Product Citations: 43

A recombinant BCG with surface-displayed antigen induces humoral and cellular immune responses.

In Scientific Reports on 16 May 2025 by Zhang, J. Y., Hu, Z. D., et al.

Bacillus Calmette-Guérin (BCG) is an attenuated vaccine widely used for tuberculosis prevention. While BCG has long been perceived as an intracellular candidate vector for delivering antigens against infectious diseases and cancers, challenges persist in inducing durable immune responses, particularly high-titer neutralizing antibodies (Nabs). Here we show that displaying antigens in the surface of BCG is a promising strategy to induce long-lasting Nabs production and T-cell responses. We constructed a recombinant BCG expressing the SARS-CoV-2 receptor-binding domain (RBD) antigen on its cell wall, termed CW-rBCG::RBD, which achieved an antigen yield approaching 850 nanograms per 107 colony-forming unit. Compared with both the parental BCG and the RBD protein subunit vaccine (RBDAS01), intravenous administration of CW-rBCG::RBD followed by a booster dose significantly enhanced Nab production and increased the frequencies of RBD-specific central memory T cells (Tcm) and T follicular helper (Tfh) cells in the spleen. In mice primed with a single dose of CW-rBCG::RBD and boosted with RBDAS01, we also observed elevated Nab titers and detectable levels of RBD-specific IgG2a antibodies at 8 weeks post-priming, responses that were not observed in the BCG-primed or RBDAS01-only groups. Furthermore, subcutaneous co-administration of CW-rBCG::RBD and RBDAS01 sustained Nab production for up to 31 weeks and maintained higher Tfh and Tcm cell frequencies compared to both BCG co-administration with RBDAS01 and RBDAS01 alone. These findings highlight an effective strategy for optimizing BCG-based vaccination and immunotherapy platforms. Subject terms: recombinant BCG; immune response; vaccines; cell wall; SARS-CoV-2 RBD.
© 2025. The Author(s).

  • Immunology and Microbiology

Dendritic cell (DC)-derived extracellular vesicles (DEVs) are promising candidates for cancer vaccines, but their therapeutic effects still need further optimization. In this study, we utilized neoantigens, lipopolysaccharide and IFN-γ to induce the maturation of DCs, and then isolated DEVs derived from these mature DCs. We showed that the immune checkpoint inhibitor (anti-CTLA-4 antibody, aCTLA-4) can improve the immunostimulatory function of DEVs by directly activating T cells through immune checkpoint signal blockade. The cytokine interleukin-12 (IL-12), as one of the third signals for T cell activation, can also enhance the capability of DEVs to activate T cells directly. Based on these findings, we designed the engineered DEVs conjugated with IL-12 and aCTLA-4 (DEV@IL-12-aCTLA-4) to improve the therapeutic potential of DEVs by providing sufficient immune regulatory signals. Moreover, the carrier property of DEVs also contributes to the delivery of IL-12 and aCTLA-4 to lymph nodes. This indicates that the conjugation of DEVs with IL-12 and aCTLA-4 constitutes a complementary approach, where IL-12 and aCTLA-4 help to enhance the T cell activation effect of DEVs, and DEVs facilitate the delivery of IL-12 and aCTLA-4. Our results showed that DEV@IL-12-aCTLA-4 can enhance the Th1 immune response and reverse exhausted CD8+ T cells in the tumour microenvironment, effectively inducing robust T cell immune responses and inhibiting tumour growth in tumour-bearing mice. Overall, this study expands the theoretical foundation of DEVs and provides a universal strategy for optimizing cancer combination immunotherapy by reprogramming DEVs.
© 2025 The Author(s). Journal of Extracellular Vesicles published by Wiley Periodicals, LLC on behalf of the International Society for Extracellular Vesicles.

  • Cancer Research
  • Immunology and Microbiology

Topical TYK2 inhibitor ameliorates psoriasis-like dermatitis via the AKT-SP1-NGFR-AP1 pathway in keratinocytes.

In Clinical and Translational Medicine on 1 March 2025 by Fang, Z., Jiang, R., et al.

