Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2015 Nov 1.
Published in final edited form as: Free Radic Biol Med. 2014 Aug 1;0:34–46. doi: 10.1016/j.freeradbiomed.2014.07.033

NADPH Oxidase- and Mitochondria-derived Reactive Oxygen Species in Proinflammatory Microglial Activation: A Bipartisan Affair?

Evan A Bordt 1, Brian M Polster 1
PMCID: PMC4252610  NIHMSID: NIHMS618638  PMID: 25091898

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 (TNF-α), interleukin-1β (IL-1β), nitric oxide (NO), and reactive oxygen species 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.

Keywords: Reactive oxygen species, NADPH oxidase, mitochondria, MitoSOX, MitoTEMPO, inflammation, NLRP3, microglia, macrophage, MAPK, NF-kappaB, TNF-alpha, IL-1beta, nitric oxide, permeability transition pore

Introduction

Microglia are macrophage-like immune cells of the central nervous system which play major roles in both health and disease [1, 2]. In health, microglia are highly motile cells that survey their surrounding environment in order to maintain homeostasis [3], promote synaptogenesis, and regulate the integrity of the blood brain barrier [4, 5]. However, microglia become activated and proliferate following acute brain injury or in chronic neurodegenerative diseases such as Parkinson's disease and Alzheimer's 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's disease [3, 8-11] 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 following brain injuries via phagocytosis of live neurons [14-16]. 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 in order 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] and [20] for detailed reviews). Treatment with the bacterial endotoxin lipopolysaccharide (LPS) 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 Nox or Phox for phagocytic 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 [25-27], and induces nuclear factor kappa-B (NF-kB) translocation from the cytosol to the nucleus [28, 29]. Initiation of NF-KB-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 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.

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-kB activation and IL-1β transcription in the mouse microglial cell line BV2 in response to treatment with an Alzheimer's disease-relevant amyloid-beta (Aβ) peptide [32]. Additional experiments using the small molecule inhibitor DPI in primary rodent microglia implicated superoxide and downstream species originating from NADPH oxidase in the LPS-mediated induction of proinflammatory cytokines [33, 34]. However DPI is not a specific inhibitor of NADPH oxidase and can influence mitochondrial superoxide production, among additional targets [35-38]. Studies using microglial cultures from phagocytic oxidase (Phox) -/- mice lacking the gp91 catalytic subunit of NADPH oxidase conclusively established NADPH oxidase as the source of LPS-stimulated extracellular superoxide production [8]. Much but not all of the intracellular ROS elevation in response to LPS was also eliminated by gp91phox knockout [8]. Interestingly, while TNF-α secretion was diminished by gp91phox deficiency, the production of NO was unaltered. This finding contrasts with a dominant negative approach to NADPH oxidase inhibition. Stable mutant p47phox subunit overexpression to prevent functional NADPH oxidase assembly impaired both inducible nitric oxide synthesis (iNOS) induction and production of NO in rat HAPI microglial cells treated with LPS [34]. The role of the p47phox and gp91phox subunits of NADPH oxidase in microglial activation was further investigated by Choi et al. in vivo following intracerebroventricular injection of LPS or Aβ1-42 peptide [39]. Inhibition of NADPH oxidase by genetic knockout of p47phox or gp91phox or by the drug apocynin promoted microglial polarization toward an M2 anti-inflammatory phenotype while reducing release of pro-inflammatory mediators [39]. Knockout of p47phox decreased TNF-α but not IL-1β measured after intracerebroventricular injection of LPS. This finding is in agreement with the in vitro studies of Qin et al. [8] and Pawate et al. [34] showing that TNF-α secretion is dependent on NADPH oxidase activity but in contrast to the results of Kang et al. showing that Aβ25-35-induced IL-1β is also impaired when blocking NADPH oxidase using DPI [32]. Finally, Loane et al. demonstrated both in vitro and in vivo that metabotropic glutamate receptor 5 agonists attenuate microglial NO and TNF-α production by inhibiting the expression and activity of NADPH oxidase [40, 41]. Collectively, these studies implicate microglial NADPH oxidase activity in the transition to a proinflammatory phenotype. However, these studies also suggest that NO, TNF-α, and IL-1β may not always be regulated in tandem. Consequently, it is important to consider the markers used to define activation as well as the activating stimulus when evaluating microglial polarization.

Fig. 1.

Fig. 1

NADPH oxidase- and mitochondria-derived reactive oxygen species pathways in microglial activation. The antioxidants DPI, apocynin, NAC, MnTMPyP, gp91ds-tat peptide, and MitoTEMPO, decrease reactive oxygen species (i), leading to a decrease in release of proinflammatory factors. Exogenous catalase but not superoxide dismutase also blocks release, suggesting that superoxide (O2-) produced by NADPH oxidase requires dismutation to hydrogen peroxide (H2O2) before it can mediate intracellular signaling (ii). Cell permeable H2O2 activates the MAP kinase signaling pathway and induces NF-kB translocation from the cytosol to the nucleus (iii), where it promotes synthesis of TNF-α and iNOS, leading to increased secretion of TNF-α and NO. Evidence also exists that reactive oxygen species upregulate and activate the NLRP3 inflammasome, initiating release of the proinflammatory cytokines IL-1β and IL-18 from their pro-forms subsequent to cleavage by caspase-1 (iv). Induction of mitochondrial superoxide by rotenone, measurement of mitochondrial reactive oxygen species by MitoSOX, and scavenging of mitochondrial reactive oxygen species by MitoTEMPO are three techniques commonly used to implicate mitochondrial reactive oxygen species in NLRP3 inflammasome activation (v). DPI, diphenylene iodonium; IL-1β, interleukin-1β; iNOS, inducible nitric oxide synthase; MnTMPyP, Mn(III)tetrakis(1-methyl-4-pyridyl)porphyrin; NAC, N-acetylcysteine; NO, nitric oxide; NLRP3, NOD-like receptor family pyrin domain-containing 3; SOD, superoxide dismutase; TNF-α, tumor necrosis factor-α.

Superoxide produced by NADPH oxidase is predominantly extracellular (Fig. 1, ii). In vivo, extracellular superoxide dismutase 3 (SOD3) dismutates superoxide to form membrane permeable H2O2 [42]. In vitro, superoxide conversion to H2O2 can be enzymatically accelerated by addition of extracellular superoxide dismutase (SOD) while extracellular H2O2 derived from superoxide can be experimentally eliminated by catalase addition. The enzyme catalase degrades H2O2 to oxygen and water [43]. Interestingly, exogenous catalase, when added in combination with LPS/IFN-γ, impaired microglial release of proinflammatory mediators while SOD did not [34]. Similarly, catalase but not SOD prevented stimulation of microglial proliferation in response to IL-1β or TNF-α [44]. These results suggest that H2O2, not superoxide, is the primary ROS responsible for mediating microglial activation and proliferation in response to proinflammatory stimuli. It is important to note that arginase has been identified as a contaminant in some commercial catalase preparations [45]. As arginase degrades L-arginine, a precursor for NO synthesis, catalase purity is an important variable to consider, particularly when using NO as a marker for activation.

In addition to H2O2 removal, the effect of H2O2 addition on microglial phenotypes has been tested. Eguchi et al. demonstrated that H2O2 increased the LPS-induced expression of iNOS, responsible for production of NO, yet found that H2O2 alone was insufficient to upregulate iNOS [46]. Another recent study demonstrated that addition of H2O2 to LPS-treated microglial cells enhanced NO production [47]. Continuous H2O2 generation by two different ROS generating systems was able to stimulate microglial proliferation [44]. These studies, and observations that NAPDH oxidase inhibition impairs but does not entirely block the release of proinflammatory mediators such as TNF-α, suggest that while H2O2 in involved in neuroinflammation, it may act more so to intensify inflammatory processes, rather than as a direct inducer of such processes. In addition, it is possible that ROS downstream of H2O2, e.g. the reactive hydroxyl radical formed from H2O2 in the presence of iron [48], also contribute to the proinflammatory phenotype.

Importantly, H2O2 derived from NADPH oxidase-generated superoxide does not only regulate microglial activation in a cell autonomous manner. Zhang et al. described upregulation of neuronal NADPH oxidase following traumatic brain injury [49]. Inhibition of NADPH oxidase using apocynin or a gp91ds-tat peptide that competitively inhibits the p47phox binding site on gp91phox was neuroprotective and attenuated microglial activation following traumatic injury to the brain [49].

Reactive oxygen species, nitric oxide, and reactive microgliosis

Microglia in vitro continue to produce proinflammatory factors following removal of activating agent [47] and microglia in vivo frequently remain activated over long time periods following injury or onset of disease [50, 51]. This continued activation is likely 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 in their continued activation through reactive microgliosis. CD11b, the MAC1 receptor which binds LPS [22], Aβ peptide [52, 53], α-synuclein [54], and high mobility group box 1 (HMGB1) [55] among several ligands, is a beta-integrin microglial cell surface molecule which demonstrates increased expression during activation. LPS and immunogenic stimuli associated with neurodegenerative disorders upregulate microglial CD11b expression [56] and increased CD11b expression corresponds to the severity of microglial activation in a variety of neuroinflammatory diseases [57]. Like the ability of antioxidants to decrease production of proinflammatory mediators, Roy et al. found that the antioxidants N-acetylcysteine (NAC) and pyrrolidine dithiocarbamate (PDTC) prevented upregulation of CD11b in vitro and in vivo [56]. H2O2 alone, or a superoxide generating system consisting of hypoxanthine and xanthine oxidase, was sufficient to elevate CD11b expression in a catalase-sensitive fashion, implicating H2O2 in the CD11b increase [56]. Intriguingly, scavenging NO with 2-(4-carboxyphenyl)-4,4,5,5-tetramethylimidazoline-1-oxyl-3-oxide (cPTIO) was able to prevent CD11b upregulation, suggesting that H2O2 upregulates CD11b indirectly through NO [56, 58]. As CD11b/MAC1 was required for NADPH oxidase activation in response to LPS [22], α-synuclein [54], or HMGB1 released by inflamed microglia and/or degenerating neurons [55], CD11b upregulation by an H2O2 and NO-mediated pathway is likely a major contributor to reactive microgliosis, as seen in a small animal model of Parkinson's disease [59]. Importantly, although compelling data suggest that ROS and NO participate in a self-amplifying loop during microglial activation, not all studies support the involvement of NO in proinflammatory cytokine production. In contrast to a role for NO in CD11b induction, Yoshino et al. observed that TNF-α produced by LPS-stimulated microglia was suppressed by the antioxidant NAC, but not by scavenging NO with cPTIO [60]. In addition, 3-(4-morpholinyl)sydnonimine (SIN-1), a donor of superoxide and NO [61], induced microglial TNF-α production in an SOD-sensitive fashion, indicating that NO was incapable of stimulating TNF-α secretion in the absence of superoxide.

Although it is often convenient to think about microglial polarization in terms of linear pathways, microglial activation does not consist of a singular cell signaling cascade, and instead comprises a multitude of pathways that converge in the phenotype seen during excessive microglial activation. The activation phenotype undoubtedly depends on the proinflammatory trigger as well as the extracellular environment. As an interesting example, a transcriptional analysis recently found that microglia isolated from an ALS mouse model differed from LPS-activated microglia as well as from M1- or M2-polarized macrophages, including in the level of proinflammatory TNF-α and IL-1β expression [62]. When investigating the role of ROS, NO, and other factors in microglial activation, it is also important to consider the experimental conditions used to measure activation. Serum promotes binding of LPS to TLR4 due to the presence of lipopolysaccharide binding protein (LBP) [63, 64]. In vitro studies variably use serum-containing or serum-free conditions to examine LPS-induced activation of microglia, potentially altering the ratio of TLR4-dependent to TLR4-independent signaling mechanisms and the activation phenotype.

