Tronco Pluripotentes Induzidas , Humanos , Proteína 1 de Troca de Ânion do Eritrócito/genética , Proteína 1 de Troca de Ânion do Eritrócito/metabolismo , Células-Tronco Pluripotentes Induzidas/metabolismo , Acidose Tubular Renal/genética , Leucócitos Mononucleares/metabolismo , Mutação
4.
Virus Res ; 323: 199009, 2023 Jan 02.
Artigo em Inglês | MEDLINE | ID: mdl-36414188

RESUMO

Influenza A virus (IAV) infection in pregnant women is a major public health concern. However, the effect of IAV infection on human embryogenesis is still unclear. Here we show that human induced pluripotent stem cells (hiPSCs) and hiPSC-derived ectodermal, mesodermal and endodermal cells are susceptible to IAV infection. These cell types stained positive for α2,6-linked sialic acid, the receptor for IAV infection expressed on the cell surface. While hiPSCs produced high viral titers for up to 7 days with increasing infected cell number suggesting that the viral progenies produced from hiPSCs without exogenous protease were infectious and could spread to other cells, the three germ-layer cells showed a decline in viral titers suggesting the lack of viral spreading. Amongst the three germ layers, endodermal cells were less susceptible than ectodermal and mesodermal cells. These results indicate the permissiveness of cells of early embryogenesis, and suggest a risk of detrimental effects of IAV infection in early human embryonic development.

5.
Int J Mol Sci ; 23(23)2022 Nov 30.
Artigo em Inglês | MEDLINE | ID: mdl-36499376

RESUMO

Persistent and efficient therapeutic protein expression in the specific target cell is a significant concern in gene therapy. The controllable integration site, suitable promoter, and proper codon usage influence the effectiveness of the therapeutic outcome. Previously, we developed a non-immunoglobulin scaffold, alpha repeat protein (αRep4E3), as an HIV-1 RNA packaging interference system in SupT1 cells using the lentiviral gene transfer. Although the success of anti-HIV-1 activity was evidenced, the integration site is uncontrollable and may not be practical for clinical translation. In this study, we use the CRISPR/Cas9 gene editing technology to precisely knock-in αRep4E3 genes into the adeno-associated virus integration site 1 (AAVS1) safe harbor locus of the target cells. We compare the αRep4E3 expression under the regulation of three different promoters, including cytomegalovirus (CMV), human elongation factor-1 alpha (EF1α), and ubiquitin C (UbC) promoters with and without codon optimization in HEK293T cells. The results demonstrated that the EF1α promoter with codon-optimized αRep4E3mCherry showed higher protein expression than other promoters with non-optimized codons. We then performed a proof-of-concept study by knocking in the αRep4E3mCherry gene at the AAVS1 locus of the Jurkat cells. The results showed that the αRep4E3mCherry-expressing Jurkat cells exhibited anti-HIV-1 activities against HIV-1NL4-3 strain as evidenced by decreased capsid (p24) protein levels and viral genome copies as compared to the untransfected Jurkat control cells. Altogether, our study demonstrates that the αRep4E3 could interfere with the viral RNA packaging and suggests that the αRep4E3 scaffold protein could be a promising anti-viral molecule that offers a functional cure for people living with HIV-1.


Assuntos
Sistemas CRISPR-Cas , HIV-1 , Humanos , Células Jurkat , Células HEK293 , HIV-1/genética , Edição de Genes , Replicação Viral/genética
6.
Sci Rep ; 12(1): 15551, 2022 09 16.
Artigo em Inglês | MEDLINE | ID: mdl-36114353

RESUMO

ß-thalassaemia is one of the most common genetic blood diseases worldwide with over 300 mutations in the HBB gene affecting red blood cell functions. Recently, advances in genome editing technology have provided a powerful tool for precise genetic correction. Generation of patient-derived induced pluripotent stem cells (iPSCs) followed by genetic correction of HBB mutations and differentiation into haematopoietic stem/progenitor cells (HSPCs) offers a potential therapy to cure the disease. However, the biggest challenge is to generate functional HSPCs that are capable of self-renewal and transplantable. In addition, functional analyses of iPSC-derived erythroid cells are hampered by poor erythroid expansion and incomplete erythroid differentiation. Previously, we generated an immortalised erythroid cell line (SiBBE) with unique properties, including unlimited expansion and the ability to differentiate into mature erythrocytes. In this study, we report a highly efficient genetic correction of HbE mutation in the SiBBE cells using the CRISPR/Cas9 system. The HbE-corrected clones restored ß-globin production with reduced levels of HbE upon erythroid differentiation. Our approach provides a sustainable supply of corrected erythroid cells and represents a valuable model for validating the therapeutic efficacy of gene editing systems.