Tyrosine kinase 2 (TYK2)-dependent cytokine signalling is integral to the pathogenesis of psoriasis. While BMS-986165, a highly selective TYK2 inhibitor, has recently been approved for oral treatment of psoriasis, its therapeutic potential via topical application remains unexplored.
We aim to investigate the efficacy of topically applying TYK2 inhibitor in psoriasis and to elucidate the underlying mechanisms driving the therapeutic effects of this delivery approach.
1.5% BMS-986165 ointment was applied topically to the back skin of imiquimod (IMQ)-induced psoriatic mice. To identify potential target cells influenced by the topical TYK2 inhibitor, we performed single cell RNA sequencing (scRNA-seq) and flow cytometry on mouse lesions. The role of TYK2 in vitro was assessed by silencing its expression or administering BMS-986165 in human keratinocytes (KCs). Mechanistic insights into TYK2 function in KCs were further investigated using RNA-seq, dual luciferase reporter assay and ChIP-qPCR.
External use of 1.5% BMS-986165 ointment significantly ameliorated the IMQ-induced psoriasis-like dermatitis. Importantly, topical TYK2 inhibitor attenuated proinflammatory capability of KCs. In vitro, TYK2 inhibition suppressed the transcription of nerve growth factor receptor (NGFR) by disrupting the AKT-SP1 signalling pathway. This impairment hindered the activation of activator protein 1 (AP1), thereby weakening the proinflammatory potential of KCs.
This study reveals a novel therapeutic potential for selective TYK2 inhibitor in topical manner on psoriasis therapy, which might prompt the development of topical treatment for psoriasis. Crucially, our findings provide an underexplored regulatory mechanism of TYK2 inhibitor in psoriasis.
Topical TYK2 inhibitor alleviates psoriasis-like dermatitis. Topical TYK2 inhibitor reduces psoriasis progression through restraining the inflammatory responses of keratinocytes. The inhibition of TYK2 regulates the inflammatory response of keratinocytes through AKT-SP1-NGFR-AP1 pathway.
© 2025 The Author(s). Clinical and Translational Medicine published by John Wiley & Sons Australia, Ltd on behalf of Shanghai Institute of Clinical Bioinformatics.

In situ visualization of endothelial cell-derived extracellular vesicle formation in steady state and malignant conditions.

In Nature Communications on 22 October 2024 by Atkin-Smith, G. K., Santavanond, J. P., et al.

Endothelial cells are integral components of all vasculature within complex organisms. As they line the blood vessel wall, endothelial cells are constantly exposed to a variety of molecular factors and shear force that can induce cellular damage and stress. However, how endothelial cells are removed or eliminate unwanted cellular contents, remains unclear. The generation of large extracellular vesicles (EVs) has emerged as a key mechanism for the removal of cellular waste from cells that are dying or stressed. Here, we used intravital microscopy of the bone marrow to directly measure the kinetics of EV formation from endothelial cells in vivo under homoeostatic and malignant conditions. These large EVs are mitochondria-rich, expose the 'eat me' signal phosphatidylserine, and can interact with immune cell populations as a potential clearance mechanism. Elevated levels of circulating EVs correlates with degradation of the bone marrow vasculature caused by acute myeloid leukaemia. Together, our study provides in vivo spatio-temporal characterization of EV formation in the murine vasculature and suggests that circulating, large endothelial cell-derived EVs can provide a snapshot of vascular damage at distal sites.
© 2024. The Author(s).

  • Mus musculus (House mouse)

A broad-spectrum multiepitope vaccine against seasonal influenza A and B viruses in mice.

In EBioMedicine on 1 August 2024 by Yuan, L., Zhang, S., et al.

Influenza viruses pose a persistent threat to global public health, necessitating the development of innovative and broadly effective vaccines.
This study focuses on a multiepitope vaccine (MEV) designed to provide broad-spectrum protection against different influenza viruses. The MEV, containing 19 B-cell linear epitopes, 7 CD4+ T cells, and 11 CD8+ T cells epitopes identified through enzyme-linked immunospot assay (ELISPOT) in influenza viruses infected mice, was administered through a regimen of two doses of DNA vaccine followed by one dose of a protein vaccine in C57BL/6 female mice.
Upon lethal challenge with both seasonal circulating strains (H1N1, H3N2, BV, and BY) and historical strains (H1N1-PR8 and H3N2-X31), MEV demonstrated substantial protection against different influenza seasonal strains, with partial efficacy against historical strains. Notably, the increased germinal centre B cells and antibody-secreting cells, along with robust T cell immune responses, highlighted the comprehensive immune defence elicited by MEV. Elevated hemagglutinin inhibition antibody was also observed against seasonal circulating and historical strains. Additionally, mice vaccinated with MEV exhibited significantly lower counts of inflammatory cells in the lungs compared to negative control groups.
Our results demonstrated the efficacy of a broad-spectrum MEV against influenza viruses in mice. Conducting long-term studies to evaluate the durability of MEV-induced immune responses and explore its potential application in diverse populations will offer valuable insights for the continued advancement of this promising vaccine.
Funding bodies are described in the Acknowledgments section.
Copyright © 2024 The Author(s). Published by Elsevier B.V. All rights reserved.

  • Mus musculus (House mouse)
  • Immunology and Microbiology
View this product on CiteAb