ImmunoCult™ Human CD3/CD28 T Cell Activator

Human T cell activation and expansion reagent

ImmunoCult™ Human CD3/CD28 T Cell Activator

Human T cell activation and expansion reagent

From: 202 USD
Catalog #
(Select a product)
Human T cell activation and expansion reagent
Add to Wish List

Product Advantages


  • Robust activation and expansion of human T cells without the use of magnetic beads, feeder cells, or antigen

  • Provides a gentle activation stimulus that maintains high viability of activated and expanded T cells

  • Highly stable, filter-sterilized soluble reagent

What Our Scientist Says

We want to make it easier to activate and expand human T cells while still maintaining a high viability of these cells. That's why we developed ImmunoCult™ Human CD3/CD28 T Cell Activator.

Jessie YuScientist
Jessie Yu, Scientist

Overview

Achieve robust activation and expansion of T cells in the absence of magnetic beads, feeder cells, or antigens.

This product’s gentle activation stimulus ensures a high viability of activated T cells, which can be further expanded in ImmunoCult™-XF T Cell Expansion Medium (Catalog #10981) or other media for culturing human T cells. Antibody complexes bind to and cross-link CD3 and CD28 cell surface ligands, thereby providing the required signals for T cell activation. 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 research applications. If you require reagents suitable for use in cell therapy manufacturing, ImmunoCult™ Human CD3/CD28 T Cell Activators (Catalog #100-0784) are produced under relevant GMPs.
Contains
• Anti-human CD3 monospecific antibody complex
• Anti-human CD28 monospecific antibody complex
Subtype
Supplements
Cell Type
T Cells, T Cells, CD4+, T Cells, CD8+
Species
Human
Application
Activation, Cell Culture, Expansion
Brand
ImmunoCult
Area of Interest
Immunology, Cell Therapy Development

Data Figures

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).

Protocols and Documentation

Find supporting information and directions for use in the Product Information Sheet or explore additional protocols below.

Document Type
Product Name
Catalog #
Lot #
Language
Catalog #
10971, 10991
Lot #
All
Language
English
Document Type
Safety Data Sheet
Catalog #
10971, 10991
Lot #
All
Language
English

Applications

This product is designed for use in the following research area(s) as part of the highlighted workflow stage(s). Explore these workflows to learn more about the other products we offer to support each research area.

Resources and Publications

Educational Materials (21)

Publications (42)

Discovery of novel disulfide-containing PD-1/PD-L1 inhibitor with in vivo influenza therapeutic efficacy Y. Hirata et al. Scientific Reports 2025 Sep

Abstract

Monoclonal antibody-based immune checkpoint inhibitors, which have brought breakthrough effects in cancer treatments, are expected to assist in the treatment of viral diseases. However, antibody therapies may cause immune-related side effects, such as inflammation and pneumonia, due to cytokine storms. Small-molecule PD-1/PD-L1 inhibitors are an alternative to monoclonal antibody-based therapeutics. We have identified a novel small-molecule PD-1/PD-L1 inhibitor having a functional group (disulfide group), namely compound 2 (molecular weight: 456.6), from our library of sulfur-containing protein–protein interaction inhibitor compounds. Compound 2 selectively bound to PD-L1 over PD-1, with the dissociation rate constant (K D ) of 77.60 ± 4.44 nM (obtained by affinity analysis) and showed promising T cell activation recovery. A molecular docking simulation study between 2 and PD-L1 suggested that 2 binds to PD-L1 in a binding mode different from those of other small-molecule PD-L1/PD-1 inhibitors. Notably, oral administration of 2 to mice pre-infected with influenza A virus (A/NWS/33, H1N1 subtype) caused a significant increase in the neutralizing antibody titers, as well as recovery from influenza-induced pneumonia. Overall, 2 provides insight for the development of therapeutic drugs against early viral infections, with both virus titer-reducing and antibody titer-boosting effects. Moreover, 2 is widely used as a rubber peptizing agent in the production process of tires and other rubber products. Our findings may provide useful information for investigating its influence on living organisms. The online version contains supplementary material available at 10.1038/s41598-025-17982-3. Subject terms: Drug discovery and development, Pharmacology, Screening, Structure-based drug design
Crosstalk with infant-derived Th17 cells, as well as exposure to IL-22 promotes maturation of intestinal epithelial cells in an enteroid model Z. Sharafian et al. Frontiers in Immunology 2025 May