Assuntos
Hemoglobina E , Talassemia beta , Sistemas CRISPR-Cas/genética , Linhagem Celular , Hemoglobina E/genética , Hemoglobina E/metabolismo , Humanos , Células-Tronco Pluripotentes Induzidas , Globinas beta/genética , Globinas beta/metabolismo , Talassemia beta/genética , Talassemia beta/metabolismo , Talassemia beta/terapia
7.
PLoS One ; 17(7): e0270963, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35797389

RESUMO

Genome editing in human induced pluripotent stem cells (hiPSCs) offers a potential tool for studying gene functions in disease models and correcting genetic mutations for cell-based therapy. Precise transgene insertion in hiPSCs represents a significant challenge. In the past decade, viral transduction has been widely used due to its high transduction efficiency; however, it can result in random transgene integration and variable transgene copy numbers. Non-viral-based strategies are generally safer but limited by their low transfection efficiency in hiPSCs. Recently, genome engineering using adeno-associated virus (AAV) vectors has emerged as a promising gene delivery approach due to AAVs' low immunogenicity, toxicity, and ability to infect a broad range of cells. The following protocol describes the workflow for genome editing in hiPSCs using the CRISPR/Cas9 ribonucleoprotein (RNP) complex combined with the recombinant AAV serotype 6 (AAV6) donor vectors to introduce a gene of interest (GOI) fused with mCherry fluorescent reporter gene into the AAVS1 safe harbor site. This approach leads to efficient transgene insertion and is applicable to precise genome editing of hiPSCs or other types of stem cells for research purposes.


Assuntos
Células-Tronco Pluripotentes Induzidas , Sistemas CRISPR-Cas/genética , Dependovirus/genética , Edição de Genes/métodos , Vetores Genéticos/genética , Humanos
8.
Stem Cell Res ; 63: 102871, 2022 08.
Artigo em Inglês | MEDLINE | ID: mdl-35853413

RESUMO

Two heterozygous mutations (p.L475P in ZYG11A and p.E51K in GATA6) were identified in a family with autosomal dominant diabetes. ZYG11A-p.L475P was proposed as a causative mutation because of the complete segregation with hyperglycemia and the proven pathogenic effect on beta-cell expansion. The modifying effect of GATA6-p.E51K was proposed owing to the earlier onset of the carriers. Herein, we establish a line of induced pluripotent stem cells (iPSCs) from peripheral blood mononuclear cells (PBMCs) of a proband who carries both mutations using Sendai viral vectors. The generated iPSC line was characterized for pluripotency, chromosomal normality, and authentication.


Assuntos
Diabetes Mellitus , Células-Tronco Pluripotentes Induzidas , Técnicas de Cultura de Células , Proteínas de Ciclo Celular/genética , Células Cultivadas , Diabetes Mellitus/metabolismo , Fator de Transcrição GATA6/genética , Fator de Transcrição GATA6/metabolismo , Vetores Genéticos , Humanos , Células-Tronco Pluripotentes Induzidas/metabolismo , Leucócitos Mononucleares/metabolismo , Mutação/genética
9.
PLoS One ; 17(3): e0264366, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35312698

RESUMO

Immunotherapy has emerged as a promising therapeutic approach for treating several forms of cancer. Adoptive cell transfer of immune cells, such as natural killer (NK) cells, provides a powerful therapeutic potential against tumor cells. In the past decades, two-dimensional (2D) tumor models have been used to investigate the effectiveness of immune cell killing. However, the 2D tumor models exhibit less structural complexity and cannot recapitulate the physiological condition of the tumor microenvironment. Thus, the effectiveness of immune cells against tumor cells using these models cannot fully be translated to clinical studies. In order to gain a deeper insight into immune cell-tumor interaction, more physiologically relevant in vivo-like three-dimensional (3D) tumor models have been developed. These 3D tumor models can mimic the dynamic cellular activities, making them a much closer representation of the in vivo tumor profiles. Here, we describe a simple and effective protocol to study the cytotoxic activity of primary human NK cells toward the 3D tumor spheroids. Our protocol includes isolation and expansion of human NK cells, labeling and formation of tumor spheroids, co-culture of NK cells and tumor spheroids, and evaluation of cytotoxic activity using a confocal microscope. This protocol is also applicable to other types of tumors and immune cells.