MAPK and NF-kB 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, both in non-microglial and microglial cell types, has focused on the MAPK signaling pathway and the downstream transcription factor NF-kB (Fig. 1, iii). Although most reports do not examine exactly how ROS increase MAPK phosphorylation during the activation process, oxidation of catalytic cysteines on MAPK-inactivating phosphatases may be involved [25-27]. A study in rat epithelial cells demonstrated that LPS treatment induces phosphorylation of the MAPKp38 subunit which was required for LPS-stimulated TNF-α production [29]. The antioxidant NAC was able to ablate p38 phosphorylation and TNF-α release while exposure to a superoxide-generating system or H2O2 was able to induce MAPK p38 phosphorylation [29]. In peripheral blood mononuclear cells, inhibition of superoxide production with DPI impaired translocation of the p50 subunit of NF-kB from the cytosol to the nucleus and subsequent production of cytokines IL-6 and IL-8 [65].

The importance of multiple MAPKs and NF-kB to the activation process specifically in microglial cells, and their regulation by ROS, has also been demonstrated through multiple approaches. Inhibition of p38, extracellular signal-related kinase (ERK)1/2, or c-Jun N-terminal kinase (JNK) in primary rat microglia impaired LPS- or Aβ1-42 stimulated production of IL-1β [66]. Similarly, treatment with the flavonoid antioxidant isoorientin or with NAC decreased LPS-induced MAPK activation, nuclear NF-kB translocation, and the production of IL-1β, TNF-α, and NO in BV2 microglial cells [67]. Multiple groups used the p47phox dominant negative approach discussed earlier to demonstrate a specific requirement for NADPH oxidase activity in p38 and ERK phosphorylation during proinflammatory microglial activation [34, 68]. Another study with BV2 cells demonstrated that fructose-1,6-bisphosphate, a glycolytic intermediate that preserves cellular antioxidant capacity through stimulation of the pentose phosphate pathway [69], decreased LPS-induced iNOS expression to a similar extent as NAC by inhibiting JNK and p38 MAPK phosphorylation [70]. The antioxidant protein peroxiredoxin I was recently identified as an endogenous negative regulator of NF-KB-mediated microglial activation [71], providing further support for the redox-sensitivity of NF-kB. Strikingly, constitutive activation of NF-kB in microglia in vivo achieved by genetic means was sufficient to induce microgliosis and neuronal death [13], confirming the central role of this transcription factor in classical M1 microglial activation.

Although most of the attention has focused on NF-kB as a redox-sensitive transcriptional regulator of microglial activation, other transcription factors are undoubtedly involved. The transcription factor p53 is a master regulator of cell-cycle control and apoptosis [72]. It responds to a variety of stressors, including oxidative damage. Inhibiting p53 in microglia using the small molecule pifithrin-α decreased LPS- or Aβ25-35-induced iNOS expression and ameliorated microglia-mediated neurotoxicity [73]. The involvement of p53 in microglial activation was also recently confirmed by genetic knockout. p53-/- microglia showed a decrease in production of proinflammatory mediators in response to IFN-γ, and displayed increased expression of genes associated with alternative activation [74]. Although it was hypothesized that p53 activation during microglial activation is induced by ROS [74], the necessity and/or sufficiency of ROS in p53 activation leading to microglial activation has yet to be established. Studies testing the effects of anti- and pro-oxidants on p53 in microglia with and without activating stimuli should prove enlightening. Finally, evidence suggests that Nuclear factor erythroid 2-related factor 2 (Nrf2), a transcriptional activator of genes regulated by antioxidant response elements (ARE), is a negative regulator of microglial activation in response to LPS [75] or the Parkinson's disease-inducing toxin 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) [76]. Drugs which activate the Nrf2 pathway such as sulforaphane [77] may mediate neuroprotection in part by preventing excessive M1 microglial polarization [75, 76, 78]. Figure 2 illustrates some of the main connections between H2O2 and proinflammatory microglial activation.

Fig 2.

Fig 2

Impact of reactive oxygen species on inflammatory pathways. Following an inflammatory stimulus (i.e. LPS, Aβ peptide), activation of NADPH oxidase and/or impairment of mitochondrial respiration results in elevated superoxide (O2-) and hydrogen peroxide (H2O2). These reactive oxygen species are implicated in NF-kB translocation to the nucleus, upregulation of CD11b/Mac1, p53 activation, and activation of the NLRP3 inflammasome. NO and TNF-α production occur subsequent to NF-kB activation. Cleavage of pro-IL-1β to the active form occurs subsequent to NLRP3 inflammasome activation. Reactive oxygen species produced by NADPH oxidase or mitochondria may also constitute an inflammatory stimulus in itself or lead to proinflammatory amplification. Aβ, amyloid-beta; IL-1β, interleukin-1β; LPS, lipopolysaccharide; NLRP3, NOD-like receptor family pyrin domain-containing 3; NO, nitric oxide; TNF-α, tumor necrosis factor-α.

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 cytoplasm. Although the preponderance of evidence supports the involvement of NADPH oxidase-derived H2O2 in microglial activation, the possibility that mitochondria-derived H2O2 contributes to the M1 transition under some scenarios has also been considered. For example, the direct microglial activation by the mitochondrial toxin rotenone observed by some was attributed to mitochondria-derived ROS [80, 81].

Rotenone inhibits complex I of the electron transport chain and stimulates mitochondrial superoxide production during the oxidation of complex I substrates [79]. Systemic rotenone administration recapitulates features of Parkinson's disease in rats, including highly selective nigrostriatal dopaminergic degeneration [82]. Early studies by Gao et al. using primary brain cell cultures suggested that microglia may contribute to rotenone-induced dopaminergic neurodegeneration [83, 84]. Low dose rotenone treatment (0.5-20 nM) did not cause significant neurotoxicity in neuron-enriched cultures, yet induced robust neurotoxicity in neuron/glia mixed cultures [83]. Although mitochondrial superoxide production is frequently cited as the link between rotenone and neurotoxicity [85, 86], inhibition of rotenone neurotoxicity by apocynin in mixed cultures suggested a surprising involvement of microglial NADPH oxidase [83, 84]. Low dose rotenone treatment (5-10 nM) elicited an increase in superoxide production by wild type microglia, but not by cells derived from gp91phox-deficient mice [84]. Furthermore, apocynin had no effect on superoxide levels or low dose rotenone neurotoxicity in Phox -/- mixed cultures, confirming the participation of microglial NAPDH oxidase in rotenone neurotoxicity [84]. Even more surprisingly, radiolabeled ligand binding assay revealed an affinity of rotenone for the catalytic gp91phox subunit of NADPH oxidase. Rotenone-gp91phox binding, which was inhibitable by DPI, induced membrane translocation of the p67phox subunit and led to NADPH oxidase-induced superoxide production [87].

Recent studies using BV2 mouse microglial cells found that rotenone treatment alone was sufficient to induce a proinflammatory activation phenotype as assessed by induction of iNOS, TNF-α, and IL-1β [80, 81]. Activation was mediated by ROS, p38 MAPK, and the NF-kB pathway [81]. Nevertheless, activation of microglia by rotenone has not always been recapitulated in primary microglial cultures ([83, 88, 89], but see [90] and [91]). The concentrations of rotenone employed in these studies varied over a 10,000-fold range (0.1 nM - 1 μM). Studies implicating rotenone in cytokine production by macrophages have ranged even higher (e.g. 5-40 μM [92, 93]). Toxicity due to rotenone has sometimes but not always been measured. This is important, as compromised cells release damage-associated molecular patterns which can promote microglial activation. As noted earlier, HMGB1 is one factor that can be released from damaged cells which propagates an activation phenotype [55]. Since ROS generally appear to augment rather than initiate activation, full microglial proinflammatory cytokine production in response to rotenone may require NAPDH oxidase- and/or mitochondria-derived ROS plus a second “hit” that is cell culture-condition or model-dependent. A rigorous establishment of dose-response relationships among rotenone-induced inhibition of mitochondrial oxygen consumption, superoxide production, toxicity, and activation markers is required and should help explain discrepant results. Nevertheless, reliance on mitochondrial inhibitors is an inherently flawed approach for determining the role of mitochondrial superoxide in microglial activation as it will likely prove impossible to completely separate out effects on mitochondrial superoxide generation from other effects on mitochondrial function. It is also generally not appreciated that rotenone, in contrast to the complex I inhibitor piericidin A [94], impairs microtubule assembly in the low nanomolar range [94-97]. A comparison of rotenone, piericidin A, and other, structurally distinct, complex I inhibitors should help untangle true contributions of mitochondrial superoxide/respiratory inhibition from effects on NADPH oxidase, microtubule polymerization, or other targets.

Importantly, in addition to ROS, altered mitochondrial and glycolytic energy metabolism were linked to BV2 microglial activation [98-100]. BV2 microglial cells significantly decreased mitochondrial oxygen consumption [98, 100] and increased glycolysis [98] upon stimulation with LPS or LPS plus IFN-γ. Consistent with other studies linking ROS to proinflammatory cytokine production, a combination of the antioxidants Trolox and edaravone or overexpression of the ROS-lowering protein Grp75/mtHsp70/mortalin was able to suppress NF-kB activation and release of TNF-α and IL-6 [98]. Inhibition of the mitochondrial ATP synthase by the drug oligomycin overcame the ability of antioxidants to attenuate secretion of these proinflammatory cytokines. Elevated glycolytic lactate production in the presence of oligomycin was suggested to be the crucial factor supplanting the effect of antioxidants on activation; sodium lactate (5 mM) mimicked the effect of oligomycin [98]. This study underscores the importance of considering effects on metabolism in addition to ROS when using mitochondrial inhibitors to modulate the activation phenotype.

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

Not long ago, Kasahara et al. 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 (ρ°) cells lacking mitochondria 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 glucose plus glucose oxidase partially rescued LPS-stimulated TNF-α secretion in ρ° cells while having no effect on their mitochondria-competent counterparts. In addition, NAC but not apocynin suppressed mitochondrial ROS levels and TNF-α secretion in LPS-treated RAW 264.7 cells, suggesting that at least in this particular cell line under the tested experimental conditions, mitochondria-derived superoxide—instead of or in addition to NADPH oxidase-derived superoxide—was required for proinflammatory cytokine production. While taking a similar approach, Nakahira et al. found that a different ρ° macrophage cell line, J774A.1, exhibited normal TNF-α secretion in response to LPS plus ATP (rather than LPS alone), but impaired IL-1β release ([93], discussed further below). Although compelling evidence as outlined above implicates ROS originating from NADPH oxidase in microglial activation, it would be interesting to test whether mitochondria-defective ρ° microglia exhibit impaired activation, suggesting a possible secondary contribution of mitochondria to the activation process.

Studies in macrophages have also indirectly implicated mitochondrial ROS in proinflammatory activation via knockout of the putative uncoupling protein UCP2. Data from multiple groups suggested that UCP2 -/- macrophages produce more mitochondrial ROS compared to wild type [102-105]. UCP2 -/-macrophages were more sensitive to induction of proinflammatory factors, with evidence for ROS-mediated protein tyrosine phosphatase inhibition [105], MAPK phosphorylation [104, 105], iNOS induction [103], and conflicting evidence on NF-kB involvement [103, 104]. In contrast to UCP2 knockout, UCP2 overexpression lowered intracellular ROS and attenuated induction of iNOS in response to LPS [106]. Rapid downregulation of UCP2 was observed in wild type macrophages stimulated with LPS; it was suggested that this decrease was necessary for a proinflammatory amplification loop mediated by mitochondrial ROS and resulting in accelerated p38 and ERK activation [104]. Consistent with this possibility, mitochondrial superoxide, as assessed by decreased activity of the superoxide-sensitive matrix enzyme aconitase, was basally elevated in the absence of UCP2, and UCP2 -/- macrophages failed to response to LPS with a further increase in mitochondrial ROS [104].

There is limited work on the involvement of UCP2 in microglial activation (as opposed to macrophage activation), however one study found an increase rather than a decrease in UCP2 mRNA in microglia following mouse ischemic brain injury [107]. Evidence from UCP2 knockout mice suggested that UCP2 may regulate mitochondrial uptake of the antioxidant glutathione rather than superoxide production directly [107]. While the role of UCP2 in mitochondrial ROS production is still debated, increasing evidence suggests that UCP2 does not even function as a proton transporter, instead likely acting as a C4 metabolite transporter [108-110]. Of note, de Bilbao et al. observed a steady-state upregulation of MnSOD in UCP2 mouse brain [107], indicating that secondary, potentially complicating changes to the proteome should be considered when using these constitutive knockout mice to assign protein function.

