当前位置:首页 > 产品中心 > 细胞培养基及添加物

细胞培养基及添加物

ImmunoCult™ Human CD3/CD28 T Cell Activator

Human T cell activation and expansion reagent

概要
技术资料
数据及文献

概要

ImmunoCult™ Human CD3/CD28 T Cell Activator is designed to activate and expand human T cells in the absence of magnetic beads, feeder cells, or antigen. ImmunoCult™ Human CD3/CD28 T Cell Activator consists of soluble tetrameric antibody complexes that bind CD3 and CD28 cell surface ligands. Binding of the tetrameric antibody complexes results in the cross-linking of CD3 and CD28 cell surface ligands, thereby providing the required primary and co-stimulatory signals for T cell activation. Activated T cells can be expanded in ImmunoCult™-XF T Cell Expansion Medium or other media for culturing human T cells supplemented with cytokines.

The ImmunoCult™ Human CD3/CD28 T Cell Activator can be used on the Seahorse XF Analyzer to measure T cell activation response and is also available as part of the Agilent Seahorse XF Hu T Cell Activation Assay Kit.

This product is designed for cell therapy research applications following the recommendations of USP<1043> on Ancillary Materials, and we can currently work with you to qualify this reagent under an approved investigational new drug (IND) or clinical trial application (CTA).

技术资料

Document Type 产品名称 Catalog # Lot # 语言
Product Information Sheet ImmunoCult™ Human CD3/CD28 T Cell Activator 10971, 10991 All English
Safety Data Sheet ImmunoCult™ Human CD3/CD28 T Cell Activator 10971, 10991 All English

数据及文献

Data

Activated Morphology of Human T Cells Stimulated With ImmunoCult™ Human CD3/CD28 T Cell Activator

Figure 1. Activated Morphology of Human T Cells Stimulated With ImmunoCult™ Human CD3/CD28 T Cell Activator

Image of human T cells isolated using the EasySep™ Human T Cell Isolation Kit (Catalog #17951), stimulated with ImmunoCult™ Human CD3/CD28 T Cell Activator, and cultured in ImmunoCult™-XF T Cell Expansion Medium (Catalog # 10981).

Activation of Human T Cells stimulated With ImmunoCult™ Human CD3/CD28 T Cell Activator

Figure 2. Activation of EasySep™-isolated Human T Cells stimulated With ImmunoCult™ Human CD3/CD28 T Cell Activator

EasySep™-isolated human T cells were stimulated with ImmunoCult™ Human CD3/CD28 T Cell Activator and cultured in ImmunoCult™-XF T Cell Expansion Medium. Activation of viable CD3+ T cells was assessed by CD25 expression using flow cytometry. On day 0, the frequency of CD25 positive cells was (A) 5.6 ± 2.4% (mean ± SD). Following 3 days of culture, the frequency of CD25 positive cells was (B) 75.4 ± 13.8% (mean ± SD) when stimulated with ImmunoCult™ Human CD3/CD28 T Cell Activator.

Robust Human T Cell Expansion with ImmunoCult™ Human CD3/CD28 T Cell Activator

Figure 3. Robust Human T Cell Expansion with ImmunoCult™ Human CD3/CD28 T Cell Activator

EasySep™-isolated human T cells were expanded over 12 days with ImmunoCult™ Human CD3/CD28 T Cell Activator in ImmunoCult™-XF T Cell Expansion Medium supplemented with Human Recombinant IL-2. On day 0, 1 x 10^6 EasySep™-isolated human T cells were stimulated with 25 μL of ImmunoCult™ Human CD3/CD28 T Cell Activator in ImmunoCult™-XF T Cell Expansion Medium supplemented with 10 ng/mL Human Recombinant IL-2. On days 3, 5, 7, and 10, viable cells were counted and fresh medium supplemented with IL-2 was added. No additional ImmunoCult™ Human CD3/CD28 T Cell Activator was added during the 12-day culture period (mean ± SD in 6 experiments with 3 donors).

Publications (6)

Science advances 2020 may Competition between PAF1 and MLL1/COMPASS confers the opposing function of LEDGF/p75 in HIV latency and proviral reactivation. R. Gao et al.

Abstract

Transcriptional status determines the HIV replicative state in infected patients. However, the transcriptional mechanisms for proviral replication control remain unclear. In this study, we show that, apart from its function in HIV integration, LEDGF/p75 differentially regulates HIV transcription in latency and proviral reactivation. During latency, LEDGF/p75 suppresses proviral transcription via promoter-proximal pausing of RNA polymerase II (Pol II) by recruiting PAF1 complex to the provirus. Following latency reversal, MLL1 complex competitively displaces PAF1 from the provirus through casein kinase II (CKII)-dependent association with LEDGF/p75. Depleting or pharmacologically inhibiting CKII prevents PAF1 dissociation and abrogates the recruitment of both MLL1 and Super Elongation Complex (SEC) to the provirus, thereby impairing transcriptional reactivation for latency reversal. These findings, therefore, provide a mechanistic understanding of how LEDGF/p75 coordinates its distinct regulatory functions at different stages of the post-integrated HIV life cycles. Targeting these mechanisms may have a therapeutic potential to eradicate HIV infection.
Cell systems 2020 mar Gut-Liver Physiomimetics Reveal Paradoxical Modulation of IBD-Related Inflammation by Short-Chain Fatty Acids. M. Trapecar et al.

