Elsevier

Free Radical Biology and Medicine

Volume 76, November 2014, Pages 34-46
Free Radical Biology and Medicine

Review Article
NADPH oxidase- and mitochondria-derived reactive oxygen species in proinflammatory microglial activation: a bipartisan affair?

https://doi.org/10.1016/j.freeradbiomed.2014.07.033Get rights and content

Highlights

  • Reactive oxygen species, particularly H2O2, participate in microglial activation.

  • This H2O2 is primarily of NADPH oxidase rather than of mitochondrial origin.

  • Common techniques used to study mitochondrial superoxide are critically evaluated.

  • Evidence linking mitochondria to NLRP3 inflammasome activation is not conclusive.

Abstract

Microglia are the resident immune cells of the brain and play major roles in central nervous system development, maintenance, and disease. Brain insults cause microglia to proliferate, migrate, and transform into one or more activated states. Classical M1 activation triggers the production of proinflammatory factors such as tumor necrosis factor-α, interleukin-1β (IL-1β), nitric oxide, and reactive oxygen species (ROS), which, in excess, can exacerbate brain injury. The mechanisms underlying microglial activation are not fully understood, yet reactive oxygen species are increasingly implicated as mediators of microglial activation. In this review, we highlight studies linking reactive oxygen species, in particular hydrogen peroxide derived from NADPH oxidase-generated superoxide, to the classical activation of microglia. In addition, we critically evaluate controversial evidence suggesting a specific role for mitochondrial reactive oxygen species in the activation of the NLRP3 inflammasome, a multiprotein complex that mediates the production of IL-1β and IL-18. Finally, the limitations of common techniques used to implicate mitochondrial ROS in microglial and inflammasome activation, such as the use of the mitochondrially targeted ROS indicator MitoSOX and the mitochondrially targeted antioxidant MitoTEMPO, are also discussed.

Introduction

Microglia are macrophage-like immune cells of the central nervous system that play major roles in both health and disease [1], [2]. In health, microglia are highly motile cells that survey their surrounding environment to maintain homeostasis [3], promote synaptogenesis, and regulate the integrity of the blood–brain barrier [4], [5]. However, microglia become activated and proliferate after acute brain injury or in chronic neurodegenerative diseases such as Parkinson disease and Alzheimer disease [1], [2]. Activated microglia undergo drastic morphological changes, transforming from a ramified to an amoeboid morphology [1], [2]. This change in morphology is thought to favor mobility and increase phagocytic activity [6]. Microglia cordon off damaged neurons and assist in the repair and regrowth of compromised cells by releasing various metabolites, growth factors, and cytokines. However, prolonged or excessive microglial activation becomes maladaptive [7]. This is partly because activation of microglia takes multiple shapes. Classical M1 microglial activation results in the proinflammatory state implicated in neurotoxicity, for instance, the degeneration of dopaminergic neurons in Parkinson disease [3], [8], [9], [10], [11] or of motor neurons in amyotrophic lateral sclerosis [12], [13]. In addition to the creation of a toxic proinflammatory milieu, activated microglia may also contribute to cell death after brain injuries via phagocytosis of live neurons [14], [15], [16]. In contrast to M1 activation, alternative microglial activation states, generally classified as M2 with further subclasses, counteract proinflammatory mediators and promote repair processes such as remodeling of the extracellular matrix and angiogenesis [17]. Impairing excessive M1 microglial activation and/or promoting a transition toward alternative M2 activation are promising neuroprotective treatment strategies receiving considerable attention [18], [19]. It is important to understand the M1 activation process in greater detail to successfully achieve this goal.

To this end, extensive progress has been made in elucidating the initial molecular signaling pathways underlying microglial activation, especially through activation of Toll-like and cytokine receptors (see [9], [20] for detailed reviews). Treatment with the bacterial endotoxin lipopolysaccharide (LPS)1 alone or in combination with the proinflammatory cytokine interferon-γ (IFN-γ) is frequently used to induce and study M1 microglial activation in vitro. These factors activate Toll-like receptor 4 (TLR4, also called CD14) and IFN-γ receptor, respectively. LPS also induces TLR4-independent signaling by binding phagocytic scavenger receptors [21] and by binding macrophage antigen complex I (MAC1, also called CD11b/CD18), a pattern recognition receptor linked to the superoxide-generating enzyme NADPH oxidase (alternatively called Phox, for phagocyte oxidase) [22]. Reactive oxygen species (ROS) production, activation of kinase cascades, and changes in gene transcription occur subsequent to receptor ligation. ROS such as hydrogen peroxide (H2O2) are suggested to act as second messengers in cytokine responses [23], [24]. H2O2 activates mitogen-activated protein kinase (MAPK) cascades, partly through oxidation of catalytic cysteines on MAPK-inactivating phosphatases [25], [26], [27], and induces nuclear factor κB (NF-κB) translocation from the cytosol to the nucleus [28], [29]. Initiation of NF-κB-dependent gene transcription is a key step in the production of proinflammatory mediators [30], [31].

