Abstract

Abstract Generation and maintenance of memory T cells within tissue sites of infection is critical for long-term antiviral protection. Current knowledge of human antiviral responses is largely derived from studies sampling peripheral blood. Yet, little is known about human tissue-localized antiviral immunity. Utilizing our human donor tissue resource established in collaboration with LiveOnNY, we investigated how tissue, virus, and age shape T cell maintenance across diverse tissues and their function when presented with antigens derived from human cytomegalovirus (HCMV) or influenza A virus (flu). Using flow cytometry, we show that virus-specific CD8 T cells are maintained in diverse tissues with subset differentiation and distribution shaped by virus specificity and age. Compared to T cells recognizing HCMV, flu-specific T cells display higher levels of residency markers—CD69 and CD103—particularly in adult compared to pediatric donors. In contrast, HCMV-specific T cells are maintained in greater abundance as terminally-differentiated effector T cells. Sequencing of the T cell receptor CDR3b chain reveals that, while both HCMV- and flu-specific T cells demonstrate clonal overlap across multiple tissues, HCMV-specific T cells display greater clonality and less diversity. When presented with viral antigens, HCMV- and flu-specific T cells are polyfunctional. Additionally, single-cell transcriptome profiling reveals tissue localization is the primary determinant of antigen-driven responses. Together, these studies demonstrate the dynamics of T cell differentiation and maintenance throughout the human body, which is primarily driven by virus specificity, and the role of tissue localization in shaping antiviral response.

Full Text
Published version (Free)

Talk to us

Join us for a 30 min session where you can share your feedback and ask us any queries you have

Schedule a call