Mitochondrial superoxide and the NLRP3 inflammasome – a murky, yet intriguing story

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 NOD-like receptor family, pyrin domain-containing 3 (NLRP3, also called NALP3) inflammasome is a protein scaffolding complex that induces the secretion of the cytokines IL-1β and IL-18 from their pro-forms by activating caspase-1 (Fig. 1, iv) [111]. Present in most immune cells including microglia, the NLRP3 inflammasome is stimulated by diverse damage-associated molecular patterns and pathogen-associated molecular patterns, such as extracellular ATP, bacterial toxins, and particulate matter [111, 112]. In microglia, fibrillar or oligomeric Aβ peptide [113, 114], fibrillar α-synuclein [115], and a neurotoxic prion peptide [116] were all shown to induce NLRP3 inflammasome-dependent IL-1β secretion. Although regulation of IL-1β release by the inflammasome appears to be similar in macrophages and microglia, microglia, in contrast to macrophages, may regulate IL-18 release at least partly by an NLRP3-independent pathway [112].

ROS have been heavily implicated as an upstream event in the activation of the NLRP3 inflammasome [117]. For example, reduction of IL-1β production was observed following treatment with antioxidants such as NAC or DPI, while in some cases H2O2 alone was sufficient to induce NLRP3 activation [118-120]. Although NADPH oxidase-derived ROS were initially suggested to be important for NLRP3 inflammasome activation [121], mouse macrophages deficient in NADPH oxidase subunits gp91phox or gp22phox remained activation-competent [122, 123]. In addition, gp91phox-defective, gp47phox-defective, and gp22phox-defective macrophages from human patients with chronic granulomatous disease all exhibited active inflammasomes in response to multiple triggers [124].

In 2011, a series of three reports implicated ROS specifically derived from mitochondria in the activation of the NLRP3 inflammasome [92, 93, 123]. Additional reports, e.g. [111, 125-127], have surfaced since. Other reports have failed to replicate the necessity of ROS in NLRP3 inflammasome activation [128] or suggested that ROS are necessary only for an LPS “priming” step involving induction of NLRP3 and pro-IL-1β protein expression, but not for actual NLRP3-dependent caspase-1 activation [129]. In contrast, a rare study performed in microglial cells rather than peripheral immune cells found that the antioxidant NAC had no effect on LPS priming while inhibiting caspase-1 dependent IL-1β secretion in response to Aβ1-42 peptide oligomers [114].

MitoSOX and MitoTEMPO—not always mitochondria-specific

Three techniques have been used often but not exclusively to implicate ROS derived from mitochondria in NLRP3 inflammasome activation: a temporal correlation between activation and increased mitochondrial ROS measured using the mitochondrially-targeted fluorescent indicator MitoSOX, inhibition of NLRP3-dependent IL-1β secretion by a mitochondrially-targeted antioxidant, e.g. MitoTEMPO or MitoQ, and inflammasome activation or amplification by a superoxide-producing mitochondrial toxin such as rotenone (Fig. 1, v). While there is clearly an increase in intracellular ROS associated with NLRP3 activation, there are several complicating factors in the implementation of these techniques that for the most part have not been appreciated. In primary macrophages or macrophage cell lines, rotenone was reported to stimulate IL-1β release [92], augment LPS plus ATP-induced IL-1β release [93], or inhibit LPS plus ATP-induced IL-1β release [130]. Difficulties associated with using mitochondrial toxins such as rotenone were discussed above and will not be elaborated further here other than to note that the metabolic consequences of respiratory inhibition in addition to increased superoxide generation merit careful consideration. Regarding MitoSOX and mitochondrially-targeted antioxidants such as MitoTEMPO, there are serious concerns that limit their utility for assigning a mitochondrial origin to ROS during the activation process.

Conjugation to the lipophilic triphenylphosphonium cation (TPP+) is a strategy for targeting drugs and fluorescent probes to the negatively charged matrix of polarized mitochondria [131-133]. Unfortunately, once reagents are labeled as “mitochondria-specific,” there is a tendency to overlook the fact that the degree of mitochondrial targeting depends on the potential across the mitochondrial inner membrane (Fig. 3). MitoSOX (also called “mito-dihydroethidium” or “mito-hydroethidine”) accumulates in the mitochondrial matrix due to a TPP+ moiety conjugated to ROS-sensitive dihydroethidium [132]. When oxidized by superoxide, MitoSOX forms mito-2-hydroxyethidium that fluoresces red upon interaction with nucleic acids—predominantly DNA in the mitochondria matrix [132, 134-136]. MitoSOX oxidation by ROS other than superoxide yields additional oxidation products that also contribute to the red fluorescence detected in cells [137].

Fig. 3.

Fig. 3

MitoSOX and MitoTEMPO are not always mitochondria-specific. A positively charged lipophilic triphenylphosphonium cation (TPP+) conjugate within the reactive oxygen species-sensitive dye MitoSOX or the antioxidant MitoTEMPO mediates their accumulation in the matrix of functional mitochondria exhibiting negative membrane potential (AY). This charge-based accumulation results in several-fold higher concentrations of MitoSOX or MitoTEMPO in the mitochondrial matrix compared to the cytoplasm, with their relative concentrations indicated by font size within the figure (i). However, during the microglial/macrophage activation process, loss of δψ due to respiratory inhibition, mitochondrial uncoupling, or opening of the mitochondrial permeability transition pore (PTP) results in charge equilibration across the mitochondrial membrane, sometimes accompanied by mitochondrial swelling and loss of outer membrane integrity. Along with other unbound, charged molecules, MitoSOX and MitoTEMPO re-equilibrate across the mitochondrial membrane into the cytoplasm and their charge-based preferential sequestration in mitochondria is lost (ii). Thus, although they may still report or scavenge reactive oxygen species, respectively, their mitochondria-specific mechanism of action is no longer maintained.

Mitochondrial electron transport chain inhibitors such as rotenone and antimycin A depolarize mitochondria and cause MitoSOX redistribution to the cytoplasm and nucleus [136]. ATP synthase reversal maintains a partial negative membrane potential by pumping protons out of the matrix at the expense of ATP— for as long as ATP levels remain sufficient—however a correction for MitoSOX redistribution in response to electron transport chain inhibition is still required [136]. Mitochondrial respiratory inhibition occurs early in the NLRP3 inflammasome activation process in response to multiple triggers, inevitably causing mitochondrial depolarization [93, 111, 128, 138]. This depolarization will lead to partial to near complete redistribution of MitoSOX out of the matrix depending on the extent to which the negative potential is lost; thus ROS detected by total cellular MitoSOX fluorescence do not necessarily originate in mitochondria. Indeed, Voloboueva and colleagues noted that MitoSOX imaging needed to be performed within a short period of time in activated BV2 microglial cells due to a time-dependent migration of the fluorescent signal to the nucleus [98]. This caveat is of particular concern because MitoSOX fluorescence in immune cells is frequently quantified by flow cytometry [134], a method that does not allow for assessment of intracellular dye distribution. We suggest that fluorescent imaging for dye distribution, positive and negative controls, and confirmation of effects using secondary methods be implemented in future studies using this problematic dye.

Like MitoSOX, MitoTEMPO depends on a TPP+ conjugate to deliver the antioxidant to the matrix of polarized mitochondria [139]. During the initial development and characterization of MitoTEMPO as a mitochondria-specific ROS scavenger, addition of only 25 nM MitoTEMPO increased mitochondrial matrix superoxide dismutation three-fold without impacting the cytoplasmic dismutation of superoxide [140]. Although early studies employing MitoTEMPO used this scavenger in the 1 nM to 5 μM range, concentrations as high as 500 μM have been employed in immune cells, usually with limited dose-response analysis, no controls employing untargeted TEMPO or TPP+ alone, and no assessment of the impact of MitoTEMPO on nonmitochondrial sources of ROS. For example, two of the main studies implicating mitochondrial ROS in NLRP3 inflammasome activation found that 500 μM MitoTEMPO impaired NLRP3 inflammasome-dependent IL-1β secretion while 100 μM MitoTEMPO was relatively ineffective [93, 111]. Even provided mitochondria are still polarized during the activation process and initially accumulate mitochondrially-targeted antioxidant, these high concentrations of MitoTEMPO will undoubtedly disrupt mitochondrial function since as little as 1 μM of the closely related antioxidants Mito-TEMPOL or MitoQ caused respiratory inhibition in cells [140]. Consequently, the ability of 500 μM MitoTEMPO to inhibit IL-1β secretion likely has nothing to do with modulation of mitochondria-derived ROS. Additional studies on how MitoTEMPO influences specific reactive oxygen species within cells (e.g. superoxide, H2O2, etc.) and characterization of non-ROS targets are clearly needed. Unfortunately, non-pharmacological approaches, such as the overexpression of SOD or catalase enzymes targeted to various subcellular compartments, have been under-utilized when assessing the contribution of mitochondria-derived superoxide and H2O2 to inflammasome activation. However, the commercially available MCAT mouse (Strain name: B6.Cg-Tg(CAG-OTC/CAT)4033Prab/J), which transgenically overexpresses catalase in the mitochondrial matrix, was recently used to show that elevating mitochondrial H2O2 degrading activity did not in fact attenuate NLRP3 inflammasome activation in bone marrow-derived macrophages in response to the most commonly used activating trigger, LPS plus extracellular ATP, although it did in response to Salmonella mutants [141]. Therefore, it appears that mitochondria-derived H2O2 downstream of matrix superoxide is not a general requirement for NLRP3 inflammasome activation, but may contribute to activation in specific contexts.

NLRP3 and brain injury

Caveats aside, the possibility that mitochondria contribute to NLRP3 activation in response to at least some triggers remains intriguing and the NLRP3 inflammasome is emerging as a new target for neuroprotection. Intracerebral hemorrhage or injection of rotenone into the mouse brain basal ganglia induced NLRP3 expression which was mitigated by MitoTEMPO injection in vivo [142]. This finding is consistent with but does not prove the proposed involvement of ROS in NLRP3 inflammasome priming [129]. NLRP3 knockdown by siRNA reduced brain edema and improved neurological functions at 24-72 hours after intracerebral hemorrhage [142], suggesting a deleterious role for the NLRP3 inflammasome following acute brain injury. Interestingly, glibenclamide (also known as glyburide), a diabetes drug proposed as a novel treatment for acute brain injury due to its ability to inhibit the Sur1-Trpm4 channel involved in brain edema [143], was also shown to inhibit the NLRP3 inflammasome [144]. Similar to NLRP3 knockdown, glibenclamide reduced neuroinflammation and cognitive impairment following subarachnoid brain hemorrhage [145].

In addition to acute brain injury, the NLRP3 inflammasome is implicated in the pathogenesis of Alzheimer's disease [146]. An increase in the cleaved form of capase-1 was detected in post-mortem hippocampal and frontal cortex brain samples of patients with Alzheimer's disease relative to controls [147], implying increased inflammasome activity. Strikingly, knockout of either NLRP3 or caspase-1 ameliorated memory loss and β-amyloid plaque burden in an Alzheimer's disease transgenic mouse model [147]. Microglia in these mice were skewed toward an M2-like phenotype, displaying increased M2 markers such as arginase-1 and IL-4 concomitant to a reduction in the M1 marker iNOS. Surprisingly, production of the proinflammatory mediators TNF-α and NO in response to fibrillar Aβ peptide in microglia or macrophages depended on NLRP3 inflammasome-dependent IL-1β production, as shown using knockout cells for NLRP3, caspase-1, or the IL-1 receptor [113]. This TNF-α/NO dependence on the NLRP3 inflammasome in response to fibrillar Aβ is in contrast to studies performed in macrophages which indicate that the NLRP3 inflammasome is not required for TNF-α/NO secretion in response to most activators, e.g. a high concentration of LPS [113] or LPS plus ATP [93]. Data showing normal TNF-α but blunted IL-1β response following LPS plus ATP treatment of ρ° macrophage cells demonstrated that IL-1β but not TNF-α was influenced by the presence or absence of respiration-competent mitochondria [93]. Pathways controlling NLRP3 inflammasome activation in response to various triggers, including the necessity of mitochondrial superoxide generation and interaction with other proinflammatory changes such as NF-KB-dependent transcription, plainly require further elucidation. Parajuli et al. recently found that even fibrillar and oligomeric Aβ peptides activated the NLRP3 inflammasome through different pathways, additionally showing a small but significant contribution of NAPDH oxidase activity to oligomeric Aβ-induced IL-1β secretion by demonstrating suppression with a cell permeable gp91ds-tat peptide inhibitor of NADPH oxidase [114].

