Natural Killer (NK)/melanoma cell interaction induces NK-mediated release of chemotactic High Mobility Group Box-1 (HMGB1) capable of amplifying NK cell …

M Parodi, M Pedrazzi, C Cantoni, M Averna… - …, 2015 - Taylor & Francis
M Parodi, M Pedrazzi, C Cantoni, M Averna, M Patrone, M Cavaletto, S Spertino, D Pende
Oncoimmunology, 2015Taylor & Francis
In this study we characterize a new mechanism by which Natural Killer (NK) cells may
amplify their recruitment to tumors. We show that NK cells, upon interaction with melanoma
cells, can release a chemotactic form of High Mobility Group Box-1 (HMGB1) protein
capable of attracting additional activated NK cells. We first demonstrate that the engagement
of different activating NK cell receptors, including those mainly involved in tumor cell
recognition can induce the active release of HMGB1. Then we show that during NK …
In this study we characterize a new mechanism by which Natural Killer (NK) cells may amplify their recruitment to tumors. We show that NK cells, upon interaction with melanoma cells, can release a chemotactic form of High Mobility Group Box-1 (HMGB1) protein capable of attracting additional activated NK cells. We first demonstrate that the engagement of different activating NK cell receptors, including those mainly involved in tumor cell recognition can induce the active release of HMGB1. Then we show that during NK-mediated tumor cell killing two HMGB1 forms are released, each displaying a specific electrophoretic mobility possibly corresponding to a different redox status. By the comparison of normal and perforin-defective NK cells (which are unable to kill target cells) we demonstrate that, in NK/melanoma cell co-cultures, NK cells specifically release an HMGB1 form that acts as chemoattractant, while dying tumor cells passively release a non-chemotactic HMGB1. Finally, we show that Receptor for Advanced Glycation End products is expressed by NK cells and mediates HMGB1-induced NK cell chemotaxis. Proteomic analysis of NK cells exposed to recombinant HMGB1 revealed that this molecule, besides inducing immediate chemotaxis, also promotes changes in the expression of proteins involved in the regulation of the cytoskeletal network. Importantly, these modifications could be associated with an increased motility of NK cells. Thus, our findings allow the definition of a previously unidentified mechanism used by NK cells to amplify their response to tumors, and provide additional clues for the emerging role of HMGB1 in immunomodulation and tumor immunity.
Taylor & Francis Online