Assuntos
Neoplasias , Esferoides Celulares , Transferência Adotiva/métodos , Linhagem Celular Tumoral , Humanos , Imunoterapia , Células Matadoras Naturais , Neoplasias/terapia , Microambiente Tumoral
10.
Stem Cell Res ; 60: 102715, 2022 04.
Artigo em Inglês | MEDLINE | ID: mdl-35193007

RESUMO

A heterozygous mutation (c.T1424C: p.L475P) in ZYG11A completely segregating with hyperglycemia in a Thai family with familial diabetes of the adulthood has been reported as a cause of cell cycle arrest in 1.1B4 cell line. This mutation is a suggestive cause of failure in adaptive beta-cell expansion which, thereby, contributes to the development of diabetes in the family. Here, an induced pluripotent stem cell (iPSC) line from peripheral blood mononuclear cells (PBMCs) of an affected family member carrying the mutation was generated using Sendai viral reprogramming. The established iPSC line is characterized and confirmed for pluripotency and chromosomal integrity.


Assuntos
Diabetes Mellitus Tipo 2 , Células-Tronco Pluripotentes Induzidas , Adulto , Pontos de Checagem do Ciclo Celular , Proteínas de Ciclo Celular/genética , Diabetes Mellitus Tipo 2/metabolismo , Humanos , Células-Tronco Pluripotentes Induzidas/metabolismo , Leucócitos Mononucleares , Mutação/genética
11.
Int J Mol Med ; 49(3)2022 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-35119077

RESUMO

Breast cancer (BC) is the most common cancer in women. Although standard treatments are successful in patients with BC diagnosed at an early stage, an alternative treatment is required for patients with advanced­stage disease who do not respond to these treatments. The concept of using chemotherapy to sensitize cancer cells to become susceptible to immunotherapy was recently introduced and may be used as an alternative treatment for BC. The chemotherapeutic drug doxorubicin has been reported to sensitize cancer cells; however, the efficacy to sensitize the solid spheroids, in addition to its underlying mechanism regarding how doxorubicin sensitizes BC, has not previously been explored. In the present study, the effectiveness of a combined treatment of doxorubicin and natural killer­92 (NK­92) cells against BC in either 2D or 3D spheroid models, and its association with Fas receptor (FasR) expression, was demonstrated. The BC (MCF7) cell line expressing a higher level of FasR was more sensitive to NK­92 cell killing than the MDA­MB­231 cell line, which expressed a lower level of FasR. A sublethal dose of doxorubicin caused a significant improvement in NK cytotoxicity. Concordantly, a significant reduction in cell viability was observed in the doxorubicin­treated MCF7 spheroids. Notably, flow cytometric analysis revealed significantly increased FasR expression in the MCF7 cells, suggesting the underlying sensitization mechanism of doxorubicin in BC was related to the FasR upregulation. The present findings supported the use of combined doxorubicin and NK immunotherapy in BC treatment.


Assuntos
Neoplasias da Mama , Receptor fas , Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/metabolismo , Linhagem Celular Tumoral , Doxorrubicina/farmacologia , Doxorrubicina/uso terapêutico , Feminino , Humanos , Células Matadoras Naturais/metabolismo , Células MCF-7
12.
Int J Mol Sci ; 23(4)2022 Feb 19.
Artigo em Inglês | MEDLINE | ID: mdl-35216446

RESUMO

Human hematopoietic stem/progenitor cell (HSPC)-based gene therapy is a promising direction for curing HIV-1-infected individuals. The zinc finger protein (2LTRZFP) designed to target the 2-LTR-circle junction of HIV-1 cDNA was previously reported as an intracellular antiviral molecular scaffold that prevents HIV integration. Here, we elucidate the efficacy and safety of using 2LTRZFP in human CD34+ HSPCs. We transduced 2LTRZFP which has the mCherry tag (2LTRZFPmCherry) into human CD34+ HSPCs using a lentiviral vector. The 2LTRZFPmCherry-transduced HSPCs were subsequently differentiated into macrophages. The expression levels of pro-apoptotic proteins of the 2LTRZFPmCherry-transduced HSPCs showed no significant difference from those of the non-transduced control. Furthermore, the 2LTRZFPmCherry-transduced HSPCs were successfully differentiated into mature macrophages, which had normal phagocytic function. The cytokine secretion assay demonstrated that 2LTRZFPmCherry-transduced CD34+ derived macrophages promoted the polarization towards classically activated (M1) subtypes. More importantly, the 2LTRZFPmCherry transduced cells significantly exhibited resistance to HIV-1 integration in vitro. Our findings demonstrate that the 2LTRZFPmCherry-transduced macrophages were found to be functionally and phenotypically normal, with no adverse effects of the anti-HIV-1 scaffold. Our data suggest that the anti-HIV-1 integrase scaffold is a promising antiviral molecule that could be applied to human CD34+ HSPC-based gene therapy for AIDS patients.