NLRP3 activation—is a mitochondrial signal required?

Even if one assumes that mitochondria-derived ROS participate in NLRP3 inflammasome activation, how also remains a contentious question. In macrophages, NLRP3 expression during the priming step depended on NF-kB [148]. Thus one possibility is that mitochondria-derived H2O2 participates in NLRP3 priming by amplifying MAPK signaling as discussed above (Fig. 1, iii). However, many studies suggest that ROS are necessary not only for inflammasome priming but also for activation. An early proposal was that ROS induce thioredoxin-interacting protein (TXNIP) dissociation from thioredoxin, allowing it to bind and activate NLRP3 [119]. However, a subsequent study failed to reproduce this finding [149]. A series of more recent studies raised the possibility that release of a mitochondrial signal is required to trigger activation of pro-caspase-1 by the NLRP3 inflammasome [93, 130, 138, 150], similar to apoptosome-mediated pro-caspase-9 activation that is triggered by the release of mitochondrial cytochrome c during apoptosis [151].

Nakahira et al. provided initial evidence that the release of mitochondrial DNA from the matrix to the cytoplasm subsequent to mitochondrial compromise is the NLRP3 trigger [93]. They and others showed that cyclosporin A, inhibitor of a large conductance mitochondrial inner membrane channel called the permeability transition pore (PTP), suppressed the release of IL-1β by activated macrophages [93, 130, 138]. The PTP is a ROS-sensitive channel that opens in response to the oxidation of thiols on pore components [152], including cysteine 203 of the cyclosporin A target cyclophilin D [153]. Therefore, in this model ROS would be acting in the mitochondrial matrix rather than the cytoplasm to initiate PTP-dependent release of mitochondrial DNA as an activating ligand of NLRP3. However, a just published study strongly challenged this model by demonstrating that knockout of cyclophilin D had no effect upon NLRP3 inflammasome activity while cyclosporin A impaired inflammasome activation equally well in wild type and cyclophilin D knockout macrophages [154]. Although it remains possible that mitochondrial DNA is released by mitochondrial damage independent of the classical cyclophlin D-dependent permeability transition pore, Allam et al. also found normal NLRP3 inflammasome activation in cells deficient in pro-apoptotic Bax and Bak or in cells overexpressing Bcl-2. These findings indicate that such mitochondrial membrane disruption would have to also occur independently of central mechanisms regulating mitochondrial permeability during apoptosis. Nevertheless, in an interesting twist, opening of the mitochondrial permeability transition pore was impaired in NLRP3 knockout macrophages [93], suggesting an upstream role for NLPR3 in mitochondrial damage, perhaps independent of the inflammasome or as part of a feedback loop. In a variation of the mitochondrial DNA efflux model, Shimada suggested that rather than simply cytoplasmic mitochondrial DNA, oxidized mitochondrial DNA was the specific trigger of inflammasome activation [138]. Thus, ROS were required both for the oxidation of mitochondrial DNA and for triggering a mitochondrial pore-dependent efflux to the cytoplasm. Subsequently, Iyer and colleagues discovered an NLRP3 inflammasome trigger, linezolid, that caused the release of IL-1β from LPS-primed macrophages in the absence of a detectable increase in mitochondrial ROS [130]. Cyclosporin A but not MitoTEMPO impaired IL-1β release. The mitochondria-specific inner membrane phospholipid cardiolipin was identified as a direct NLRP3 activating ligand, apparently without a requirement for mitochondrial ROS or mitochondrial DNA release [130]. Cardiolipin, along with microtubules, appeared to also be acquired for the recruitment of NLRP3 to mitochondria, where NLRP3 bound the adaptor protein Apoptosis-associated speck-like protein containing CARD (ASC) to form functional inflammasome complexes [130, 150].

Reactive oxygen species and NLRP3 activation—challenges and final thoughts

Overall, the evidence that mitochondria are involved in NLRP3 inflammasome activation in some way is intriguing. Nevertheless, a unifying model is needed to assimilate the interesting but not always consistent evidence implicating their participation. Challenging all of the work implicating mitochondria-derived ROS in inflammasome activation, Münoz-Planillo et al. found that pore-forming gramicidin, which collapses the plasma membrane sodium and potassium gradients, initiated NLRP3-dependent IL-1β release from macrophages prior to any mitochondrial dysfunction and without ROS production [128]. They failed to reproduce NLRP3 inflammasome activation by H2O2, rotenone, or the mitochondrial complex III inhibitor antimycin A [128]. Furthermore, several antioxidants including NAC did not inhibit LPS priming or gramicidin-triggered NLRP3 activation in their hands. In addition, Jabaut et al. found a poor correlation between the effect of mitochondria-targeted treatments on NLRP3 inflammasome activation and their effect on ROS production [127].

All tested NLRP3 inflammasome activators cause cytoplasmic potassium efflux [128, 155]. Münoz-Planillo et al. demonstrated that the removal of extracellular potassium alone was sufficient to cause activation of the NLRP3 inflammasome by causing a drop in cytoplasmic potassium below a critical threshold, in the apparent absence of mitochondrial ROS or a mitochondria-derived activating ligand [128].

In striking contrast to the stimulatory role proposed for mitochondrial matrix superoxide, evidence suggests that cytoplasmic superoxide is a negative regulator of NLRP3-dependent IL-1β secretion [156]. Macrophages deficient in the cytoplasmic Cu/Zn SOD enzyme (SOD1) exhibited impaired caspase-1 activation mediated by reversible oxidation and glutathionylation of cysteine residues on caspase-1 [156]. Thus, the spatial and temporal distribution of ROS may profoundly affect how they influence priming and/or activation of the NLRP3 inflammasome. Ever improving techniques to measure site-specific ROS production within individual intracellular compartments should help to unravel the many complexities of how these species regulate NLRP3 inflammasome activation.

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 myeloid origin, differences in resting environment (brain vs. periphery) have led to some genetic and phenotypic differences between microglia and macrophages. Utilizing direct RNA sequencing of microglia and macrophages isolated from the same mouse, Hickman et al. found substantial differences in the expression of transcripts between the two cells types, with microglia expressing higher levels of several genes associated with the sensing of endogenous activating ligands [158]. The interplay between NO and ROS was discussed above, and may account for important differences in the reaction of macrophages or microglia to proinflammatory stimuli. Intriguingly, a study by Toku et al. found that IFN-γ stimulated NO production in macrophages, but failed to elicit an NO response in microglia [159]. Chao et al. demonstrated that the flavonoid naringenin inhibited iNOS and cyclooxygenase-2 expression more effectively in macrophages than in microglia [160]. While macrophages share many characteristics with microglia, further study into the role of ROS, particularly mitochondrial superoxide, in the activation of microglia and its NLRP3 inflammasome need to be completed.

M1 vs. M2 activation

The majority of this review has focused upon 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. While M1 polarization may be beneficial immediately following brain injury by phagocytic clearing of debris, M2 polarization may help to resolve a neuroinflammatory response by “putting the breaks” on proinflammatory cytokine production before levels become harmful. IL-4 is often employed to initiate M2 polarization in cultured microglia and induces prototypic M2 markers such as arginase expression [89]. Although possible roles for mitochondrial dysfunction and ROS production in M2 activation have not yet been widely investigated, one study found that low, non-toxic doses of the mitochondrial electron transport chain inhibitors rotenone and 3-nitroproprionic acid (3-NP) impaired IL-4-induced M2 microglial activation in vitro, as assessed by arginase activity. IL-4 treatment also decreased LPS-induced production of proinflammatory factors, consistent with a role for anti-inflammatory IL-4 in checking the M1 response. Interestingly, addition of rotenone or 3-NP, to inhibit complex I or II of the mitochondrial electron transport chain, respectively, prevented IL-4 from decreasing LPS-induced proinflammatory cytokines [89]. As inhibition of the electron transport chain is likely to produce superoxide, it is possible that superoxide or downstream ROS plays an inhibitory role in alternative (M2) microglial activation; i.e. ROS may in part promote M1 activation by downregulating the M2 “breaks” on the M1 response. Microglia within p47phox deficient mice defective in NADPH-mediated superoxide production acquired an M2 rather than an M1 phenotype in response to LPS, consistent with an inhibitory role for ROS in M2 polarization [39]. Further studies on how mitochondrial dysfunction, ROS production, and NADPH oxidase interact following anti-inflammatory microglial stimulation are clearly needed before any conclusions on the precise role(s) of ROS production in M2 activation can be drawn.

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, while even more demonstrate that antioxidants impair activation. However, concerns exist with the methodology used to quantify and modulate ROS, particularly superoxide of mitochondrial origin. It is noteworthy that flawed approaches such as the use of non-specific inhibitors have been overwhelmingly used to implicate mitochondrial ROS in microglial or macrophage activation whereas the involvement of NADPH-derived superoxide and H2O2 is supported by in vitro and in vivo gene knockout and dominant negative experiments. The recent demonstration that inhibition of NLRP3 inflammasome activation by cyclosporin A is not due to inhibition of cyclophilin D-dependent mitochondrial permeability transition pore opening refutes a major line of evidence linking mitochondrial superoxide-induced pore formation to IL-1β production [154]. Nevertheless, as NADPH oxidase knockout cells still produce proinflammatory factors such as IL-1β, it is important that additional studies are performed on the sources and species of ROS implicated in microglial activation, particularly with regard to the NLRP3 inflammasome. Modification of mitochondrial ROS in microglia by ectopic alteration of MnSOD and catalase expression should help in this regard.

Inflammasome-independent functions for NLRP3 is also an emerging area of investigation that may impact brain disorders such as Alzheimer's disease and intracerebral hemorrhage where aberrant NLRP3 expression is implicated. In addition to a possible upstream role for NLRP3 in mitochondrial permeability transition pore opening [93], new evidence suggests that NLRP3 can increase ROS production independent of inflammasome activation [162]. As the permeability transition pore is a ROS-sensitive channel [152], NLRP3 may indirectly activate pore opening by elevating mitochondrial matrix superoxide. Establishment of the subcellular localization of NLRP3 and its proximity to the electron transport chain will be crucial to understanding if and how it regulates mitochondrial and/or cellular ROS production. It is also essential that more microglia-specific studies are performed, particularly with regard to NLRP3 inflammasome activation, as the biochemical events associated with changes in ROS and M1 polarization may not always be identical in macrophages and microglial cells.

Finally, we would be remiss if we did not mention that NADPH oxidase enzymes and mitochondria are not the exclusive producers of ROS in activated microglia. Cyclooxygenase enzymes and nitric oxide synthase [163] are but two of several potential additional culprits.

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.

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.

