Macrophage activation as an archetype of mitochondrial repurposing

https://doi.org/10.1016/j.mam.2019.100838Get rights and content

Abstract

Mitochondria are metabolic organelles essential not only for energy transduction, but also a range of other functions such as biosynthesis, ion and metal homeostasis, maintenance of redox balance, and cell signaling. A hallmark example of how mitochondria can rebalance these processes to adjust cell function is observed in macrophages. These innate immune cells are responsible for a remarkable breadth of processes including pathogen elimination, antigen presentation, debris clearance, and wound healing. These diverse, polarized functions often include similarly disparate alterations in the metabolic phenotype associated with their execution. In this chapter, mitochondrial bioenergetics and signaling are viewed through the lens of macrophage polarization: both classical, pro-inflammatory activation and alternative, anti-inflammatory activation are associated with substantive changes to mitochondrial metabolism. Emphasis is placed on recent evidence that aims to clarify the essential – rather than associative – mitochondrial alterations, as well as accumulating data suggesting a degree of plasticity within the metabolic phenotypes that can support pro- and anti-inflammatory functions.

Introduction

Mitochondria play essential roles in cell physiology including, and extending far beyond, serving as the sites of oxidative phosphorylation. In addition to synthesizing the majority of ATP in most cell types (Nicholls and Ferguson, 2013; Pagliarini and Rutter, 2013), mitochondria are also hubs for biosynthesis, Ca2+ handling, iron homeostasis, redox balance, and signal transduction (Chandel, 2014; Murphy and Hartley, 2018; Shadel, 2012). Mitochondrially derived signals can be as varied as protein release from the intermembrane space (Jiang and Wang, 2004), metabolite efflux (Ryan et al., 2019), redox signals [e.g. reactive oxygen species (ROS) production] (Finkel, 2011; Murphy, 2009; Reczek and Chandel, 2015), and release of damage-associated molecular patterns (DAMPs) (Grazioli and Pugin, 2018; Picca et al., 2017).

It is increasingly appreciated that the roles of mitochondria in energy metabolism and as a signaling organelle are not necessarily discrete. In fact, one purpose of mitochondria as a signaling organelle is as a communicator of metabolic health status, serving a checkpoint or feedback function to instruct nuclear gene expression and cellular function (Chandel, 2015). Perhaps reflective of this, some proteins with essential roles in energy transduction have dual functions in pathways associated with cell death and disease. For example, cytochrome c is an essential component of the electron transport chain, but also triggers caspase-mediated apoptosis when released to the cytoplasm upon mitochondrial outer membrane permeabilization (Jiang and Wang, 2004; Tait and Green, 2010). Similarly, the FOF1-ATP synthase that generates ATP during oxidative phosphorylation also dimerizes to form the mitochondrial permeability transition pore, a channel causing inner membrane permeability and causative of pathologies such as ischemic heart injury and muscular dystrophy (Bernardi, 2013). This mitochondrial plasticity in shifting from energy powerhouse to signaling platform often involves a classical definition of reduced or impaired mitochondrial function (lowered oxidative phosphorylation, redox imbalance, etc.) (Jazwinski, 2013). Remarkably, however, emerging evidence in murine bone marrow-derived macrophages (BMDMs) suggests this does not necessarily translate to reduced or impaired cellular function.

Macrophages are cells of the innate immune system responsible for a remarkable breadth of functions essential to human health including killing pathogens, clearing subcellular debris, and repairing tissue (Adams and Hamilton, 1984). In addition to their role in healthy physiology, targeting macrophage function may lead to improved therapies for a variety of pathologies including tissue fibrosis, cancer, and metabolic disease (Wynn and Vannella, 2016).

Macrophages were first described over 130 years ago by Metchnikoff, who suggested that the observed phagocytosis by ameboid cells could be a means of host defense rather than a deleterious consequence of tissue damage or infection (Gordon, 2016). Specificity for the functional role and activation state of a macrophage at any given time is set by coordinating intrinsic, extrinsic, and environmental cues (Murray, 2017). This allows for the broad and sometimes opposing range of functions (e.g. “kill or repair”) these cells must execute. This so-called polarization state is often simplified through the classification of pro-inflammatory “M1” and anti-inflammatory “M2” macrophages (Gordon and Taylor, 2005). The nomenclature is drawn from the response to cytokines secreted by subsets of CD4+ helper T (Th) cells. A Th1 response is associated with immunity to bacteria and infections as well as secretion of interferon-γ  (IFN-γ) and tumor necrosis factor-α (TNF-α), whereas a Th2 response counteracts the microbicidal Th1 response and is associated with allergy and helminth immunity along with production of interleukin-4 (IL-4), IL-10, and IL-13.

