HIF-1α is an essential mediator of IFN-γ–dependent immunity to Mycobacterium tuberculosis

J Braverman, KM Sogi, D Benjamin… - The Journal of …, 2016 - journals.aai.org
The Journal of Immunology, 2016journals.aai.org
The cytokine IFN-γ coordinates macrophage activation and is essential for control of
pathogens, including Mycobacterium tuberculosis. However, the mechanisms by which IFN-
γ controls M. tuberculosis infection are only partially understood. In this study, we show that
the transcription factor hypoxia-inducible factor-1α (HIF-1α) is an essential mediator of IFN-γ–
dependent control of M. tuberculosis infection both in vitro and in vivo. M. tuberculosis
infection of IFN-γ–activated macrophages results in a synergistic increase in HIF-1α protein …
Abstract
The cytokine IFN-γ coordinates macrophage activation and is essential for control of pathogens, including Mycobacterium tuberculosis. However, the mechanisms by which IFN-γ controls M. tuberculosis infection are only partially understood. In this study, we show that the transcription factor hypoxia-inducible factor-1α (HIF-1α) is an essential mediator of IFN-γ–dependent control of M. tuberculosis infection both in vitro and in vivo. M. tuberculosis infection of IFN-γ–activated macrophages results in a synergistic increase in HIF-1α protein levels. This increase in HIF-1α levels is functionally important, as macrophages lacking HIF-1α are defective for IFN-γ–dependent control of infection. RNA-sequencing demonstrates that HIF-1α regulates nearly one-half of all IFN-γ–inducible genes during infection of macrophages. In particular, HIF-1α regulates production of important immune effectors, including inflammatory cytokines and chemokines, eicosanoids, and NO. In addition, we find that during infection HIF-1α coordinates a metabolic shift to aerobic glycolysis in IFN-γ–activated macrophages. We find that this enhanced glycolytic flux is crucial for IFN-γ–dependent control of infection in macrophages. Furthermore, we identify a positive feedback loop between HIF-1α and aerobic glycolysis that amplifies macrophage activation. Finally, we demonstrate that HIF-1α is crucial for control of infection in vivo as mice lacking HIF-1α in the myeloid lineage are strikingly susceptible to infection and exhibit defective production of inflammatory cytokines and microbicidal effectors. In conclusion, we have identified HIF-1α as a novel regulator of IFN-γ–dependent immunity that coordinates an immunometabolic program essential for control of M. tuberculosis infection in vitro and in vivo.
journals.aai.org