Assuntos
Infecções por HIV/metabolismo , HIV-1/patogenicidade , Células-Tronco Hematopoéticas/metabolismo , Macrófagos/metabolismo , Células-Tronco/metabolismo , Dedos de Zinco/fisiologia , Antígenos CD34/metabolismo , Terapia Genética/métodos , Humanos
13.
Cell Tissue Res ; 388(1): 89-104, 2022 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-35072793

RESUMO

Peripheral arterial disease (PAD) is caused by atherosclerotic plaque accumulation, which results in ischemia in lower extremity ischemia. Cell-based therapy using endothelial progenitor cells (EPCs) or endothelial cells (ECs) has been challenging due to an insufficient number and replicative senescence of primary cells isolated from patients. To overcome this limitation, we generated induced pluripotent stem cells (iPSCs) from a patient with PAD for the first time. The patient-specific iPSCs have unlimited proliferation and can be used to generate a clinically relevant number of functional ECs. Here we developed a strategy to efficiently generate high EC yields within 5 days of differentiation. The generated iPSC-derived ECs from a PAD patient were phenotypically and functionally similar to the primary blood outgrowth endothelial cells (BOECs) and iPSC-ECs derived from healthy donors as evidenced by expression of EC-specific markers, capillary-like tube-forming potential, and the ability to uptake acetylated low-density lipoprotein (Ac-LDL). Our approach may provide an alternative renewable source of large-scale ECs for regenerative therapy. This study represents the first step toward the development of an autologous cell-based strategy for the treatment of PAD in the future.


Assuntos
Células-Tronco Pluripotentes Induzidas , Doença Arterial Periférica , Diferenciação Celular/fisiologia , Células Cultivadas , Células Endoteliais/metabolismo , Humanos , Isquemia/metabolismo , Isquemia/terapia , Doença Arterial Periférica/metabolismo , Doença Arterial Periférica/terapia
14.
Methods Mol Biol ; 2454: 607-624, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-33945142

RESUMO

Advances in induced pluripotent stem cell (iPSC) technology provide a renewable source of cells for tissue regeneration and therefore hold great promise for cell replacement therapy. However, immune rejection of allograft due to human leukocyte antigen (HLA) mismatching remains a major challenge. Considerable efforts have been devoted to overcoming the immunogenicity of allograft transplantation. One of the approaches is an elimination of HLA molecules on the surface of allogeneic cells using genome editing technology to generate universal stem cells. Here, we present a simple and effective genome editing approach to knockout the ß-2-immunoglobulin (B2M) gene, which encodes B2M protein that forms a heterodimer with HLA class I proteins, in induced pluripotent stem cells (iPSCs) leading to HLA class I (HLA-I) depletion. We also describe detailed procedures for validation of the B2M-knockout iPSCs using flow cytometry, and genotypic analysis for potential off-target regions. Our protocol is also applicable for knocking out other genes in iPSCs and other cell types.


Assuntos
Células-Tronco Pluripotentes Induzidas , Sistemas CRISPR-Cas , Edição de Genes , Antígenos HLA/genética , Antígenos de Histocompatibilidade Classe I/metabolismo , Humanos , Ribonucleoproteínas/metabolismo
15.
Methods Mol Biol ; 2454: 423-442, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-33755900

RESUMO

One of the major obstacles for adoptive cell transfer (ACT) of T cells is the loss of effector function and proliferative ability of isolated antigen-specific T cells after prolonged ex vivo expansion. To overcome this issue, induced pluripotent stem cells (iPSCs), which have unlimited proliferation and differentiation potential, can be used to generate a large number of antigen-specific T cells. Here, we describe an efficient differentiation protocol for the generation of cytotoxic CD8+ T cells from human T cell-derived iPSCs (T-iPSCs). The protocol consists of three main steps including differentiation of T-iPSCs toward hematoendothelial progenitors (HEPs), co-culture of HEPs with OP9-DL1 cells, and stimulation of T cell receptor (TCR) signaling to obtain CD8 single-positive (SP) T cells. This culture system is simple and efficient; therefore, will offer a powerful tool for studying T cell development and applications in adoptive immunotherapy.


