loader
Home/
Journal of Immunology, Infection & Inflammatory Diseases

Full Text


Editorial

NOX2-Derived ROS: Not Just For Microbial Killing Anymore

Jamel El-Benna and Pham My-Chan Dang

Correspondence Address :

Jamel El-Benna
Universite Paris Diderot
Sorbonne Paris Cite
INSERM-U1149
CNRS-ERL8252
Centre de Recherche sur l Inflammation
Laboratoired Excellence Inflamex
Faculte de Medecine Bichat
Paris
France
Email: james.elbenna@inserm.fr

Received on: January 17, 2017 , Accepted on: January 25, 2017 , Published on: January 31, 2017

Citation: Jamel El-Benna, Pham My-Chan Dang (2017) NOX2-Derived ROS: Not Just For Microbial Killing Anymore

Copyright: 2017 Jamel El-Benna, et al. This is an open-access article distributed under the terms of the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.

  • Abstract

  • Fulltext

  • References

  • Tables & Figures

  • Download PDF

Abstract

Fulltext
             Neutrophils are the first cells to migrate out of the blood to the infection site during bacterial or fungal invasion [1]. After phagocytosis of the pathogen, the neutrophils produce and use different toxic agents to kill and eliminate the pathogen. Among these agents, reactive oxygen species (ROS) such as superoxide anion (O2-.), hydrogen peroxide (H2O2), hydroxyl radical (OH dot) and hypochlorous acid (HOCl) are pro-oxidants that react quickly with the various biological molecules (lipids, proteins and DNA) of the pathogens or the host [2-4]. The enzyme responsible for ROS production is called the phagocyte NADPH oxidase (NOX2). It is composed of two trans-membrane proteins (p22phox and gp91phox) that form the flavocytochrome b558, and four soluble cytosolic proteins (p47phox, p67phox, and p40phox and a small G-protein, Rac2 in neutrophils or Rac1 in other cells) [2-4]. In resting cells, the membrane and cytosolic components of NOX2 are segregated, but assemble at the membrane upon activation. For several decades, the role of NOX2-derived ROS was thought to be restricted to its toxic and killing properties to eliminate pathogens during the immune response. In fact, this function is well illustrated by chronic granulomatous disease (CGD), a genetic defect in which one of the NOX2 subunits is missing, resulting in absence of ROS production, and of killing/elimination of pathogens [2,3]. The generation of NOX2 knockout mice confirmed the requirement of NOX2-derived ROS for bacterial killing [5,6]. Although the bactericidal effect of NOX2-derived ROS is well established in vitro and in vivo in neutrophils, other effects of NOX2-derived ROS are also very important in innate and adaptive immunity. Indeed, NOX2 is also expressed in other immune cells, such as eosinophils, monocytes, macrophages, dendritic cells, and B and T lymphocytes, where they exert other functions [7,8]. In addition, some ROS such as H2O2 can diffuse outside the cells to act as a "paracrine-redox signaling agent", thus altering the functions of neighboring cells and potentially modulating the immune response [9,10]. ROS, and particularly the diffusible H2O2, can oxidize cellular macromolecules (proteins, lipids, DNA), leading to tissue injury or to the alteration of cellular functions such as an imbalance of intracellular signaling pathways, production of cytokines, and activation and release of proteases [11,12].
          Although NOX2 is also required for pathogen killing by monocytes and macrophages, it can regulate phagosomal protein degradation for antigen presentation, especially in macrophages, through oxidative inactivation of cysteines in cathepsin [13]. NOX2 also participates to cell signaling and protein synthesis in monocytes and macrophages [12]. Dendritic cells, which are professional antigen-presenting cells and key players in adaptive immunity, have developed a mild phagocytic pH environment to prevent complete degradation of peptides, thus allowing antigen presentation [14]. In these cells, NOX2 