Abstract

Although the association between the microbiome and IBD and liver diseases is known, the cause and effect remain elusive. By connecting human microphysiological systems of the gut, liver, and circulating Treg and Th17 cells, we created a multi-organ model of ulcerative colitis (UC) ex vivo. The approach shows microbiome-derived short-chain fatty acids (SCFAs) to either improve or worsen UC severity, depending on the involvement of effector CD4 T cells. Using multiomics, we found SCFAs increased production of ketone bodies, glycolysis, and lipogenesis, while markedly reducing innate immune activation of the UC gut. However, during acute T cell-mediated inflammation, SCFAs exacerbated CD4+ T cell-effector function, partially through metabolic reprograming, leading to gut barrier disruption and hepatic injury. These paradoxical findings underscore the emerging utility of human physiomimetic technology in combination with systems immunology to study causality and the fundamental entanglement of immunity, metabolism, and tissue homeostasis.
Scientific reports 2020 jun A Simple and Scalable Strategy for Analysis of Endogenous Protein Dynamics. M. K. Schwinn et al.

Abstract

The ability to analyze protein function in a native context is central to understanding cellular physiology. This study explores whether tagging endogenous proteins with a reporter is a scalable strategy for generating cell models that accurately quantitate protein dynamics. Specifically, it investigates whether CRISPR-mediated integration of the HiBiT luminescent peptide tag can easily be accomplished on a large-scale and whether integrated reporter faithfully represents target biology. For this purpose, a large set of proteins representing diverse structures and functions, some of which are known or potential drug targets, were targeted for tagging with HiBiT in multiple cell lines. Successful insertion was detected for 86{\%} of the targets, as determined by luminescence-based plate assays, blotting, and imaging. In order to determine whether endogenously tagged proteins yield more representative models, cells expressing HiBiT protein fusions either from endogenous loci or plasmids were directly compared in functional assays. In the tested cases, only the edited lines were capable of accurately reproducing the anticipated biology. This study provides evidence that cell lines expressing HiBiT fusions from endogenous loci can be rapidly generated for many different proteins and that these cellular models provide insight into protein function that may be unobtainable using overexpression-based approaches.
Stem Cell Research 2019 oct Detection of all adult Tau isoforms in a 3D culture model of iPSC-derived neurons L. Miguel et al.

Abstract

Tauopathies are a class of neurodegenerative diseases characterized by the presence of pathological intracellular deposits of Tau proteins. Six isoforms of Tau are expressed in the adult human brain, resulting from alternative splicing of the MAPT gene. Tau splicing is developmentally regulated such that only the smallest Tau isoform is expressed in fetal brain, contrary to the adult brain showing the expression of all 6 isoforms. Induced Pluripotent Stem Cell (iPSC) technology has opened up new perspectives in human disease modeling, including tauopathies. However, a major challenge to in vitro recapitulation of Tau pathology in iPSC-derived neurons is their relative immaturity. In this study, we examined the switch in Tau splicing from fetal-only to all adult Tau isoforms during the differentiation of iPSC-derived neurons in a new 3D culture system. First, we showed that iPSC-induced neurons inside Matrigel-coated alginate capsules were able to differentiate into cortical neurons. Then, using a new assay that allowed both the qualitative and the quantitative analysis of all adult MAPT mRNA isoforms individually, we demonstrated that BrainPhys-maintained neurons expressed the 6 adult MAPT mRNA transcripts from 25 weeks of maturation, making this model highly suitable for modeling Tau pathology and therapeutic purposes.
Nature medicine 2018 OCT Translational control of tumor immune escape via the eIF4F-STAT1-PD-L1 axis in melanoma. M. Cerezo et al.

Abstract

Preventing the immune escape of tumor cells by blocking inhibitory checkpoints, such as the interaction between programmed death ligand-1 (PD-L1) and programmed death-1 (PD-1) receptor, is a powerful anticancer approach. However, many patients do not respond to checkpoint blockade. Tumor PD-L1 expression is a potential efficacy biomarker, but the complex mechanisms underlying its regulation are not completely understood. Here, we show that the eukaryotic translation initiation complex, eIF4F, which binds the 5' cap of mRNAs, regulates the surface expression of interferon-$\gamma$-induced PD-L1 on cancer cells by regulating translation of the mRNA encoding the signal transducer and activator of transcription 1 (STAT1) transcription factor. eIF4F complex formation correlates with response to immunotherapy in human melanoma. Pharmacological inhibition of eIF4A, the RNA helicase component of eIF4F, elicits powerful antitumor immune-mediated effects via PD-L1 downregulation. Thus, eIF4A inhibitors, in development as anticancer drugs, may also act as cancer immunotherapies.
Cancer cell 2018 FEB Eradication of Triple-Negative Breast Cancer Cells by Targeting Glycosylated PD-L1. C.-W. Li et al.

Abstract

Protein glycosylation provides proteomic diversity in regulating protein localization, stability, and activity; it remains largely unknown whether the sugar moiety contributes to immunosuppression. In the study of immune receptor glycosylation, we showed that EGF induces programmed death ligand 1 (PD-L1) and receptor programmed cell death protein 1 (PD-1) interaction, requiring beta$-1,3-N-acetylglucosaminyl transferase (B3GNT3) expression in triple-negative breast cancer. Downregulation of B3GNT3 enhances cytotoxic T cell-mediated anti-tumor immunity. A monoclonal antibody targeting glycosylated PD-L1 (gPD-L1) blocks PD-L1/PD-1 interaction and promotes PD-L1 internalization and degradation. In addition to immune reactivation, drug-conjugated gPD-L1 antibody induces a potent cell-killing effect as well as a bystander-killing effect on adjacent cancer cells lacking PD-L1 expression without any detectable toxicity. Our work suggests targeting protein glycosylation as a potential strategy to enhance immune checkpoint therapy.
View All Publications