Abbreviations

amyloid-beta

ARE

antioxidant response element

cPTIO

2-(4-Carboxyphenyl)-4,4,5,5-tetramethylimidazoline-1-oxyl-3-oxide

DPI

diphenylene iodonium

ERK

extracellular signal-related kinase

H2O2

hydrogen peroxide

HMGB1

high mobility group box 1

IFN-γ

interferon-γ

IL

interleukin

iNOS

inducible nitric oxide synthase

JNK

c-Jun N-terminal kinase

LPS

lipopolysaccharide

MAC1

macrophage antigen complex 1

MAPK

mitogen-activated protein kinase

δψ

mitochondrial membrane potential

NAC

N-acetylcysteine

NF-κB

nuclear factor kappa-B

NLRP3

NOD-like receptor family pyrin domain-containing 3

NO

nitric oxide

Nox

NADPH oxidase

Nrf2

Nuclear factor erythroid 2-related factor 2

PDTC

pyrrolidine dithiocarbamate

Phox

phagocyte oxidase

PTP

permeability transition pore

ρ°

rho zero

ROS

reactive oxygen species

SOD

superoxide dismutase

TLR

toll-like receptor

TNF-α

tumor necrosis factor- α

TPP+

triphenylphosphonium cation

Footnotes

Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final citable form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