activity is much lower than in neutrophils, monocytes and macrophages, but regulates phagosomal proteolysis through pH alkalinization. Indeed, dismutation of O2-. into H2O2 consumes protons present in the phagosome, thus preventing intraphagosomal acidification and lowering protease activity for a better antigen processing. Thus, the principal role of NOX2 in dendritic cells is to control antigen cross-presentation. In circulating T and B cells, the expression of NOX2 has not been well established; however, it is well known that EBV-transformed B lymphocytes express all NOX2 subunits and produce ROS [8]. It was also reported that T cells express NOX2, which acts as a regulator of signaling pathways and cytokine production in these cells [7]. However phagocytes-derived ROS can dampen T-cell-dependent inflammation through alteration of the T-cell membrane oxidation status and of Th17/Treg cell development [10].
        In conclusion, NOX2-derived ROS were first considered as destroying/killing agents in professional phagocytes, and this function is well established. However, more subtle functions of NOX2-derived ROS, either in neutrophils or other cells, are emerging. These effects are either due to NOX2 expressed by the cell or to a diffusible ROS such as hydrogen peroxide acting as a signaling agent. Further studies will be needed to better understand these novel functions in immunity.
References
1. Nauseef WM, Borregaard N. Neutrophils at work. Nature Immunology. 2014;15:602-611.
2. Nauseef WM. How human neutrophils kill and degrade microbes:an integrated view. Immunological Reviews. 2007;219(1):88-102.
3.El-Benna J, Dang PM, Gougerot-Pocidalo MA, Elbim C. Phagocyte NADPH oxidase: a multicomponent enzyme es sential for host defenses. Archivum Immunologiae Therapiae Experimentalis. 2005;53(3):199-206.
4. El-Benna J, Hurtado-Nedelec M, Marzaioli V, Marie JC, Gougerot-Pocidalo MA, Dang PM. Priming of the neutrophil respiratory burst: role in host defense and inflammation. Immunological Reviews. 2016;273(1):180-193.
5. Morgenstern DE, Gifford MA, Li LL, Doerschuk CM, Dinauer MC. Absence of respiratory burst in X-linked chronic granulomatous disease mice leads to abnormalities in both host defense and inflammatory response to Aspergillus fumigatus. Journal of Experimental Medicine. 1997;185(2):207-218.
6. Jackson SH, Gallin JI, Holland SM. The p47phox mouse knock-out model of chronic granulomatous disease. J Exp Med. 1995;182(3):751-758.
7. Jackson SH, Devadas S, Kwon J, Pinto LA, Williams MS. T cells express a phagocyte-type NADPH oxidase that is activated after T cell receptor stimulation. Nat Immunol. 2004;5(8):818-827.
8. Belambri SA, Hurtado-Nedelec M, Senator A, et al. Phosphorylation of p47phox is required for receptor-mediated NADPH oxidase/NOX2 activation in Epstein-Barr virus-transformed human B lymphocytes. Am J Blood Res. 2012;2(3):187-193.
9. Reth M. Hydrogen peroxide as second messenger in lymphocyte activation. Nature Immunology. 2002;3(12):1129-1134.
10. Holmdahl R, Sareila O, Pizzolla A, et al. Hydrogen peroxide as an immunological transmitter regulating autoreactive T cells. Antioxidants & Redox Signaling. 2012;18(12):1463-1474.
11. Winterbourn CC. Reconciling the chemistry and biology of reactive oxygen species. Nature Chemical Biology. 2008;4(5):278-286.
12. Fialkow L, Wang Y, Downey GP. Reactive oxygen and nitrogen species as signaling molecules regulating neutrophil function. Free Radic Biol Med. 2007;42(2):153-164.
13. Rybicka JM, Balce DR, Khan MF, Krohn RM, Yates RM. NADPH oxidase activity controls phagosomal proteolysis in macrophages through modulation of the lumenal redox environment of phagosomes. Proc Natl Acad Sci U S A. 2010;107(23):10496-10501.
14. Kotsias F, Hoffmann E, Amigorena S, Savina A. Reactive oxygen species production in the phagosome: impact on antigen presentation in dendritic cells. Antioxid Redox Signal. 2013;18(6):714-729.
Tables & Figures

Download PDF