Here, we review evidence linking ROS to the proinflammatory M1 microglial activation state and the NOD-like receptor family pyrin domain-containing 3 (NLRP3) inflammasome complex, a mediator of caspase-1-dependent interleukin-1β (IL-1β) secretion. We also discuss the extent to which mitochondrial dysfunction can be incriminated. Importantly, although much can be extrapolated from macrophages and other immune cells that share many features of microglial activation, precise roles for ROS and the NLRP3 inflammasome in the activation of primary microglia require further elucidation.

Section snippets

A role for NADPH oxidase in the proinflammatory state

Early work linking ROS to the M1 microglial activation state used a pharmacological approach (Fig. 1, i). Kang et al. demonstrated that the superoxide dismutase (SOD)/catalase mimetic Mn(III)tetrakis(1-methyl-4-pyridyl)porphyrin (MnTMPyP) or the NADPH oxidase/flavoprotein inhibitor diphenylene iodonium (DPI) reduced NF-κB activation and IL-1β transcription in the mouse microglial cell line BV2 in response to treatment with an Alzheimer disease-relevant amyloid-β (Aβ) peptide [32]. Additional

Reactive oxygen species, nitric oxide, and reactive microgliosis

Microglia in vitro continue to produce proinflammatory factors after removal of activating agent [47], and microglia in vivo frequently remain activated over long time periods after injury or onset of disease [50], [51]. This continued activation is probably due to a phenomenon termed reactive microgliosis, a self-propelling cycle mediated by intracellular ROS and NO that serves to maintain the M1 phenotype [47]. Hence, ROS play a vital role in both the initial activation of microglia and their

MAPK and NF-κB as hydrogen peroxide-sensitive regulators of microglial activation

The precise mechanisms by which ROS, and H2O2 in particular, promote microglial proliferation and the M1 transition remain incompletely characterized. Much of the work on the proinflammatory role of ROS, in both nonmicroglial and microglial cell types, has focused on the MAPK signaling pathway and the downstream transcription factor NF-κB (Fig. 1, iii). Although most reports do not examine exactly how ROS increase MAPK phosphorylation during the activation process, oxidation of catalytic

Rotenone and microglial activation—is mitochondrial superoxide involved?

To this point we have focused on studies pointing to a role for NADPH oxidase in microglial activation. However, mitochondria are a possible source of the component of LPS-induced intracellular ROS that was not eliminated by gp91phox knockout [8]. Superoxide anion is the primary ROS produced by mitochondria, mostly in the matrix, but it does not readily cross biological membranes [79]. MnSOD (SOD2) efficiently converts mitochondrial matrix superoxide into H2O2, which diffuses into the

A proinflammatory amplification loop initiated by mitochondrial superoxide? Lessons from macrophages

Not long ago, Kasahara et al. [101] took an interesting alternative approach to address whether there is a mitochondrial contribution to the activation phenotype in macrophages, the peripheral “cousins” of microglia. Rho zero (ρ0) cells lacking mitochondrial DNA were created from the RAW 264.7 macrophage cell line. The cells exhibited defective oxygen consumption, diminished ROS production in response to LPS, and attenuated TNF-α and IL-6 secretion [101]. An H2O2-generating system consisting of

NADPH oxidase-independent reactive oxygen species and NLRP3

Our discussion to this point has centered mostly on TNF-α; however, IL-1β is also one of the major proinflammatory cytokines in the brain. Release of IL-1β, in contrast to TNF-α, is regulated enzymatically through caspase-1 activity. The NLRP3 (also called NALP3) inflammasome is a protein scaffolding complex that induces the secretion of the cytokines IL-1β and IL-18 from their proforms by activating caspase-1 (Fig. 1, iv) [111]. Present in most immune cells including microglia, the NLRP3