References

  • 1.Kreutzberg GW. Microglia: a sensor for pathological events in the CNS. Trends Neurosci. 1996;19:312–8. doi: 10.1016/0166-2236(96)10049-7. [DOI] [PubMed] [Google Scholar]
  • 2.Schwarz JM, Bilbo SD. Sex, glia, and development: interactions in health and disease. Horm Behav. 2012;62:243–53. doi: 10.1016/j.yhbeh.2012.02.018. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Davis DS, Carson MJ. An Introduction to CNS-Resident Microglia: Definitions, Assays, and Functional Roles in Health and Disease. In: Cui C, Grandison L, Noronha A, editors. Neural-Immune Interactions in Brain Function and Alcohol Disorders. New York: Springer Press; 2013. pp. 3–29. [Google Scholar]
  • 4.Tremblay ME, Stevens B, Sierra A, Wake H, Bessis A, Nimmerjahn A. The role of microglia in the healthy brain. J Neurosci. 2011;31:16064–9. doi: 10.1523/JNEUROSCI.4158-11.2011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Horvath RJ, Deleo JA. Differential migration, LPS-induced cytokine, chemokine, and NO expression in immortalized BV-2 and HAPI cell lines and primary microglial cultures. J Neurochem. 2008 doi: 10.1111/j.1471-4159.2008.05633.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Lull ME, Block ML. Microglial activation and chronic neurodegeneration. Neurotherapeutics. 2010;7:354–65. doi: 10.1016/j.nurt.2010.05.014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Block ML, Hong JS. Microglia and inflammation-mediated neurodegeneration: multiple triggers with a common mechanism. Prog Neurobiol. 2005;76:77–98. doi: 10.1016/j.pneurobio.2005.06.004. [DOI] [PubMed] [Google Scholar]
  • 8.Qin L, Liu Y, Wang T, Wei SJ, Block ML, Wilson B, et al. NADPH oxidase mediates lipopolysaccharide-induced neurotoxicity and proinflammatory gene expression in activated microglia. J Biol Chem. 2004;279:1415–21. doi: 10.1074/jbc.M307657200. [DOI] [PubMed] [Google Scholar]
  • 9.Block ML, Zecca L, Hong JS. Microglia-mediated neurotoxicity: uncovering the molecular mechanisms. Nat Rev Neurosci. 2007;8:57–69. doi: 10.1038/nrn2038. [DOI] [PubMed] [Google Scholar]
  • 10.Wang T, Liu B, Qin L, Wilson B, Hong JS. Protective effect of the SOD/catalase mimetic MnTMPyP on inflammation-mediated dopaminergic neurodegeneration in mesencephalic neuronal-glial cultures. J Neuroimmunol. 2013;147:68–72. doi: 10.1016/j.jneuroim.2003.10.040. [DOI] [PubMed] [Google Scholar]
  • 11.Cunningham C. Microglia and neurodegeneration: the role of systemic inflammation. Glia. 2013;61:71–90. doi: 10.1002/glia.22350. [DOI] [PubMed] [Google Scholar]
  • 12.Boillee S, Yamanaka K, Lobsiger CS, Copeland NG, Jenkins NA, Kassiotis G, et al. Onset and progression in inherited ALS determined by motor neurons and microglia. Science. 2006;312:1389–92. doi: 10.1126/science.1123511. [DOI] [PubMed] [Google Scholar]
  • 13.Frakes AE, Ferraiuolo L, Haidet-Phillips AM, Schmelzer L, Braun L, Miranda CJ, et al. Microglia induce motor neuron death via the classical NF-kappaB pathway in amyotrophic lateral sclerosis. Neuron. 2014;81:1009–23. doi: 10.1016/j.neuron.2014.01.013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Neniskyte U, Neher JJ, Brown GC. Neuronal death induced by nanomolar amyloid beta is mediated by primary phagocytosis of neurons by microglia. J Biol Chem. 2011;286:39904–13. doi: 10.1074/jbc.M111.267583. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Fricker M, Oliva-Martin MJ, Brown GC. Primary phagocytosis of viable neurons by microglia activated with LPS or Abeta is dependent on calreticulin/LRP phagocytic signalling. J Neuroinflammation. 2012;9:196. doi: 10.1186/1742-2094-9-196. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Neher JJ, Emmrich JV, Fricker M, Mander PK, Thery C, Brown GC. Phagocytosis executes delayed neuronal death after focal brain ischemia. Proc Natl Acad Sci U S A. 2013;110:E4098–E4107. doi: 10.1073/pnas.1308679110. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Colton CA. Heterogeneity of microglial activation in the innate immune response in the brain. J Neuroimmune Pharmacol. 2009;4:399–418. doi: 10.1007/s11481-009-9164-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Kumar A, Loane DJ. Neuroinflammation after traumatic brain injury: opportunities for therapeutic intervention. Brain Behav Immun. 2012;26:1191–201. doi: 10.1016/j.bbi.2012.06.008. [DOI] [PubMed] [Google Scholar]
  • 19.Kumar A, Stoica BA, Sabirzhanov B, Burns MP, Faden AI, Loane DJ. Traumatic brain injury in aged animals increases lesion size and chronically alters microglial/macrophage classical and alternative activation states. Neurobiol Aging. 2013;34:1397–411. doi: 10.1016/j.neurobiolaging.2012.11.013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Saijo K, Glass CK. Microglial cell origin and phenotypes in health and disease. Nat Rev Immunol. 2011;11:775–87. doi: 10.1038/nri3086. [DOI] [PubMed] [Google Scholar]
  • 21.Wilkinson K, El KJ. Microglial scavenger receptors and their roles in the pathogenesis of Alzheimer's disease. Int J Alzheimers Dis. 2012;2012:489456. doi: 10.1155/2012/489456. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Pei Z, Pang H, Qian L, Yang S, Wang T, Zhang W, et al. MAC1 mediates LPS-induced production of superoxide by microglia: the role of pattern recognition receptors in dopaminergic neurotoxicity. Glia. 2007;55:1362–73. doi: 10.1002/glia.20545. [DOI] [PubMed] [Google Scholar]
  • 23.Palmer HJ, Paulson KE. Reactive oxygen species and antioxidants in signal transduction and gene expression. Nutr Rev. 1997;55:353–61. doi: 10.1111/j.1753-4887.1997.tb01561.x. [DOI] [PubMed] [Google Scholar]
  • 24.Lander HM. An essential role for free radicals and derived species in signal transduction. FASEB J. 1997;11:118–24. [PubMed] [Google Scholar]
  • 25.Hecht D, Zick Y. Selective inhibition of protein tyrosine phosphatase activities by H2O2 and vanadate in vitro. Biochem Biophys Res Commun. 1992;188:773–9. doi: 10.1016/0006-291x(92)91123-8. [DOI] [PubMed] [Google Scholar]
  • 26.Denu JM, Tanner KG. Specific and reversible inactivation of protein tyrosine phosphatases by hydrogen peroxide: evidence for a sulfenic acid intermediate and implications for redox regulation. Biochemistry. 1998;37:5633–42. doi: 10.1021/bi973035t. [DOI] [PubMed] [Google Scholar]
  • 27.Kamata H, Honda S, Maeda S, Chang L, Hirata H, Karin M. Reactive oxygen species promote TNFalpha-induced death and sustained JNK activation by inhibiting MAP kinase phosphatases. Cell. 2005;120:649–61. doi: 10.1016/j.cell.2004.12.041. [DOI] [PubMed] [Google Scholar]
  • 28.Martindale JL, Holbrook NJ. Cellular response to oxidative stress: signaling for suicide and survival. J Cell Physiol. 2002;192:1–15. doi: 10.1002/jcp.10119. [DOI] [PubMed] [Google Scholar]
  • 29.Haddad JJ, Land SC. Redox/ROS regulation of lipopolysaccharide-induced mitogen-activated protein kinase (MAPK) activation and MAPK-mediated TNF-alpha biosynthesis. Br J Pharmacol. 2002;135:520–36. doi: 10.1038/sj.bjp.0704467. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Zeng KW, Zhang T, Fu H, Liu GX, Wang XM. Schisandrin B exerts anti-neuroinflammatory activity by inhibiting the Toll-like receptor 4-dependent MyD88/IKK/NF-?B signaling pathway in lipopolysaccharide-induced microglia. Eur J Pharmacol. 2012;692:29–37. doi: 10.1016/j.ejphar.2012.05.030. [DOI] [PubMed] [Google Scholar]
  • 31.Zheng W, Zheng X, Liu S, Ouyang H, Levitt RC, Candiotti KA, et al. TNF? and IL-1? are mediated by both TLR4 and Nod1 pathways in the cultured HAPI cells stimulated by LPS. Biochem Biophys Res Commun. 2012;420:762–7. doi: 10.1016/j.bbrc.2012.03.068. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Kang J, Park EJ, Jou I, Kim JH, Joe EH. Reactive oxygen species mediate A beta(25-35)-induced activation of BV-2 microglia. Neuroreport. 2001;12:1449–52. doi: 10.1097/00001756-200105250-00030. [DOI] [PubMed] [Google Scholar]
  • 33.Qin L, Li G, Qian X, Liu Y, Wu X, Liu B, et al. Interactive role of the toll-like receptor 4 and reactive oxygen species in LPS-induced microglia activation. Glia. 2005;52:78–84. doi: 10.1002/glia.20225. [DOI] [PubMed] [Google Scholar]
  • 34.Pawate S, Shen Q, Fan F, Bhat NR. Redox regulation of glial inflammatory response to lipopolysaccharide and interferongamma. J Neurosci Res. 2004;77:540–51. doi: 10.1002/jnr.20180. [DOI] [PubMed] [Google Scholar]
  • 35.Aldieri E, Riganti C, Polimeni M, Gazzano E, Lussiana C, Campia I, et al. Classical inhibitors of NOX NAD(P)H oxidases are not specific. Curr Drug Metab. 2008;9:686–96. doi: 10.2174/138920008786049285. [DOI] [PubMed] [Google Scholar]
  • 36.Majander A, Finel M, Wikstrom M. Diphenyleneiodonium inhibits reduction of iron-sulfur clusters in the mitochondrial NADH-ubiquinone oxidoreductase (Complex I) J Biol Chem. 1994;269:21037–42. [PubMed] [Google Scholar]
  • 37.Li Y, Trush MA. Diphenyleneiodonium, an NAD(P)H oxidase inhibitor, also potently inhibits mitochondrial reactive oxygen species production. Biochem Biophys Res Commun. 1998;253:295–9. doi: 10.1006/bbrc.1998.9729. [DOI] [PubMed] [Google Scholar]
  • 38.Lambert AJ, Buckingham JA, Boysen HM, Brand MD. Diphenyleneiodonium acutely inhibits reactive oxygen species production by mitochondrial complex I during reverse, but not forward electron transport. Biochim Biophys Acta. 2008;1777:397–403. doi: 10.1016/j.bbabio.2008.03.005. [DOI] [PubMed] [Google Scholar]
  • 39.Choi SH, Aid S, Kim HW, Jackson SH, Bosetti F. Inhibition of NADPH oxidase promotes alternative and anti-inflammatory microglial activation during neuroinflammation. J Neurochem. 2012;120:292–301. doi: 10.1111/j.1471-4159.2011.07572.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Loane DJ, Stoica BA, Pajoohesh-Ganji A, Byrnes KR, Faden AI. Activation of metabotropic glutamate receptor 5 modulates microglial reactivity and neurotoxicity by inhibiting NADPH oxidase. J Biol Chem. 2009;284:15629–39. doi: 10.1074/jbc.M806139200. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Loane DJ, Stoica BA, Byrnes KR, Jeong W, Faden AI. Activation of mGluR5 and inhibition of NADPH oxidase improves functional recovery after traumatic brain injury. J Neurotrauma. 2013;30:403–12. doi: 10.1089/neu.2012.2589. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Fukai T, Folz RJ, Landmesser U, Harrison DG. Extracellular superoxide dismutase and cardiovascular disease. Cardiovasc Res. 2002;55:239–49. doi: 10.1016/s0008-6363(02)00328-0. [DOI] [PubMed] [Google Scholar]
  • 43.Melov S, Ravenscroft J, Malik S, Gill MS, Walker DW, Clayton PE, et al. Extension of life-span with superoxide dismutase/catalase mimetics. Science. 2000;289:1567–9. doi: 10.1126/science.289.5484.1567. [DOI] [PubMed] [Google Scholar]
  • 44.Mander PK, Jekabsone A, Brown GC. Microglia proliferation is regulated by hydrogen peroxide from NADPH oxidase. J Immunol. 2006;176:1046–52. doi: 10.4049/jimmunol.176.2.1046. [DOI] [PubMed] [Google Scholar]
  • 45.Esch F, Lin KI, Hills A, Zaman K, Baraban JM, Chatterjee S, et al. Purification of a multipotent antideath activity from bovine liver and its identification as arginase: nitric oxide-independent inhibition of neuronal apoptosis. J Neurosci. 1998;18:4083–95. doi: 10.1523/JNEUROSCI.18-11-04083.1998. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Eguchi H, Fujiwara N, Sakiyama H, Yoshihara D, Suzuki K. Hydrogen peroxide enhances LPS-induced nitric oxide production via the expression of interferon beta in BV-2 microglial cells. Neurosci Lett. 2011;494:29–33. doi: 10.1016/j.neulet.2011.02.047. [DOI] [PubMed] [Google Scholar]
  • 47.Lijia Z, Zhao S, Wang X, Yang J. A self-propelling cycle mediated by reactive oxide species and nitric oxide exists in LPS-activated microglia. Neurochem International. 2012 doi: 10.1016/j.neuint.2012.09.002. [DOI] [PubMed] [Google Scholar]
  • 48.Winterbourn CC. Toxicity of iron and hydrogen peroxide: the Fenton reaction. Toxicol Lett. 1995;82-83:969–74. doi: 10.1016/0378-4274(95)03532-x. [DOI] [PubMed] [Google Scholar]
  • 49.Zhang QG, Laird MD, Han D, Nguyen K, Scott E, Dong Y, et al. Critical role of NADPH oxidase in neuronal oxidative damage and microglia activation following traumatic brain injury. PLoS One. 2012;7:e34504. doi: 10.1371/journal.pone.0034504. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Kumar A, Loane DJ. Neuroinflammation after traumatic brain injury: opportunities for therapeutic intervention. Brain Behav Immun. 2012;26:1191–201. doi: 10.1016/j.bbi.2012.06.008. [DOI] [PubMed] [Google Scholar]
  • 51.Loane DJ, Kumar A, Stoica BA, Cabatbat R, Faden AI. Progressive neurodegeneration after experimental brain trauma: association with chronic microglial activation. J Neuropathol Exp Neurol. 2014;73:14–29. doi: 10.1097/NEN.0000000000000021. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Goodwin JL, Kehrli ME, Jr, Uemura E. Integrin Mac-1 and beta-amyloid in microglial release of nitric oxide. Brain Res. 1997;768:279–86. doi: 10.1016/s0006-8993(97)00653-7. [DOI] [PubMed] [Google Scholar]
  • 53.Zhang D, Hu X, Qian L, Chen SH, Zhou H, Wilson B, et al. Microglial MAC1 receptor and PI3K are essential in mediating beta-amyloid peptide-induced microglial activation and subsequent neurotoxicity. J Neuroinflammation. 2011;8:3. doi: 10.1186/1742-2094-8-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Zhang W, Dallas S, Zhang D, Guo JP, Pang H, Wilson B, et al. Microglial PHOX and Mac-1 are essential to the enhanced dopaminergic neurodegeneration elicited by A30P and A53T mutant alpha-synuclein. Glia. 2007;55:1178–88. doi: 10.1002/glia.20532. [DOI] [PubMed] [Google Scholar]
  • 55.Gao HM, Zhou H, Zhang F, Wilson BC, Kam W, Hong JS. HMGB1 acts on microglia Mac1 to mediate chronic neuroinflammation that drives progressive neurodegeneration. J Neurosci. 2011;31:1081–92. doi: 10.1523/JNEUROSCI.3732-10.2011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Roy A, Jana A, Yatish K, Freidt MB, Fung YK, Martinson JA, et al. Reactive oxygen species up-regulate CD11b in microglia via nitric oxide: Implications for neurodegenerative diseases. Free Radic Biol Me. 2008;45:686–99. doi: 10.1016/j.freeradbiomed.2008.05.026. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Akiyama H, McGeer PL. Brain microglia constitutively express beta-2 integrins. J Neuroimmunol. 1990;30:81–93. doi: 10.1016/0165-5728(90)90055-r. [DOI] [PubMed] [Google Scholar]