Abstract

The intestinal epithelium of human infants is developmentally immature compared to that of adults. Exactly how this immaturity affects key epithelial functions and their interactions with nearby immune cells remains an understudied area of research, partly due to limited access to non-diseased infant gut tissues. Human intestinal organoids, or “mini guts” generated from tissue stem cells, are promising models for investigating intestinal biology and disease mechanisms. These three-dimensional structures closely mimic their tissue of origin, including cellular physiology and genetics. We have also previously shown that neonatal Th17 cells represent a distinct cell population with a cytokine profile skewed toward IL-22 production rather than IL-17A, as seen in adult Th17 cells. In this study, we sought to model the impact of neonatal-derived Th17 cytokine, namely IL-22 and the intestinal epithelium using infant-derived ileal enteroids. We generated enteroids from ileal biopsies from infants (< 6 months old) and cultured them for seven days with standard organoid growth media, organoid media supplemented with conditioned media from cord-blood-derived Th17 cells, or media supplemented with recombinant IL-22. We assessed morphological changes and conducted transcriptomics profiling via RNAseq. Exposing enteroids to neonatal Th17-cells-derived conditioned media led to enhanced growth, maturation, and differentiation as compared to control media. These effects were ablated when an IL-22 neutralizing antibody was used, while conversely, supplementing with recombinant IL-22 mimicked the Th17 effects, increasing intestinal epithelial cell proliferation and inducing marked differentiation of secretory cells. Our transcriptomic profiling similarly demonstrated significant changes in response to IL-22 with downregulation of Wnt and Notch signaling and upregulation of immune pathways, particularly interferon signaling. The transcriptomic data also suggested that IL-22 treatment led to changes in cell type composition with an increase in stem- and progenitor cells at the expense of enterocytes. Taken together, our data suggests that early-life intestinal development is likely influenced by IL-22-dependent crosstalk between the infant epithelium and exposure to neighboring Th17 cells. This promotes epithelial cell maturation and immune readiness, reflected at both the morphological and molecular levels. Our work also provides a relevant framework for studying healthy infant gut development, which can be further leveraged to examine early-life gastrointestinal disorders, model complex human disease, and therapeutic testing while reducing reliance on animal models.
Cell trajectory modulation: rapid microfluidic biophysical profiling of CAR T cell functional phenotypes Nature Communications 2025 May

Abstract

Chimeric Antigen Receptor (CAR) T cell therapy is a pivotal treatment for hematological malignancies. However, CAR T cell products exhibit batch-to-batch variability in cell number, quality, and in vivo efficacy due to donor-to-donor heterogeneity, and pre/post-manufacturing processes, and the manufacturing of such products necessitates careful testing, both post-manufacturing and pre-infusion. Here, we introduce the Cell Trajectory Modulation (CTM) assay, a microfluidic, label-free approach for the rapid evaluation of the functional attributes of CAR T cells based on biophysical features (i.e., size, deformability). CTM assay correlates with phenotypic metrics, including CD4:CD8 ratio, memory subtypes, and cytotoxic activity. Validated across multiple donors and culture platforms, the CTM assay requires fewer than 10,000 cells and delivers results within 10 minutes. Compared to labeled flow cytometry processing, the CTM assay offers real-time data to guide adaptive manufacturing workflows. Thus, the CTM assay offers an improvement over existing phenotypic assessments, marking a step forward in advancing CAR T cell therapy manufacturing. CAR T cell manufacturing faces significant challenges that impact cell quality and in vivo efficacy. This necessitates reliable cellular characterization methods. Here the authors present a real-time, label-free, microfluidic method that profiles cellular biophysical properties and correlates them to activation state and CAR T potency, facilitating the rapid phenotypic cell assessment during production.