Microglia vs macrophages

The vast majority of studies on NLRP3 inflammasome activation were performed in macrophages or other peripheral immune cells, particularly with regard to mitochondrial involvement. It is accepted by most that microglia are simply the resident macrophages in the brain and indeed the two phagocytic cell types are quite similar. For example, antigens that are commonly used to identify microglia, such as CD11b or CD68, are also present in macrophages [157]. However, although both cell types are of

M1 vs M2 activation

The majority of this review has focused on proinflammatory microglial activation. This mechanism of activation is referred to as a “classical” phenotype, or M1 activation [161]. However, microglia can also exhibit an “alternative” M2 activation [19], which serves to reduce production of proinflammatory mediators and promote brain repair. Whereas M1 polarization may be beneficial immediately after brain injury by phagocytic clearing of debris, M2 polarization may help to resolve a

Concluding remarks

Although it is generally accepted that reactive oxygen species, and in particular H2O2, play a role in M1 microglial activation, there is still disagreement as to the extent. A number of studies indicate that pro-oxidants cause microglial activation, whereas even more demonstrate that antioxidants impair activation. However, concerns exist about the methodology used to quantify and modulate ROS, particularly superoxide of mitochondrial origin. It is noteworthy that flawed approaches such as the

Acknowledgments

The authors acknowledge funding from NIH R01 NS085165 to B.M.P. In addition, the authors thank Dr. David Loane for helpful feedback during the preparation of the review.

References (163)

  • H. Kamata et al.

    Reactive oxygen species promote TNFalpha-induced death and sustained JNK activation by inhibiting MAP kinase phosphatases

    Cell

    (2005)
  • KW Zeng et al.

    Schisandrin B exerts anti-neuroinflammatory activity by inhibiting the Toll-like receptor 4-dependent MyD88/IKK/NF-κB signaling pathway in lipopolysaccharide-induced microglia

    Eur. J. Pharmacol.

    (2012)
  • W Zheng et al.

    TNFα and IL-1β are mediated by both TLR4 and Nod1 pathways in the cultured HAPI cells stimulated by LPS

    Biochem. Biophys. Res. Commun.

    (2012)
  • A. Majander et al.

    Diphenyleneiodonium inhibits reduction of iron–sulfur clusters in the mitochondrial NADH-ubiquinone oxidoreductase (Complex I)

    J. Biol. Chem.

    (1994)
  • Y. Li et al.

    an NAD(P)H oxidase inhibitor, also potently inhibits mitochondrial reactive oxygen species production

    Biochem. Biophys. Res. Commun.

    (1998)
  • A.J. Lambert et al.

    Diphenyleneiodonium acutely inhibits reactive oxygen species production by mitochondrial complex I during reverse, but not forward electron transport

    Biochim. Biophys. Acta

    (2008)
  • D.J. Loane et al.

    Activation of metabotropic glutamate receptor 5 modulates microglial reactivity and neurotoxicity by inhibiting NADPH oxidase

    J. Biol. Chem.

    (2009)
  • H Eguchi et al.

    Hydrogen peroxide enhances LPS-induced nitric oxide production via the expression of interferon beta in BV-2 microglial cells

    Neurosci. Lett.

    (2011)
  • Z Lijia et al.

    A self-propelling cycle mediated by reactive oxide species and nitric oxide exists in LPS-activated microglia

    Neurochem. Int.

    (2012)
  • C.C. Winterbourn

    Toxicity of iron and hydrogen peroxide: the Fenton reaction

    Toxicol. Lett.

    (1995)
  • A Kumar et al.

    Neuroinflammation after traumatic brain injury: opportunities for therapeutic intervention

    Brain Behav. Immun.

    (2012)
  • J.L. Goodwin et al.

    Uemura E. Integrin Mac-1 and beta-amyloid in microglial release of nitric oxide

    Brain Res.

    (1997)
  • D. Zhang et al.

    Microglial MAC1 receptor and PI3K are essential in mediating beta-amyloid peptide-induced microglial activation and subsequent neurotoxicity

    J. Neuroinflammation

    (2011)
  • A Roy et al.

    Reactive oxygen species up-regulate CD11b in microglia via nitric oxide: implications for neurodegenerative diseases

    Free Radic. Biol. Med.

    (2008)
  • H Akiyama et al.