  • 58.Roy A, Fung YK, Liu X, Pahan K. Up-regulation of microglial CD11b expression by nitric oxide. J Biol Chem. 2006;281:14971–80. doi: 10.1074/jbc.M600236200. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Hu X, Zhang D, Pang H, Caudle WM, Li Y, Gao H, et al. Macrophage antigen complex-1 mediates reactive microgliosis and progressive dopaminergic neurodegeneration in the MPTP model of Parkinson's disease. J Immunol. 2008;181:7194–204. doi: 10.4049/jimmunol.181.10.7194. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Yoshino Y, Yamamoto S, Kohsaka S, Oshiro S, Nakajima K. Superoxide anion contributes to the induction of tumor necrosis factor alpha (TNF?) through activation of the MKK3/6-p38 MAPK cascade in rat microglia. Brain Res. 2011;8:1–12. doi: 10.1016/j.brainres.2011.09.009. [DOI] [PubMed] [Google Scholar]
  • 61.Hogg N, Darley-Usmar VM, Wilson MT, Moncada S. Production of hydroxyl radicals from the simultaneous generation of superoxide and nitric oxide. Biochem J. 1992;281:419–24. doi: 10.1042/bj2810419. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Chiu IM, Morimoto ET, Goodarzi H, Liao JT, O'Keeffe S, Phatnani HP, et al. A neurodegeneration-specific gene-expression signature of acutely isolated microglia from an amyotrophic lateral sclerosis mouse model. Cell Rep. 2013;4:385–401. doi: 10.1016/j.celrep.2013.06.018. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Schutt C, Schilling T, Kruger C. sCD14 prevents endotoxin inducible oxidative burst response of human monocytes. Allerg Immunol (Leipz) 1991;37:159–64. [PubMed] [Google Scholar]
  • 64.Fan MH, Klein RD, Steinstraesser L, Merry AC, Nemzek JA, Remick DG, et al. An essential role for lipopolysaccharide-binding protein in pulmonary innate immune responses. Shock. 2002;18:248–54. doi: 10.1097/00024382-200209000-00008. [DOI] [PubMed] [Google Scholar]
  • 65.Simard JC, Cesaro A, Chapeton-Montes J, Tardif M, Antoine F, Girard D, et al. S100A8 and S100A9 induce cytokine expression and regulate the NLRP3 inflammasome via ROS-dependent activation of NF-?B(1.) PLoS One. 2013;8:e72138. doi: 10.1371/journal.pone.0072138. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Kim SH, Smith CJ, Van Eldik LJ. Importance of MAPK pathways for microglial pro-inflammatory cytokine IL-1 beta production. Neurobiol Aging. 2004;25:431–9. doi: 10.1016/S0197-4580(03)00126-X. [DOI] [PubMed] [Google Scholar]
  • 67.Yuan L, Wu Y, Ren X, Liu Q, Wang J, Liu X. Isoorientin attenuates lipopolysaccharide-induced proinflammatory responses through down-regulation of ROS-related MAPK/NF-?B signaling pathway in BV-2 microglia. Mol Cell Biochem. 2013 doi: 10.1007/s11010-013-1854-9. [DOI] [PubMed] [Google Scholar]
  • 68.Yang CS, Lee HM, Lee JY, Kim JA, Lee SJ, Shin DM, et al. Reactive oxygen species and p47phox activation are essential for the Mycobacterium tuberculosis-induced pro-inflammatory response in murine microglia. J Neuroinflammation. 2007;4:27. doi: 10.1186/1742-2094-4-27. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Vexler ZS, Wong A, Francisco C, Manabat C, Christen S, Tauber M, et al. Fructose-1,6-bisphosphate preserves intracellular glutathione and protects cortical neurons against oxidative stress. Brain Res. 2003;960:90–8. doi: 10.1016/s0006-8993(02)03777-0. [DOI] [PubMed] [Google Scholar]
  • 70.Kim YC, Park TY, Baik E, Lee SH. Fructose-1,6-bisphosphate attenuates induction of nitric oxide synthase in microglia stimulated with lipopolysaccharide. Life Sci. 2012;90:365–72. doi: 10.1016/j.lfs.2011.12.011. [DOI] [PubMed] [Google Scholar]
  • 71.Kim SU, Park YH, Min JS, Sun HN, Han YH, Hua JM, et al. Peroxiredoxin I is a ROS/p38 MAPK-dependent inducible antioxidant that regulates NF-?B-mediated iNOS induction and microglial activation. J Neuroimmunol. 2013;259:26–36. doi: 10.1016/j.jneuroim.2013.03.006. [DOI] [PubMed] [Google Scholar]
  • 72.Kastan MB, Canman CE, Leonard CJ. P53, cell cycle control and apoptosis: implications for cancer. Cancer Metastasis Rev. 1995;14:3–15. doi: 10.1007/BF00690207. [DOI] [PubMed] [Google Scholar]
  • 73.Davenport CM, Sevastou IG, Hooper C, Pocock JM. Inhibiting p53 pathways in microglia attenuates microglial-evoked neurotoxicity following exposure to Alzheimer peptides. J Neurochem. 2010;112:552–63. doi: 10.1111/j.1471-4159.2009.06485.x. [DOI] [PubMed] [Google Scholar]
  • 74.Jayadev S, Nesser NK, Hopkins S, Myers SJ, Case A, Lee RJ, et al. Transcription factor p53 influences microglial activation phenotype. Glia. 2011;59:1402–13. doi: 10.1002/glia.21178. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Innamorato NG, Rojo AI, Garcia-Yague AJ, Yamamoto M, de Ceballos ML, Cuadrado A. The transcription factor Nrf2 is a therapeutic target against brain inflammation. J Immunol. 2008;181:680–9. doi: 10.4049/jimmunol.181.1.680. [DOI] [PubMed] [Google Scholar]
  • 76.Rojo AI, Innamorato NG, Martin-Moreno AM, de Ceballos ML, Yamamoto M, Cuadrado A. Nrf2 regulates microglial dynamics and neuroinflammation in experimental Parkinson's disease. Glia. 2010;58:588–98. doi: 10.1002/glia.20947. [DOI] [PubMed] [Google Scholar]
  • 77.Greco T, Fiskum G. Neuroprotection through stimulation of mitochondrial antioxidant protein expression. J Alzheimers Dis. 2010;20(Suppl 2):S427–S437. doi: 10.3233/JAD-2010-100519. [DOI] [PubMed] [Google Scholar]
  • 78.Brandenburg LO, Kipp M, Lucius R, Pufe T, Wruck CJ. Sulforaphane suppresses LPS-induced inflammation in primary rat microglia. Inflamm Res. 2010;59:443–50. doi: 10.1007/s00011-009-0116-5. [DOI] [PubMed] [Google Scholar]
  • 79.Andreyev AY, Kushnareva YE, Starkov AA. Mitochondrial metabolism of reactive oxygen species. Biochemistry (Mosc) 2005;70:200–14. doi: 10.1007/s10541-005-0102-7. [DOI] [PubMed] [Google Scholar]
  • 80.Yuan YH, Sun JD, Wu MM, Hu JF, Peng SY, Chen NH. Rotenone could activate microglia through NF-kB associated pathway. Neurochem Res. 2013;38:1553–60. doi: 10.1007/s11064-013-1055-7. [DOI] [PubMed] [Google Scholar]
  • 81.Gao F, Chen D, Hu Q, Wang G. Rotenone directly induces BV2 cell activation via the p38 MAPK pathway. PLoS One. 2013;8:e72046. doi: 10.1371/journal.pone.0072046. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82.Betarbet R, Sherer TB, MacKenzie G, Garcia-Osuna M, Panov AV, Greenamyre JT. Chronic systemic pesticide exposure reproduces features of Parkinson's disease. Nat Neurosci. 2000;3:1301–6. doi: 10.1038/81834. [DOI] [PubMed] [Google Scholar]
  • 83.Gao HM, Hong JS, Zhang W, Liu B. Distinct role for microglia in rotenone-induced degeneration of dopaminergic neurons. J Neurosci. 2002;22:782–90. doi: 10.1523/JNEUROSCI.22-03-00782.2002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.Gao HM, Liu B, Hong JS. Critical role for microglial NADPH oxidase in rotenone-induced degeneration of dopaminergic neurons. J Neurosci. 2003;23:6181–7. doi: 10.1523/JNEUROSCI.23-15-06181.2003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.Sherer TB, Betarbet R, Testa CM, Seo BB, Richardson JR, Kim JH, et al. Mechanism of toxicity in rotenone models of Parkinson's disease. J Neurosci. 2003;23:10756–64. doi: 10.1523/JNEUROSCI.23-34-10756.2003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86.Sanders LH, Greenamyre JT. Oxidative damage to macromolecules in human Parkinson disease and the rotenone model. Free Radic Biol Med. 2013;62:111–20. doi: 10.1016/j.freeradbiomed.2013.01.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87.Zhou H, Zhang F, Chen SH, Zhang D, Wilson B, Hong JS, et al. Rotenone activates phagocyte NADPH oxidase by binding to its membrane subunit gp91phox. Free Radic Biol Med. 2012;52:303–13. doi: 10.1016/j.freeradbiomed.2011.10.488. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.Klintworth H, Garden G, Xia Z. Rotenone and paraquat do not directly activate microglia or induce inflammatory cytokine release. Neurosci Lett. 2009;462:1–5. doi: 10.1016/j.neulet.2009.06.065. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89.Ferger AI, Campanelli L, Reimer V, Muth KN, Merdian I, Ludolph AC, et al. Effects of mitochondrial dysfunction on the immunological properties of microglia. J Neuroinflammation. 2010;7:45. doi: 10.1186/1742-2094-7-45. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.Zhou F, Wu JY, Sun XL, Yao HH, Ding JH, Hu G. Iptakalim alleviates rotenone-induced degeneration of dopaminergic neurons through inhibiting microglia-mediated neuroinflammation. Neuropsychopharmacology. 2007;32:2570–80. doi: 10.1038/sj.npp.1301381. [DOI] [PubMed] [Google Scholar]
  • 91.Zhou F, Yao HH, Wu JY, Ding JH, Sun T, Hu G. Opening of microglial K(ATP) channels inhibits rotenone-induced neuroinflammation. J Cell Mol Med. 2008;12:1559–70. doi: 10.1111/j.1582-4934.2007.00144.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92.Zhou R, Yazdi AS, Menu P, Tschopp J. A role for mitochondria in NLRP3 inflammasome activation. Nature. 2011;469:221–5. doi: 10.1038/nature09663. [DOI] [PubMed] [Google Scholar]
  • 93.Nakahira K, Haspel JA, Rathinam VA, Lee SJ, Dolinay T, Lam HC, et al. Autophagy proteins regulate innate immune responses by inhibiting the release of mitochondrial DNA mediated by the NALP3 inflammasome. Nat Immunol. 2011;12:222–30. doi: 10.1038/ni.1980. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94.Choi WS, Palmiter RD, Xia Z. Loss of mitochondrial complex I activity potentiates dopamine neuron death induced by microtubule dysfunction in a Parkinson's disease model. J Cell Biol. 2011;192:873–82. doi: 10.1083/jcb.201009132. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95.Brinkley BR, Barham SS, Barranco SC, Fuller GM. Rotenone inhibition of spindle microtubule assembly in mammalian cells. Exp Cell Res. 1974;85:41–6. doi: 10.1016/0014-4827(74)90210-9. [DOI] [PubMed] [Google Scholar]
  • 96.Marshall LE, Himes RH. Rotenone inhibition of tubulin self-assembly. Biochim Biophys Acta. 1978;543:590–4. doi: 10.1016/0304-4165(78)90315-x. [DOI] [PubMed] [Google Scholar]
  • 97.Ren Y, Liu W, Jiang H, Jiang Q, Feng J. Selective vulnerability of dopaminergic neurons to microtubule depolymerization. J Biol Chem. 2005;280:34105–12. doi: 10.1074/jbc.M503483200. [DOI] [PubMed] [Google Scholar]
  • 98.Voloboueva LA, Emery JF, Sun X, Giffard RG. Inflammatory response of microglial BV-2 cells includes a glycolytic shift and is modulated by mitochondrial glucose-regulated protein 75/mortalin. FEBS Lett. 2013;587:756–62. doi: 10.1016/j.febslet.2013.01.067. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99.Moss DW, Bates TE. Activation of murine microglial cell lines by lipopolysaccharide and interferon-gamma causes NO-mediated decreases in mitochondrial and cellular function. Eur J Neurosci. 2001;13:529–38. doi: 10.1046/j.1460-9568.2001.01418.x. [DOI] [PubMed] [Google Scholar]
  • 100.Chenais B, Morjani H, Drapier JC. Impact of endogenous nitric oxide on microglial cell energy metabolism and labile iron pool. J Neurochem. 2002;81:615–23. doi: 10.1046/j.1471-4159.2002.00864.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101.Kasahara E, Sekiyama A, Hori M, Hara K, Takahashi N, Konishi M, et al. Mitochondrial density contributes to the immune response of macrophages to lipopolysaccharide via the MAPK pathway. FEBS Lett. 2011;585:2263–8. doi: 10.1016/j.febslet.2011.05.049. [DOI] [PubMed] [Google Scholar]
  • 102.Arsenijevic D, Onuma H, Pecqueur C, Raimbault S, Manning BS, Miroux B, et al. Disruption of the uncoupling protein-2 gene in mice reveals a role in immunity and reactive oxygen species production. Nat Genet. 2000;26:435–9. doi: 10.1038/82565. [DOI] [PubMed] [Google Scholar]
  • 103.Bai Y, Onuma H, Bai X, Medvedev AV, Misukonis M, Weinberg JB, et al. Persistent nuclear factorkappa B activation in Ucp2-/- mice leads to enhanced nitric oxide and inflammatory cytokine production. J Biol Chem. 2005;280:19062–9. doi: 10.1074/jbc.M500566200. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104.Emre Y, Hurtaud C, Nubel T, Criscuolo F, Ricquier D, Cassard-Doulcier AM. Mitochondria contribute to LPS-induced MAPK activation via uncoupling protein UCP2 in macrophages. Biochem J. 2007;402:271–8. doi: 10.1042/BJ20061430. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105.Basu BW, Kar S, Mukherjee M, Chande AG, Mukhopadhyaya R, Das PK. Uncoupling protein 2 negatively regulates mitochondrial reactive oxygen species generation and induces phosphatase-mediated anti-inflammatory response in experimental visceral leishmaniasis. J Immunol. 2011;187:1322–32. doi: 10.4049/jimmunol.1004237. [DOI] [PubMed] [Google Scholar]
  • 106.Kizaki T, Suzuki K, Hitomi Y, Taniguchi N, Saitoh D, Watanabe K, et al. Uncoupling protein 2 plays an important role in nitric oxide production of lipopolysaccharide-stimulated macrophages. Proc Natl Acad Sci U S A. 2002;99:9392–7. doi: 10.1073/pnas.142206299. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107.de BF, Arsenijevic D, Vallet P, Hjelle OP, Ottersen OP, Bouras C, et al. Resistance to cerebral ischemic injury in UCP2 knockout mice: evidence for a role of UCP2 as a regulator of mitochondrial glutathione levels. J Neurochem. 2004;89:1283–92. doi: 10.1111/j.1471-4159.2004.02432.x. [DOI] [PubMed] [Google Scholar]
  • 108.Echtay KS, Roussel D, St-Pierre J, Jekabsons MB, Cadenas S, Stuart JA, et al. Superoxide activates mitochondrial uncoupling proteins. Nature. 2002;415:96–9. doi: 10.1038/415096a. [DOI] [PubMed] [Google Scholar]
  • 109.Cannon B, Shabalina IG, Kramarova TV, Petrovic N, Nedergaard J. Uncoupling proteins: a role in protection against reactive oxygen species--or not? Biochim Biophys Acta. 2006;1757:449–58. doi: 10.1016/j.bbabio.2006.05.016. [DOI] [PubMed] [Google Scholar]
