Abstract

CD8 T cell responses have three phases: expansion, contraction, and memory. Dynamic alterations in proliferation and apoptotic rates control CD8 T cell numbers at each phase, which in turn dictate the magnitude of CD8 T cell memory. Identification of signaling pathways that control CD8 T cell memory is incomplete. The PI3K/Akt signaling pathway controls cell growth in many cell types by modulating the activity of FOXO transcription factors. But the role of FOXOs in regulating CD8 T cell memory remains unknown. We show that phosphorylation of Akt, FOXO and mTOR in CD8 T cells occurs in a dynamic fashion in vivo during an acute viral infection. To elucidate the potentially dynamic role for FOXO3 in regulating homeostasis of activated CD8 T cells in lymphoid and non-lymphoid organs, we infected global and T cell-specific FOXO3-deficient mice with Lymphocytic Choriomeningitis Virus (LCMV). We found that FOXO3 deficiency induced a marked increase in the expansion of effector CD8 T cells, preferentially in the spleen, by T cell-intrinsic mechanisms. Mechanistically, the enhanced accumulation of proliferating CD8 T cells in FOXO3-deficient mice was not attributed to an augmented rate of cell division, but instead was linked to a reduction in cellular apoptosis. These data suggested that FOXO3 might inhibit accumulation of growth factor-deprived proliferating CD8 T cells by reducing their viability. By virtue of greater accumulation of memory precursor effector cells during expansion, the numbers of memory CD8 T cells were strikingly increased in the spleens of both global and T cell-specific FOXO3-deficient mice. The augmented CD8 T cell memory was durable, and FOXO3 deficiency did not perturb any of the qualitative attributes of memory T cells. In summary, we have identified FOXO3 as a critical regulator of CD8 T cell memory, and therapeutic modulation of FOXO3 might enhance vaccine-induced protective immunity against intracellular pathogens.

Highlights

  • The ability of the immune system to respond rapidly and vigorously to antigen re-exposure is termed immunological memory, which is one of the tenets of adaptive immunity [1,2]

  • We have identified the transcription factor FOXO3 as a crucial regulator of the magnitude of CD8 T cell memory

  • Loss of FOXO3 activity in T cells led to a durable increase in the number of memory CD8 T cells, and the functional quality of FOXO3-deficient memory CD8 T cells was unaffected by FOXO3 deficiency

Read more

Summary

Introduction

The ability of the immune system to respond rapidly and vigorously to antigen re-exposure is termed immunological memory, which is one of the tenets of adaptive immunity [1,2]. As compared to naıve T cells, memory T cells are hyper-reactive to antigenic stimulation and swiftly proliferate and/or differentiate into effector cells to confer protective immunity expeditiously [5,6,7,8]. The ability of memory T cells to confer protective immunity depends upon the number and quality of memory T cells [5,9,10,11,12,13]. During a CD8 T cell response, engagement of the TCR, along with appropriate co-stimulatory and inflammatory signals, activate naıve T cells to proliferate and differentiate into effector cells [1,4,8,13,14]. The number of memory CD8 T cells generated depends largely upon the magnitude of the expansion of MPECs during the T cell response. Substantial progress has been made in deciphering the extracellular signals and transcription factors that regulate the differentiation of MPECs [1], but the signaling pathways that govern the number of MPECs, their differentiation into memory CD8 T cells, and the maintenance of CD8 T cell memory are not fully understood

Methods
Results
Discussion
Conclusion
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