24,25 An FcR-mediated activity of a broadly reactive HIV neutrali

24,25 An FcR-mediated activity of a broadly reactive HIV neutralizing monoclonal antibody (mAb) has also been shown to contribute to protective efficacy in a macaque challenge model,26 further invoking a role of NK cells. Moreover, the recent modest success of

the RV144 HIV clinical vaccine trial in Thailand27 has been suggested to be partly the result of ADCC activity elicited by the vaccine regimen.28 Hence, there is heightened interest in the HIV vaccine field in NK-cell-mediated effector functions. Despite the potential role played by NK cells during innate and adaptive immune responses against HIV/SIV, and the utility of rhesus macaque models, the variety and function of roles Staurosporine of different macaque NK cell subpopulations have not been exhaustively explored. Previous reports have described macaque circulatory NK cells as CD3− CD8α+ CD20−/dim NKG2A+ cells that can be further divided into four subpopulations based on their CD56 and CD16 expression patterns.29–31 However, CD8α expression on different human NK cell subsets is variable,32,33 and therefore CD8α expression Opaganib manufacturer is not necessarily a requisite marker for NK cell phenotyping. In this regard, a minor subset of CD8α− NK cells has been recently identified in healthy and HIV-infected chimpanzees.34 Furthermore, it has been shown that peripheral

blood mononuclear cells (PBMCs) from HIV-infected mothers and their infants that strongly respond to HIV-1 peptide stimulation [by up-regulating interferon-γ (IFN-γ) and interleukin-2 (IL-2) production in both CD3− CD8− and CD3− CD8+ cells] are less likely to transmit and acquire infection, respectively.35 For the reasons mentioned above, in the present study we evaluated the presence of NK cell lineage markers on macaque CD3− CD14− CD20−/dim CD8α− PBMCs, and the potential of these cells to mediate functional responses. Using multi-parametric flow cytometry, we identified a subpopulation of

circulatory CD8α− NK cells in naive and SIV-infected macaques that expressed the CD56 and/or CD16 NK cell lineage markers. A subset of these CD3− CD14− CD20−/dim CD8α− cells (from now on referred to as CD8α− NK cells) also co-expressed granzyme B, perforin, NKG2D and KIR2D. Upon cytokine triclocarban stimulation, CD8α− NK cells up-regulated CD69 expression and IFN-γ mRNA transcription and produced low levels of tumour necrosis factor-α (TNF-α). Importantly, enriched CD8α− NK cells were capable of mediating direct cell lysis as well as antibody-dependent killing, suggesting a potential for contributing to both innate and adaptive immune responses. Rhesus macaques (n = 30, 17 naive and 13 chronically infected with SIV) used in this study were housed at the National Institutes of Health (NIH) Division of Veterinary Resources (Bethesda, MD), at Bioqual, Inc.

Comments are closed.