Pro-inflammatory activation can occur via detection of pathogens by pattern recognition receptors (PRRs), proteins present at both the plasma and endosomal membranes (O'Neill et al., 2013). PRRs bind subsets of molecular scaffolds typically associated with microbes [pathogen associated molecular patterns (PAMPs)] or tissue damage [damage associated molecular patterns (DAMPS)]. Innate PRRs are broadly classified in two groups: membrane-bound, including Toll-like receptors (TLRs) and C-type lectin receptors (CLRs), and cytoplasmic, such as NOD-like receptors (NLRs) and RIG-I-like receptors (RLRs) (Akira and Takeda, 2004). Regardless of their localization, PRRs function by receptor-mediated activation of broad transcriptional programs, resulting in an orchestrated inflammatory response to eliminate pathogens or damaged cells (Barton and Medzhitov, 2003).

In addition to activation by PAMPs and DAMPs, macrophages can also be polarized to a range of activation states by secreted cytokines. This can occur in both paracrine [e.g. IFN-γ or anti-inflammatory IL-4 secreted by lymphocytes] and autocrine [e.g. pro-inflammatory interleukin-1β (IL-1β) or anti-inflammatory transforming growth factor-β (TGF-β)] manners upon receptor binding (Mosser and Edwards, 2008). In some cases, activation of PRR- and cytokine receptor-mediated activation integrate to amplify the inflammatory response. In the hallmark example, activation of TLR4, which recognizes the gram-negative bacteria membrane component lipopolysaccharide (LPS), synergizes with IFN-γ to create a powerful, composite pro-inflammatory signal (Held et al., 1999).

It is often accepted that the M1/M2 framework is based on rigidly prescribed conditions that rarely, if ever, occur physiologically, and do not necessarily reflect the spectral nature of macrophage activation (Martinez and Gordon, 2014). Nonetheless, the use of tightly controlled, in vitro polarization assays coupled with modern metabolic techniques has revealed profound and unexpected roles for several aspects of mitochondrial function in the control of the innate immune response.

An essential role for metabolism in macrophage function may be somewhat obvious in the traditional definition, as activation-induced phagocytosis, motility, and cytokine synthesis are all ATP-consuming processes that require commensurate changes in ATP production. Indeed, early biochemical studies showed that glucose consumption in naïve, unstimulated macrophages is far below its enzymatic capacity, suggesting an ability to quickly upregulate metabolic pathways in response to external stimuli (Newsholme et al., 1986). Abundant genetic and pharmacologic evidence now exists to demonstrate several essential roles for metabolism in shaping macrophage function, and these are surveyed in multiple comprehensive reviews (Arts et al., 2016a; Benmoussa et al., 2018; Caputa et al., 2019; Geeraerts et al., 2017; Langston et al., 2017; O'Neill and Pearce, 2016; Odegaard and Chawla, 2011; Russell et al., 2019; Van den Bossche et al., 2017; Viola et al., 2019; Weinberg et al., 2015). Such roles include increased and/or rerouted metabolic flux through specific pathways, post-translational modifications by metabolic intermediates, and signaling initiated by metabolites, redox triggers, or nucleic acids.

In the present manuscript, we focus on mitochondria as a hub for both energy transduction as well as metabolite and signal generation, and discuss how these different modes are used to shape macrophage function. It is unquestioned that, in murine macrophages, oxidative metabolism is associated with anti-inflammatory activation (stimulation with IL-4 ± IL-13), and classical inflammatory activation (LPS ± IFN-γ) involves the collapse of oxidative phosphorylation and repurposing of mitochondria towards accumulation of metabolites and other pro-inflammatory triggers. However, the essential, targetable metabolic requirements to adjust macrophage function may not be as black-or-white as initially thought. In fact, a crude analogy could be drawn to macrophage polarization itself. Similarly to how the M1/M2 paradigm is perhaps too bifurcated and discrete to reflect the graded nature of macrophage function (Martinez and Gordon, 2014; Murray, 2017), so too may be a one-size-fits-all approach that suggests ‘polarized’ metabolic phenotypes are indispensable for pro- or anti-inflammatory macrophage activation. Rather, accumulating evidence suggests bioenergetic and signaling roles for mitochondria in both pro- and anti-inflammatory macrophage activation, although these manifest in different ways.

Section snippets

Overview of mitochondrial energy metabolism

Of course, the best described function of mitochondria is energy metabolism and the production of ATP through oxidative phosphorylation (Nicholls and Ferguson, 2013). Mitochondria generate ATP through a series of energy transducing processes. The chemical energy in nutrients such as sugars, amino acids, and fatty acids is first harvested through the tricarboxylic acid (TCA) cycle (Fig. 1). The TCA cycle (or Krebs cycle) is a series of eight consecutive enzymes that oxidize energy-rich

Mitochondrial signaling in cell physiology

One of the more exciting and remarkable developments in mitochondrial biology over the past decade has been appreciation of the myriad ways in which the organelle links metabolism to cell physiology via TCA cycle metabolites (Dang and Su, 2017; Lu and Thompson, 2012; Mills et al., 2017; Ryan et al., 2019; Sciacovelli and Frezza, 2017; Sivanand et al., 2018). Much like the bioenergetic alterations discussed previously, early inspiration likely came from the field of cancer metabolism following

Concluding remarks

In addition to autoimmune diseases that may be expected to have a dysregulated balance in the macrophage polarization state, it is now well accepted that an imbalance between pro- and anti-inflammatory macrophage function can underlie a range of diseases as varied as diet-induced obesity (Lumeng et al., 2008), insulin resistance (Olefsky and Glass, 2010), cardiovascular disease (Bolego et al., 2013; Zhou and Tian, 2018), non-alcoholic steatotic hepatitis (Kazankov et al., 2019),

Acknowledgements

Anthony E. Jones is supported by the UCLA Tumor Cell Biology Training Program (USHHS Ruth L. Kirschstein Institutional National Research Service Award #T32 CA009056).

References (177)

  • V. Adam-Vizi et al.

    Calcium and mitochondrial reactive oxygen species generation: how to read the facts

    J. Alzheimer's Dis.

    (2010)
  • D.O. Adams et al.

    The cell biology of macrophage activation

    Annu. Rev. Immunol.

    (1984)
  • L.B. Adams et al.

    Microbiostatic effect of murine-activated macrophages for Toxoplasma gondii. Role for synthesis of inorganic nitrogen oxides from L-arginine

    J. Immunol.

    (1990)
  • S.M.U. Ahmed et al.

    Nrf2 signaling pathway: pivotal roles in inflammation

    Biochim. Biophys. Acta (BBA) - Mol. Basis Dis.

    (2017)
  • S. Akira et al.

    Toll-like receptor signalling

    Nat. Rev. Immunol.

    (2004)
  • D. Arsenijevic et al.

    Disruption of the uncoupling protein-2 gene in mice reveals a role in immunity and reactive oxygen species production

    Nat. Genet.

    (2000)
  • R.J.W. Arts et al.

    Immunometabolic circuits in trained immunity

    Semin. Immunol.

    (2016)
  • R.J.W. Arts et al.

    Glutaminolysis and fumarate accumulation integrate immunometabolic and epigenetic programs in trained immunity

    Cell Metabol.

    (2016)
  • S.H. Baik et al.

    A breakdown in metabolic reprogramming causes microglia dysfunction in alzheimer's disease

    Cell Metabol.

    (2019)
  • J.D. Bailey et al.

    Nitric oxide modulates metabolic remodeling in inflammatory macrophages through TCA cycle regulation and itaconate accumulation

    Cell Rep.

    (2019)
  • M. Bambouskova et al.

    Electrophilic properties of itaconate and derivatives regulate the IκBζ-ATF3 inflammatory axis

    Nature

    (2018)
  • G.M. Barton et al.

    Toll-like receptor signaling pathways

    Science

    (2003)
  • K. Benmoussa et al.

    How mitochondrial metabolism contributes to macrophage phenotype and functions

    J. Mol. Biol.

    (2018)
  • P. Bernardi

    The mitochondrial permeability transition pore: a mystery solved?

    Front. Physiol.

    (2013)
  • C. Bolego et al.

    Macrophage function and polarization in cardiovascular disease a role of estrogen signaling?

    Arterioscler. Thromb. Vasc. Biol.

    (2013)
  • M.D. Brand et al.

    Assessing mitochondrial dysfunction in cells

    Biochem. J.

    (2011)
  • J.M. Buescher et al.

    A roadmap for interpreting 13 C metabolite labeling patterns from cells

    Curr. Opin. Biotechnol.

    (2015)
  • A.M. Cameron et al.

    Inflammatory macrophage dependence on NAD + salvage is a consequence of reactive oxygen species–mediated DNA damage

    Nat. Immunol.

    (2019)
  • B. Cannon et al.

    Brown adipose tissue: function and physiological significance

    Physiol. Rev.

    (2004)
  • G. Caputa et al.

    Metabolic adaptations of tissue-resident immune cells

    Nat. Immunol.

    (2019)
  • M.K. Cathcart

    Regulation of superoxide anion production by NADPH oxidase in monocytes/macrophages: contributions to atherosclerosis

    Arterioscler. Thromb. Vasc. Biol.

    (2004)
  • S.M. Ceccarelli et al.

    Carnitine palmitoyltransferase (CPT) modulators: a medicinal chemistry perspective on 35 years of research

    J. Med. Chem.

    (2011)
  • N.S. Chandel

    Evolution of mitochondria as signaling organelles

    Cell Metabol.

    (2015)
  • N.S. Chandel

    Mitochondria as signaling organelles

    BMC Biol.

    (2014)
  • D. Chen et al.

    Chloroquine modulates antitumor immune response by resetting tumor-associated macrophages toward M1 phenotype

    Nat. Commun.

    (2018)
  • E.T. Chouchani et al.

    Cardioprotection by S-nitrosation of a cysteine switch on mitochondrial complex i

    Nat. Med.

    (2013)
  • E.T. Chouchani et al.

    Ischaemic accumulation of succinate controls reperfusion injury through mitochondrial ROS

    Nature

    (2014)
  • E.T. Chouchani et al.

    A unifying mechanism for mitochondrial superoxide production during ischemia-reperfusion injury

    Cell Metabol.

    (2016)
  • H.R. Christofk et al.

    The M2 splice isoform of pyruvate kinase is important for cancer metabolism and tumour growth

    Nature

    (2008)
  • M.W.J. Cleeter et al.

    Reversible inhibition of cytochrome c oxidase, the terminal enzyme of the mitochondrial respiratory chain, by nitric oxide. Implications for neurodegenerative diseases

    FEBS Lett.

    (1994)
  • E. Clementi et al.

    Persistent inhibition of cell respiration by nitric oxide: crucial role of S-nitrosylation of mitochondrial complex I and protective action of glutathione

    Proc. Natl. Acad. Sci. U. S. A

    (1998)
  • T. Cordes et al.

    Itaconic acid: the surprising role of an industrial compound as a mammalian antimicrobial metabolite

    Annu. Rev. Nutr.

    (2015)
  • T. Cordes et al.

    Immunoresponsive gene 1 and itaconate inhibit succinate dehydrogenase to modulate intracellular succinate levels

    J. Biol. Chem.

    (2016)
  • A.J. Covarrubias et al.

    Control of macrophage metabolism and activation by mTOR and Akt signaling

    Semin. Immunol.

    (2015)
  • A.J. Covarrubias et al.

    Akt-mTORC1 signaling regulates Acly to integrate metabolic input to control of macrophage activation

    Elife

    (2016)
  • T. Cramer et al.

    HIF-1α is essential for myeloid cell-mediated inflammation

    Cell

    (2003)
  • A.R. Crofts

    The cytochrome bc 1 complex: function in the context of structure

    Annu. Rev. Physiol.

    (2004)
  • L. Dang et al.

    Isocitrate dehydrogenase mutation and ( R )-2-Hydroxyglutarate: from basic discovery to therapeutics development

    Annu. Rev. Biochem.

    (2017)
  • D.P. De Souza et al.

    Autocrine IFN-I inhibits isocitrate dehydrogenase in the TCA cycle of LPS-stimulated macrophages

    J. Clin. Investig.

    (2019)
  • D. De Stefani et al.

    Structure and function of the mitochondrial calcium uniporter complex

    Biochim. Biophys. Acta Mol. Cell Res.

    (2014)
  • A. Dhir et al.

    Mitochondrial double-stranded RNA triggers antiviral signalling in humans

    Nature

    (2018)
  • A.S. Divakaruni et al.

    The regulation and physiology of mitochondrial proton leak

    Physiology

    (2011)
  • A.S. Divakaruni et al.

    Etomoxir inhibits macrophage polarization by disrupting CoA homeostasis

    Cell Metabol.

    (2018)
  • A.S. Divakaruni et al.

    Analysis and interpretation of microplate-based oxygen consumption and pH data

    Methods in Enzymology

    (2014)
  • S.K. Dolan et al.

    The glyoxylate shunt, 60 Years on

    Annu. Rev. Microbiol.

    (2018)
  • M.R. Duchen

    Mitochondria and calcium: from cell signalling to cell death

    J. Physiol.

    (2000)
  • R. El-Khoury et al.

    Engineering the alternative oxidase gene to better understand and counteract mitochondrial defects: state of the art and perspectives

    Br. J. Pharmacol.

    (2014)
  • B. Everts et al.

    Commitment to glycolysis sustains survival of NO-producing inflammatory dendritic cells

    Blood

    (2012)
  • B.A. Fensterheim et al.

    The TLR4 agonist monophosphoryl lipid A drives broad resistance to infection via dynamic reprogramming of macrophage metabolism

    J. Immunol.

    (2018)
  • T. Finkel

    Signal transduction by reactive oxygen species

    J. Cell Biol.

    (2011)
  • Cited by (0)

    View full text