Project description:Assessing transcriptomic differences between activation and non-activation of CAR T cells expressing different Chimeric Cytokine Receptors
Project description:Failure of adoptive T cell therapies in cancer patients is linked to limited T cell expansion and persistence, even in memory-prone 41BB-(BBz)-based chimeric antigen receptor (CAR) T cells. In murine CD8+ T cells, SUV39H1 promotes differentiation and expansion of effector CD8+ T cells during acute infection by Listeria monocytogenes by silencing stemness and memory genes (Pace et al. Science, 2018). The purpuse of this study is to investigate the transcriptomic differences of SUV39H1 knock-out versus mock human 41BBz-CAR T cells by Nanostring at different cycles of restimulation.
Project description:This dataset is a four-ligand x three-genotype Affymetrix microarray analysis of the regulation of liver genes in the mouse by the constitutive androstane receptor (CAR). 24 female mice of mixed background (C57BL/6x129Sv) were divided into three groups: wild-type (contains only mouse CAR; mCAR), CAR.KO = knockout mice (mice ablated for mCAR gene; mCAR -/-), and CAR.AH= contains human CAR transgene under the control of the mouse albumin promoter in the mCAR -/- background. Each of the three groups underwent four different treatment regimens: CO = corn oil vehicle control, PB = phenobarbitol (100 mg/kg/day), an anti-convulsant agent which can translocate both mCAR and hCAR into the nucleus to turn on target gene expression, TC = TCPOBOP, a potent non-metabolized ligand of mCAR (3 mg/kg), CITCO = a hCAR specific ligand (30 mg/kg/day). Two mice were used per treatment group and each mouse RNA was used for one chip.
Project description:In mouse bone marrow, mesenchymal stem cells (MSC) has the potential to form osteocytes, adipocytes and cartilage. In the process of osteogenesis, MSCs differenetiate into stromal cells, such as CAR cells. Osteoblast is responsible for the formation of osteocytes and osteoblasts may be differentiated from a subset of CAR cells. Dmp1-Cre targeted CAR cells are thought to enrich for a osteoblast progenitor population. We used microarrays to detail the gene expression profiles among Dmp1-Cre targeted and non-targeted CAR cells. Gene expression diffferences were compared to support the hypothesis that Dmp1-Cre targeted CAR cells may be enriched for osteoblast progenitors. Dmp1-Cre targeted and non-targeted CAR cells were FACS sorted from three mice. RNA were extracted from these sorted cells and processed for microarray using Affymetrix mogene 1.0 ST chip. Cells from one mouse represent one sample
Project description:Comparison of gene expression by Nanostring of activated and non activated CAR T cells expressing a CD19 CAR in the absence or presence of the dSHP2 module
Project description:The objective of this experiment was to characterized the molecular mechanisms behind the different effects of PD-1 disruption in low affinity (LA) and high affinity (HA) HER2-28z CAR-T cells. The transcriptomic profile of mock and PD-1 KO CAR-T cells following antigen recognition was compared by using the nCounter® CAR-T Characterization Gene Expression Panel (Nanostring). RNA samples were isolated from CAR-T cells generated from three independent donors.
Project description:CAR-T cell therapy against MM currently shows promising results, but usually with serious toxicities. CAR-NK cells may exert less toxicity when redirected against resistant myeloma cells. CARs can be designed through the use of receptors, such as NKG2D, which recognizes a wide range of ligands to provide broad target specificity. Here, we test this approach by analyzing the anti-tumor activity of activated and expanded NK cells (NKAE) and CD45RA- T cells from MM patients that were engineered to express an NKG2D-based CAR. NKAE cells were cultured with irradiated Clone9.mbIL21 cells. Then, cells were transduced with an NKG2D-4-1BB-CD3z-CAR. CAR-NKAE cells exhibited no evidence of genetic abnormalities. Although memory T cells were more stably transduced, CAR-NKAE cells exhibited greater in vitro cytotoxicity against MM cells, while showing minimal activity against healthy cells. In vivo, CAR-NKAE cells mediated highly efficient abrogation of MM growth, and two of the treated mice remained disease-free. Overall, these results demonstrate that it is feasible to modify autologous NKAE cells from MM patients to safely express a NKG2D-CAR. Additionally, autologous CAR-NKAE cells display enhanced anti-myeloma activity demonstrating that they could be an effective strategy against MM supporting the development of NKG2D-CAR NK cell therapy for MM.
Project description:Purpose: Compared the differences in the transcriptome of T cells versus EpCAM CAR-T cells and EpCAM CAR-T cells versus rapamycin-pretreated EpCAM CAR-T cells.Methods: Next-generation sequencing (NGS) has revolutionized the systems-based analysis of cellular pathways. The goals of this study are to compare NGS-derived transcriptome profiling (RNA-seq) between T cells and EpCAM CAR-T cells,and between EpCAM CAR-T cells and rapamycin-pretreated EpCAM CAR-T cells. Results: RNA-seq showed that CAR-T cells significantly upregulated the expression of exhaustion markers compared to T cells. Gene Set Enrichment Analysis (GSEA) analysis showed that CAR-T cells cells were enriched for signatures of apoptosis. Compared with T cells, Kyoto Encyclopedia of Genes and Genomessignaling(KEGG) pathway analysis showed that the PI3K/AKT/mTOR signaling pathway was significantly enriched. Likewise, GSEA showed that the mTOR signaling pathway was significantly upregulated in CAR-T cells. Next, we analyzed the RNA-seq data between CAR-T cells and rapamycin-pretreated CAR-T cells. RNA-seq showed that rapamycin-treated CAR T down-regulated the expression of exhaustion markers compared with untreated CAR-T cells. GSEA analysis showed that the apoptosis-related gene set was enriched in untreated CAR-T cells.Conclusions:In vitro culture promotes the terminal differentiation of CAR-T cells.CAR-T cells present a highly activated mTOR phenotype. Rapamycin pretreatment prevents CAR-T cells terminal differentiation.