Assuntos
Células-Tronco Pluripotentes Induzidas , Linfócitos T CD8-Positivos , Diferenciação Celular/fisiologia , Linhagem da Célula , Humanos , Imunoterapia Adotiva/métodos
16.
Front Immunol ; 12: 759558, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34650571

RESUMO

Adoptive cell therapy (ACT) using chimeric antigen receptor (CAR) T cells holds impressive clinical outcomes especially in patients who are refractory to other kinds of therapy. However, many challenges hinder its clinical applications. For example, patients who undergo chemotherapy usually have an insufficient number of autologous T cells due to lymphopenia. Long-term ex vivo expansion can result in T cell exhaustion, which reduces the effector function. There is also a batch-to-batch variation during the manufacturing process, making it difficult to standardize and validate the cell products. In addition, the process is labor-intensive and costly. Generation of universal off-the-shelf CAR T cells, which can be broadly given to any patient, prepared in advance and ready to use, would be ideal and more cost-effective. Human induced pluripotent stem cells (iPSCs) provide a renewable source of cells that can be genetically engineered and differentiated into immune cells with enhanced anti-tumor cytotoxicity. This review describes basic knowledge of T cell biology, applications in ACT, the use of iPSCs as a new source of T cells and current differentiation strategies used to generate T cells as well as recent advances in genome engineering to produce next-generation off-the-shelf T cells with improved effector functions. We also discuss challenges in the field and future perspectives toward the final universal off-the-shelf immunotherapeutic products.


Assuntos
Imunoterapia Adotiva , Células-Tronco Pluripotentes Induzidas/imunologia , Linfopenia/terapia , Receptores de Antígenos Quiméricos/imunologia , Linfócitos T/imunologia , Humanos , Células-Tronco Pluripotentes Induzidas/citologia , Linfopenia/imunologia , Linfócitos T/citologia
17.
Stem Cell Res ; 56: 102551, 2021 10.
Artigo em Inglês | MEDLINE | ID: mdl-34628247

RESUMO

Allogeneic cell-based therapy is emerging as a promising approach in regenerative medicine. However, rejection of allograft due to mismatch of human leukocyte antigens (HLAs) remains a major concern after transplantation. Here, we generated a homozygous B2M knockout induced pluripotent stem cell (iPSC) line, lacking the expression of HLA class I (HLA-I) molecules, using a CRISPR/Cas9 system. The established iPSC line, MUSIi001-A-1, can serve as an in vitro model for studying immunological responses against allogeneic grafts and provides a prototype for "off-the-shelf" allogeneic cell products for future cell-based therapy.


Assuntos
Células-Tronco Pluripotentes Induzidas , Microglobulina beta-2 , Sistemas CRISPR-Cas/genética , Antígenos HLA , Humanos , Medicina Regenerativa , Transplante Homólogo , Microglobulina beta-2/genética
18.
Methods Mol Biol ; 2211: 193-211, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33336279

RESUMO

HbE/ß-thalassemia is one of the most common thalassemic syndromes in Southeast Asia and Thailand. Patients have mutations in ß hemoglobin (HBB) gene resulting in decreased and/or abnormal production of ß hemoglobin. Here, we describe a protocol for CRISPR/Cas9-mediated gene correction of the mutated hemoglobin E from one allele of the HBB gene by homology-directed repair (HDR) in HbE/ß-thalassemia patient-derived induced pluripotent stem cells (iPSCs) using a CRISPR/Cas9 plasmid-based transfection method and a single-stranded DNA oligonucleotide (ssODN) repair template harboring the correct nucleotides. Our strategy allows the seamless HbE gene correction with the editing efficiency (HDR) up to 3%, as confirmed by Sanger sequencing. This protocol provides a simple one-step genetic correction of HbE mutation in the patient-derived iPSCs. Further differentiation of the corrected iPSCs into hematopoietic stem/progenitor cells will provide an alternative renewable source of cells for the application in autologous transplantation in the future.