  • 110.Vozza A, Parisi G, De LF, Lasorsa FM, Castegna A, Amorese D, et al. UCP2 transports C4 metabolites out of mitochondria, regulating glucose and glutamine oxidation. Proc Natl Acad Sci U S A. 2014;111:960–5. doi: 10.1073/pnas.1317400111. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 111.Heid ME, Keyel PA, Kamga C, Shiva S, Watkins SC, Salter RD. Mitochondrial reactive oxygen species induces NLRP3-dependent lysosomal damage and inflammasome activation. J Immunol. 2014;191:5230–8. doi: 10.4049/jimmunol.1301490. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 112.Hanamsagar R, Torres V, Kielian T. Inflammasome activation and IL-1?/IL-18 processing are influenced by distinct pathways in microglia. J Neurochem. 2011;119:736–48. doi: 10.1111/j.1471-4159.2011.07481.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 113.Halle A, Hornung V, Petzold GC, Stewart CR, Monks BG, Reinheckel T, et al. The NALP3 inflammasome is involved in the innate immune response to amyloid-beta. Nat Immunol. 2008;9:857–65. doi: 10.1038/ni.1636. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 114.Parajuli B, Sonobe Y, Horiuchi H, Takeuchi H, Mizuno T, Suzumura A. Oligomeric amyloid beta induces IL-1beta processing via production of ROS: implication in Alzheimer's disease. Cell Death Dis. 2013;4:e975;. doi: 10.1038/cddis.2013.503. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 115.Codolo G, Plotegher N, Pozzobon T, Brucale M, Tessari I, Bubacco L, et al. Triggering of inflammasome by aggregated alpha-synuclein, an inflammatory response in synucleinopathies. PLoS One. 2013;8:e55375. doi: 10.1371/journal.pone.0055375. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 116.Shi F, Yang L, Kouadir M, Yang Y, Wang J, Zhou X, et al. The NALP3 inflammasome is involved in neurotoxic prion peptide-induced microglial activation. J Neuroinflammation. 2012;9:73. doi: 10.1186/1742-2094-9-73. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 117.Martinon F. Signaling by ROS drives inflammasome activation. Eur J Immunol. 2010;40:616–9. doi: 10.1002/eji.200940168. [DOI] [PubMed] [Google Scholar]
  • 118.Bauernfeind F, Ablasser A, Bartok E, Kim S, Schmid-Burgk J, Cavlar T, et al. Inflammasomes: current understanding and open questions. Cell Mol Life Sci. 2011;68:765–83. doi: 10.1007/s00018-010-0567-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 119.Zhou R, Tardivel A, Thorens B, Choi I, Tschopp J. Thioredoxin-interacting protein links oxidative stress to inflammasome activation. Nat Immunol. 2010;11:136–40. doi: 10.1038/ni.1831. [DOI] [PubMed] [Google Scholar]
  • 120.Kaushik DK, Gupta M, Kumawat KL, Basu A. NLRP3 inflammasome: key mediator of neuroinflammation in murine Japanese encephalitis. PLoS One. 2012;7:e32270. doi: 10.1371/journal.pone.0032270. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 121.Dostert C, Petrilli V, Van Bruggen R, Steele C, Mossman BT, Tschopp J. Innate immune activation through Nalp3 inflammasome sensing of asbestos and silica. Science. 2008;320:674–7. doi: 10.1126/science.1156995. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 122.Hornung V, Bauernfeind F, Halle A, Samstad EO, Kono H, Rock KL, et al. Silica crystals and aluminum salts activate the NALP3 inflammasome through phagosomal destabilization. Nat Immunol. 2008;9:847–56. doi: 10.1038/ni.1631. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 123.Bulua AC, Simon A, Maddipati R, Pelletier M, Park H, Kim KY, et al. Mitochondrial reactive oxygen species promote production of proinflammatory cytokines and are elevated in TNFR1-associated periodic syndrome (TRAPS) J Exp Med. 2011;208:519–33. doi: 10.1084/jem.20102049. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 124.Meissner F, Serger RA, Moshous D, Fischer A, Reichenbach J, Zychlinsky A. Inflammasome activation in NADPH oxidase defective mononuclear phagocytes from patients with chronic granulomatous disease. Blood. 2010;116:1570–3. doi: 10.1182/blood-2010-01-264218. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 125.Oh JY, Ko JH, Lee HJ, Yu JM, Choi H, Kim MK, et al. Mesenchymal stem/stromal cells inhibit the NLRP3 inflammasome by decreasing mitochondrial reactive oxygen species. Stem Cells. 2013 doi: 10.1002/stem.1608. [DOI] [PubMed] [Google Scholar]
  • 126.Dashdorj A, Jyothi KR, Lim S, Jo A, Nguyen MN, Ha J, et al. Mitochondria-targeted antioxidant MitoQ ameliorates experimental mouse colitis by suppressing NLRP3 inflammasome-mediated inflammatory cytokines. BMC Med. 2013;11:178. doi: 10.1186/1741-7015-11-178. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 127.Jabaut J, Ather JL, Taracanova A, Poynter ME, Ckless K. Mitochondria-targeted drugs enhance Nlrp3 inflammasome-dependent IL-1beta secretion in association with alterations in cellular redox and energy status. Free Radic Biol Med. 2013;60:233–45. doi: 10.1016/j.freeradbiomed.2013.01.025. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 128.Munoz-Planillo R, Kuffa P, Martinez-Colon G, Smith BL, Rajendiran TM, Nunez G. K(+) efflux is the common trigger of NLRP3 inflammasome activation by bacterial toxins and particulate matter. Immunity. 2013;38:1142–53. doi: 10.1016/j.immuni.2013.05.016. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 129.Bauernfeind F, Bartok E, Rieger A, Franchi L, Nunez G, Hornung V. Cutting edge: reactive oxygen species inhibitors block priming, but not activation, of the NLRP3 inflammasome. J Immunol. 2011;187:613–7. doi: 10.4049/jimmunol.1100613. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 130.Iyer SS, He Q, Janczy JR, Elliott EI, Zhong Z, Olivier AK, et al. Mitochondrial cardiolipin is required for Nlrp3 inflammasome activation. Immunity. 2013;39:311–23. doi: 10.1016/j.immuni.2013.08.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 131.Smith RA, Porteous CM, Coulter CV, Murphy MP. Selective targeting of an antioxidant to mitochondria. Eur J Biochem. 1999;263:709–16. doi: 10.1046/j.1432-1327.1999.00543.x. [DOI] [PubMed] [Google Scholar]
  • 132.Robinson KM, Janes MS, Pehar M, Monette JS, Ross MF, Hagen TM, et al. Selective fluorescent imaging of superoxide in vivo using ethidium-based probes. Proc Natl Acad Sci U S A. 2006;103:15038–43. doi: 10.1073/pnas.0601945103. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 133.Murphy MP, Smith RA. Drug delivery to mitochondria: the key to mitochondrial medicine. Adv Drug Deliv Rev. 2000;41:235–50. doi: 10.1016/s0169-409x(99)00069-1. [DOI] [PubMed] [Google Scholar]
  • 134.Mukhopadhyay P, Rajesh M, Yoshihiro K, Hasko G, Pacher P. Simple quantitative detection of mitochondrial superoxide production in live cells. Biochem Biophys Res Commun. 2007;358:203–8. doi: 10.1016/j.bbrc.2007.04.106. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 135.Robinson KM, Janes MS, Beckman JS. The selective detection of mitochondrial superoxide by live cell imaging. Nat Protoc. 2008;3:941–7. doi: 10.1038/nprot.2008.56. [DOI] [PubMed] [Google Scholar]
  • 136.Johnson-Cadwell LI, Jekabsons MB, Wang A, Polster BM, Nicholls DG. ‘Mild Uncoupling’ does not decrease mitochondrial superoxide levels in cultured cerebellar granule neurons but decreases spare respiratory capacity and increases toxicity to glutamate and oxidative stress. J Neurochem. 2007;101:1619–31. doi: 10.1111/j.1471-4159.2007.04516.x. [DOI] [PubMed] [Google Scholar]
  • 137.Zielonka J, Kalyanaraman B. Hydroethidine- and MitoSOX-derived red fluorescence is not a reliable indicator of intracellular superoxide formation: another inconvenient truth. Free Radic Biol Me. 2010;48:983–1001. doi: 10.1016/j.freeradbiomed.2010.01.028. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 138.Shimada K, Crother TR, Karlin J, Dagvadorj J, Chiba N, Chen S, et al. Oxidized mitochondrial DNA activates the NLRP3 inflammasome during apoptosis. Immunity. 2012;36:401–14. doi: 10.1016/j.immuni.2012.01.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 139.Dikalova AE, Bikineyeva AT, Budzyn K, Nazarewicz RR, McCann L, Lewis W, et al. Therapeutic targeting of mitochondrial superoxide in hypertension. Circ Res. 2010;107:106–16. doi: 10.1161/CIRCRESAHA.109.214601. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 140.Reily C, Mitchell T, Chacko BK, Benavides G, Murphy MP, Darley-Usmar V. Mitochondrially targeted compounds and their impact on cellular bioenergetics. Redox Biol. 2013;1:86–93. doi: 10.1016/j.redox.2012.11.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 141.Wynosky-Dolfi MA, Snyder AG, Philip NH, Doonan PJ, Poffenberger MC, Avizonis D, et al. Oxidative metabolism enables Salmonella evasion of the NLRP3 inflammasome. J Exp Med. 2014;211:653–68. doi: 10.1084/jem.20130627. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 142.Ma Q, Chen S, Hu Q, Feng H, Zhang JH, Tang J. NLRP3 inflammasome contributes to inflammation after intracerebral hemorrhage. Ann Neurol. 2014;75:209–19. doi: 10.1002/ana.24070. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 143.Kurland DB, Tosun C, Pampori A, Karimy JK, Caffes NM, Gerzanich V, et al. Glibenclamide for the treatment of acute CNS injury. Pharmaceuticals (Basel) 2013;6:1287–303. doi: 10.3390/ph6101287. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 144.Lamkanfi M, Mueller JL, Vitari AC, Misaghi S, Fedorova A, Deshayes K, et al. Glyburide inhibits the Cryopyrin/Nalp3 inflammasome. J Cell Biol. 2009;187:61–70. doi: 10.1083/jcb.200903124. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 145.Tosun C, Kurland DB, Mehta R, Castellani RJ, deJong JL, Kwon MS, et al. Inhibition of the Sur1-Trpm4 channel reduces neuroinflammation and cognitive impairment in subarachnoid hemorrhage. Stroke. 2013;44:3522–8. doi: 10.1161/STROKEAHA.113.002904. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 146.Goldmann T, Tay TL, Prinz M. Love and death: microglia, NLRP3 and the Alzheimer's brain. Cell Res. 2013;23:595–6. doi: 10.1038/cr.2013.24. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 147.Heneka MT, Kummer MP, Stutz A, Delekate A, Schwartz S, Vieira-Saecker A, et al. NLRP3 is activated in Alzheimer's disease and contributes to pathology in APP/PS1 mice. Nature. 2013;493:674–8. doi: 10.1038/nature11729. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 148.Bauernfeind FG, Horvath G, Stutz A, Alnemri ES, MacDonald K, Speert D, et al. Cutting edge: NF-kappaB activating pattern recognition and cytokine receptors license NLRP3 inflammasome activation by regulating NLRP3 expression. J Immunol. 2009;183:787–91. doi: 10.4049/jimmunol.0901363. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 149.Masters SL, Dunne A, Subramanian SL, Hull RL, Tannahill GM, Sharp FA, et al. Activation of the NLRP3 inflammasome by islet amyloid polypeptide provides a mechanism for enhanced IL-1beta in type 2 diabetes. Nat Immunol. 2010;11:897–904. doi: 10.1038/ni.1935. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 150.Misawa T, Takahama M, Kozaki T, Lee H, Zou J, Saitoh T, et al. Microtubule-driven spatial arrangement of mitochondria promotes activation of the NLRP3 inflammasome. Nat Immunol. 2013;14:454–60. doi: 10.1038/ni.2550. [DOI] [PubMed] [Google Scholar]
  • 151.Li P, Nijhawan D, Budihardjo I, Srinivasula SM, Ahmad M, Alnemri ES, et al. Cytochrome c and dATP-dependent formation of Apaf-1/caspase-9 complex initiates an apoptotic protease cascade. Cell. 1997;91:479–89. doi: 10.1016/s0092-8674(00)80434-1. [DOI] [PubMed] [Google Scholar]
  • 152.Kowaltowski AJ, Castilho RF, Vercesi AE. Mitochondrial permeability transition and oxidative stress. FEBS Lett. 2001;495:12–5. doi: 10.1016/s0014-5793(01)02316-x. [DOI] [PubMed] [Google Scholar]
  • 153.Nguyen TT, Stevens MV, Kohr M, Steenbergen C, Sack MN, Murphy E. Cysteine 203 of cyclophilin D is critical for cyclophilin D activation of the mitochondrial permeability transition pore. J Biol Chem. 2011;286:40184–92. doi: 10.1074/jbc.M111.243469. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 154.Allam R, Lawlor KE, Yu EC, Mildenhall AL, Moujalled DM, Lewis RS, et al. Mitochondrial apoptosis is dispensable for NLRP3 inflammasome activation but non-apoptotic caspase-8 is required for inflammasome priming. EMBO Rep. 2014 doi: 10.15252/embr.201438463. in press. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 155.Petrilli V, Papin S, Dostert C, Mayor A, Martinon F, Tschopp J. Activation of the NALP3 inflammasome is triggered by low intracellular potassium concentration. Cell Death Differ. 2007;14:1583–9. doi: 10.1038/sj.cdd.4402195. [DOI] [PubMed] [Google Scholar]
  • 156.Meissner F, Molawi K, Zychlinsky A. Superoxide dismutase 1 regulates caspase-1 and endotoxic shock. Nat Immunol. 2008;9:866–72. doi: 10.1038/ni.1633. [DOI] [PubMed] [Google Scholar]
  • 157.Guillemin GJ, Brew BJ. Microglia, macrophages, perivascular macrophages, and pericytes: a review of function and identification. J Leukoc Biol. 2004;75:388–97. doi: 10.1189/jlb.0303114. [DOI] [PubMed] [Google Scholar]
  • 158.Hickman SE, Kingery ND, Ohsumi TK, Borowsky ML, Wang LC, Means TK, et al. The microglial sensome revealed by direct RNA sequencing. Nat Neurosci. 2013;16:1896–905. doi: 10.1038/nn.3554. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 159.Toku K, Tanaka J, Fujikata S, Hamamoto Y, Horikawa Y, Miyoshi K, et al. Distinctions between microglial cells and peripheral macrophages with regard to adhesive activities and morphology. J Neurosci Res. 1999;57:855–65. [PubMed] [Google Scholar]
  • 160.Chao CL, Weng CS, Chang NC, Lin JS, Kao ST, Ho FM. Naringenin more effectively inhibits inducible nitric oxide synthase and cyclooxygenase-2 expression in macrophages than in microglia. Nutr Res. 2010;30:858–64. doi: 10.1016/j.nutres.2010.10.011. [DOI] [PubMed] [Google Scholar]
  • 161.Lynch MA. The multifaceted profile of activated microglia. Mol Neurobiol. 2009;40:139–56. doi: 10.1007/s12035-009-8077-9. [DOI] [PubMed] [Google Scholar]
  • 162.Bracey NA, Gershkovich B, Chun J, Vilaysane A, Meijndert HC, Wright JR, Jr, et al. Mitochondrial NLRP3 Protein Induces Reactive Oxygen Species to Promote Smad Protein Signaling and Fibrosis Independent from the Inflammasome. J Biol Chem. 2014;289:19571–84. doi: 10.1074/jbc.M114.550624. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 163.Porasuphatana S, Tsai P, Rosen GM. The generation of free radicals by nitric oxide synthase. Comp Biochem Physiol C Toxicol Pharmacol. 2003;134:281–9. doi: 10.1016/s1532-0456(02)00271-5. [DOI] [PubMed] [Google Scholar]

RESOURCES