    Brain microglia constitutively express beta-2 integrins

    J. Neuroimmunol.

    (1990)
  • A. Roy et al.

    Up-regulation of microglial CD11b expression by nitric oxide

    J. Biol. Chem.

    (2006)
  • Y Yoshino et al.

    Superoxide anion contributes to the induction of tumor necrosis factor alpha (TNFα) through activation of the MKK3/6–p38 MAPK cascade in rat microglia

    Brain Res.

    (2011)
  • I.M. Chiu et al.

    A neurodegeneration-specific gene-expression signature of acutely isolated microglia from an amyotrophic lateral sclerosis mouse model

    Cell Rep

    (2013)
  • S.H. Kim et al.

    Importance of MAPK pathways for microglial pro-inflammatory cytokine IL-1 beta production

    Neurobiol. Aging

    (2004)
  • ZS Vexler et al.

    Fructose-1,6-bisphosphate preserves intracellular glutathione and protects cortical neurons against oxidative stress

    Brain Res.

    (2003)
  • YC Kim et al.

    Fructose-1,6-bisphosphate attenuates induction of nitric oxide synthase in microglia stimulated with lipopolysaccharide

    Life Sci.

    (2012)
  • SU Kim et al.

    Peroxiredoxin I is a ROS/p38 MAPK-dependent inducible antioxidant that regulates NF-κB-mediated iNOS induction and microglial activation

    J. Neuroimmunol.

    (2013)
  • L.H. Sanders et al.

    Oxidative damage to macromolecules in human Parkinson disease and the rotenone model

    Free Radic. Biol. Med.

    (2013)
  • H. Zhou et al.

    Rotenone activates phagocyte NADPH oxidase by binding to its membrane subunit gp91phox

    Free Radic. Biol. Med.

    (2012)
  • H. Klintworth et al.

    Rotenone and paraquat do not directly activate microglia or induce inflammatory cytokine release

    Neurosci. Lett.

    (2009)
  • B.R. Brinkley et al.

    Rotenone inhibition of spindle microtubule assembly in mammalian cells

    Exp. Cell Res.

    (1974)
  • L.E. Marshall et al.

    Rotenone inhibition of tubulin self-assembly

    Biochim. Biophys. Acta

    (1978)
  • Y. Ren et al.

    Selective vulnerability of dopaminergic neurons to microtubule depolymerization

    J. Biol. Chem.

    (2005)
  • LA Voloboueva et al.

    Inflammatory response of microglial BV-2 cells includes a glycolytic shift and is modulated by mitochondrial glucose-regulated protein 75/mortalin

    FEBS Lett.

    (2013)
  • DS Davis et al.

    An introduction to CNS-resident microglia: definitions, assays, and functional roles in health and disease

  • ME Tremblay et al.

    The role of microglia in the healthy brain

    J. Neurosci.

    (2011)
  • RJ Horvath et al.

    Differential migration, LPS-induced cytokine, chemokine, and NO expression in immortalized BV-2 and HAPI cell lines and primary microglial cultures

    J. Neurochem.

    (2008)
  • ML Block et al.

    Microglia-mediated neurotoxicity: uncovering the molecular mechanisms

    Nat. Rev. Neurosci.

    (2007)
  • T Wang et al.

    Protective effect of the SOD/catalase mimetic MnTMPyP on inflammation-mediated dopaminergic neurodegeneration in mesencephalic neuronal–glial cultures

    J. Neuroimmunol.

    (2013)
  • C. Cunningham

    Microglia and neurodegeneration: the role of systemic inflammation

    Glia

    (2013)
  • S. Boillee et al.

    Onset and progression in inherited ALS determined by motor neurons and microglia

    Science

    (2006)
  • M. Fricker et al.

    Primary phagocytosis of viable neurons by microglia activated with LPS or Abeta is dependent on calreticulin/LRP phagocytic signalling

    J. Neuroinflammation

    (2012)
  • J.J. Neher et al.

    Phagocytosis executes delayed neuronal death after focal brain ischemia

    Proc. Natl. Acad. Sci. USA

    (2013)
  • C.A. Colton

    Heterogeneity of microglial activation in the innate immune response in the brain

    J. Neuroimmune Pharmacol

    (2009)
  • K. Saijo et al.

    Microglial cell origin and phenotypes in health and disease

    Nat. Rev. Immunol.

    (2011)
  • Cited by (154)

    View all citing articles on Scopus
    View full text