Assuntos
Sistemas CRISPR-Cas , Edição de Genes , Hemoglobina E/genética , Células-Tronco Pluripotentes Induzidas/metabolismo , Talassemia beta/genética , Diferenciação Celular/genética , Clonagem Molecular , Edição de Genes/métodos , Expressão Gênica , Marcação de Genes , Vetores Genéticos/genética , Células-Tronco Hematopoéticas/citologia , Células-Tronco Hematopoéticas/metabolismo , Humanos , Células-Tronco Pluripotentes Induzidas/citologia , Mutação , RNA Guia de Cinetoplastídeos/genética , Reparo de DNA por Recombinação , Globinas beta/genética
19.
Methods Mol Biol ; 2211: 213-229, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33336280

RESUMO

Cell-mediated cytotoxicity plays an important role in several fundamental immunological processes and is crucial for biological evaluation in in vitro studies. In order to determine the immunological activities of the cells, an assay should be safe, reproducible, and cost-effective. Here, we present a simple and cost-effective approach for evaluation of natural killer (NK) cell-mediated cytotoxicity by generating a CRISPR/Cas9-mediated GFP reporter knock-in in the target cell line, K562, and the non-target cell line, Raji, using a plasmid-based transfection method. The GFP+ target cells facilitate tracking of the immune cell killing assay, which avoids the need for multiple cell labeling with fluorescent dyes. Our approach is also applicable to the genome editing of other target cell types for functional analysis of effector cells.


Assuntos
Sistemas CRISPR-Cas , Citotoxicidade Imunológica/genética , Edição de Genes , Genes Reporter , Linhagem Celular Tumoral , Evolução Clonal/genética , Citometria de Fluxo , Expressão Gênica , Técnicas de Introdução de Genes , Proteínas de Fluorescência Verde/genética , Humanos , Células K562 , Plasmídeos/genética
20.
Stem Cell Res Ther ; 11(1): 481, 2020 11 11.
Artigo em Inglês | MEDLINE | ID: mdl-33176890

RESUMO

BACKGROUND: Human induced pluripotent stem cells (hiPSCs) offer a renewable source of cells for the generation of hematopoietic cells for cell-based therapy, disease modeling, and drug screening. However, current serum/feeder-free differentiation protocols rely on the use of various cytokines, which makes the process very costly or the generation of embryoid bodies (EBs), which are labor-intensive and can cause heterogeneity during differentiation. Here, we report a simple feeder and serum-free monolayer protocol for efficient generation of iPSC-derived multipotent hematoendothelial progenitors (HEPs), which can further differentiate into endothelial and hematopoietic cells including erythroid and T lineages. METHODS: Formation of HEPs from iPSCs was initiated by inhibition of GSK3 signaling for 2 days followed by the addition of VEGF and FGF2 for 3 days. The HEPs were further induced toward mature endothelial cells (ECs) in an angiogenic condition and toward T cells by co-culturing with OP9-DL1 feeder cells. Endothelial-to-hematopoietic transition (EHT) of the HEPs was further promoted by supplementation with the TGF-ß signaling inhibitor. Erythroid differentiation was performed by culturing the hematopoietic stem/progenitor cells (HSPCs) in a three-stage erythroid liquid culture system. RESULTS: Our protocol significantly enhanced the number of KDR+ CD34+ CD31+ HEPs on day 5 of differentiation. Further culture of HEPs in angiogenic conditions promoted the formation of mature ECs, which expressed CD34, CD31, CD144, vWF, and ICAM-1, and could exhibit the formation of vascular-like network and acetylated low-density lipoprotein (Ac-LDL) uptake. In addition, the HEPs were differentiated into CD8+ T lymphocytes, which could be expanded up to 34-fold upon TCR stimulation. Inhibition of TGF-ß signaling at the HEP stage promoted EHT and yielded a large number of HSPCs expressing CD34 and CD43. Upon erythroid differentiation, these HSPCs were expanded up to 40-fold and displayed morphological changes following stages of erythroid development. CONCLUSION: This protocol offers an efficient and simple approach for the generation of multipotent HEPs and could be adapted to generate desired blood cells in large numbers for applications in basic research including developmental study, disease modeling, and drug screening as well as in regenerative medicine.


Assuntos
Células-Tronco Pluripotentes Induzidas , Diferenciação Celular , Células Endoteliais , Quinase 3 da Glicogênio Sintase , Células-Tronco Hematopoéticas , Humanos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA