BMB Reports 2020; 53(1): 35-46  https://doi.org/10.5483/BMBRep.2020.53.1.274
Emerging perspectives on mitochondrial dysfunction and inflammation in Alzheimer’s disease
Seung-Min Yoo#, Jisu Park#, Seo-Hyun Kim & Yong-Keun Jung*
School of Biological Sciences, Seoul National University, Seoul 08826, Korea
Correspondence to: Tel: +82-2-880-4401; Fax: +82-2-8808-4185; E-mail: ykjung@snu.ac.kr
#These authors contributed equally to this work.
Received: October 28, 2019; Published online: January 31, 2020.
© Korean Society for Biochemistry and Molecular Biology. All rights reserved.

ABSTRACT

Despite enduring diverse insults, mitochondria maintain normal functions through mitochondrial quality control. However, the failure of mitochondrial quality control resulting from excess damage and mechanical defects causes mitochondrial dysfunction, leading to various human diseases. Recent studies have reported that mitochondrial defects are found in Alzheimer’s disease (AD) and worsen AD symptoms. In AD pathogenesis, mitochondrial dysfunction-driven generation of reactive oxygen species (ROS) and their contribution to neuronal damage has been widely studied. In contrast, studies on mitochondrial dysfunction-associated inflammatory responses have been relatively scarce. Moreover, ROS produced upon failure of mitochondrial quality control may be linked to the inflammatory response and influence the progression of AD. Thus, this review will focus on inflammatory pathways that are associated with and initiated through defective mitochondria and will summarize recent progress on the role of mitochondria-mediated inflammation in AD. We will also discuss how reducing mitochondrial dysfunction-mediated inflammation could affect AD.

Keywords: Alzheimer's disease, Dysfunction, Inflammation, Mitochondria
INTRODUCTION

Mitochondria play a wide range of roles in apoptosis, calcium homeostasis, cell proliferation, production of metabolic substrates, and inflammation, in addition to their primary responsibility of energy production in cells (1). Mitochondria operate various defense mechanisms from the protein level to the organelle level through mitochondrial quality control (MQC) to maintain normal functions (2). Notably, the failure of MQC results in mitochondrial dysfunction, which has been frequently associated with many diseases (3). Interestingly, damaged mitochondria are also found in the brains of patients with AD, and mitochondrial dysfunction is known to accelerate AD symptoms. In recent years, accumulating evidence has highlighted the essential role of inflammation in AD. Here, we provide an overview of the features of inflammation and mitochondrial dysfunction, and the mechanisms underlying the mitochondria-mediated inflammatory response in AD pathogenesis (Fig. 1). Furthermore, we explore possible ways of adjusting MQC and inflammation to ameliorate AD symptoms and pathogenesis (Table 1).

ROLE OF INFLAMMATION AND MITOCHONDRIA IN AD

Inflammation as a central mechanism of AD

Recent findings strengthen the implication of inflammation on the pathogenesis of AD. Genetic studies have consistently identified a list of genes that can act as risk factors for AD, regardless of amyloid-beta (Aβ) signal transduction. Triggering receptor expressed on myeloid cells 2 (TREM2) is expressed on microglial membranes, recognizes lipoproteins and phospholipids, and is involved in phagocytosis of microglial cells. Lack of TREM2 suppresses tau disease, gliosis, and neuroinflammation, because it helps the microglia respond to damage caused by tau disease (4). Besides, Apolipoprotein E (ApoE) is secreted by microglia and astrocytes and has three alleles, ε2, ε3, and ε4. In the central nervous system, ApoE binds to ApoE receptors present on nerve cells to regulate the development of the central nervous system and recovery of nerve defects. Among them, the ApoE4 allele is a genetic risk factor for sporadic AD (5), and as the ε4 gene increases, the age of onset of AD decreases. It is known that an impaired function of ApoE4 adversely affects Aβ removal and Aβ-induced inflammatory response (6, 7). As mentioned above, inflammatory process-linked proteins, such as TREM2 and ApoE, may act independently of Aβ signaling. In a bioinformatics study conducted by Zhang et al., the immune-and microglia-specific pathway, including TYROBP which is restricted to cells involved in the innate immunity, was also identified as a critical regulator of AD pathogenesis (8).

Since the 1980s, researchers have found components of the immune response, such as immunoglobulins and complement proteins near Aβ and elevated levels of cytokines and chemokines, in AD brains (9, 10). It has also been shown that anti-inflammatory drugs activate microglia and lower Aβ42 in vivo in mouse models (11, 12). Several human clinical trials have revealed that anti-inflammatory drugs reduce the risk of AD (13, 14). Thus, many researchers now agree that an association between neuroinflammation and AD pathogenesis exists and that AD pathogenesis and inflammation are the cause and effect of each other, regardless of what is triggered first. In the case of acute inflammation, microglia eliminate Aβ and prevent the ensuing detrimental consequences. Contrastingly, cytokines, chemokines, and ROS are over-produced by immune cells and exacerbate neurotoxicity in chronic inflammation. Whereas the former is beneficial in relieving neuropathology, the latter aggravates neurotoxicity. Next, we investigate the roles of inflammation with the opposing side to the pathogenesis of AD.

Neuroprotective inflammation in the pathogenesis of AD: Many studies have demonstrated that overexpression of inflammatory mediators in the AD mouse model plays a beneficial role in pathogenesis. Whereas aged amyloid precursor protein (APP) transgenic (TG) mice display increased production of astroglial TGF-β1 and reduction in the number of parenchymal amyloid plaques, mice expressing hAPP and TGF-β1 show Aβ accumulation in cerebral blood vessels (15). In the study conducted by Wyss-Coray et al., researchers observed that hAPP/TGF-β1 mice have markedly higher levels of C3, a component of the complement system, than do hAPP mice, and inhibition of C3 activation causes an increase in Aβ deposition and the number of degenerating neurons (16). Furthermore, pathogenic Aβ is eliminated by immune-related clearance mechanisms. For example, low-density lipoprotein receptor-related protein 1 (LRP-1) mediates the uptake of Aβ in astrocytes and neurons (17, 18). ATP-binding cassette subfamily A member 7 (ABCA7) also participates in phagocytic clearance of Aβ in the brain (19). These results suggest that immune activation and the subsequent microglial activation help relieve AD pathology.

Detrimental effect of inflammation on AD: Based on various AD mouse models, it is known that higher levels of cytokines trigger inflammation and thereby exacerbate AD pathology. The APP TG mouse model with exogenous expression of interleukin (IL)-10 in the brain displays elevated Aβ accumulation and memory deficit (20). The most common mechanism whereby Aβ is produced because of inflammation is inflammation-mediated regulation of APP, β-secretase 1 (BACE1), and γ-secretase expression. Activation of immune-related transcriptional factor NK-κB was reported to cause APP upregulation in neurons (21). Interferon (IFN)-γ also induces BACE1 expression through the JAK2-ERK1/2 signaling pathway in astrocytes (22). In addition, inflammation is thought to play a role in tau pathology in AD. Lipopolysaccharide (LPS) administration induces inflammation and aggravates tau pathology in the 3xTg AD mouse model (23). These results offer compelling evidence for the harmful effects of inflammation on AD pathogenesis.

Mitochondrial cascade in AD

Mitochondria are considered to play a critical role in the pathology of AD. Neurons need to produce large amounts of neurotransmitters and establish membrane excitability. Since mitochondria are responsible for ATP production, iron homeostasis, and Ca2+ signaling, neuronal viability relies highly on mitochondrial function. For example, mitochondria in the presynaptic nerve terminal primarily regulate presynaptic calcium at central glutamatergic terminals (24). Axon regeneration is also facilitated by increasing mitochondrial motility and recovering the energy deficit in mature neurons (25). Thus, mitochondrial defects are commonly observed in neurodegenerative diseases, including amyotrophic lateral sclerosis (ALS), Parkinson’s disease (PD), and AD. In ALS and PD, mitochondrial dysfunction and impaired mitochondrial fusion cause neuronal loss (26); mitochondria also play a pivotal role in the loss of hippocampal and cortical neurons in AD.

Electron microscopic studies have revealed that AD brains display abnormal mitochondrial structure in the hippocampus, acoustic cortex, frontal cortex, and cerebellum (27). A recent report identified a mitochondrial fission-arrest phenotype and elongated interconnected mitochondria in the hippocampus and entorhinal cortex of patients with AD (28). In addition to the structural abnormality, AD brains also exhibit mitochondrial malfunction, such as changes in glucose metabolism and oxygen consumption. The activity of the pyruvate dehydrogenase complex (PDHC) and 2-ketoglutarate dehydrogenase complex is reduced in the affected regions of AD brains (29, 30). The activity of complex I, complex II-III, and cytochrome oxidase is reduced in the cortex of AD brains (31). Increased oxidative damage to mitochondrial DNA is found in AD (32). Despite these studies on the corruption of mitochondrial function in AD brains, it is still debatable whether mitochondrial disruption is the main cause of AD or occurs as a consequence of pathological conditions in AD. This section describes the correlation between abnormal mitochondria and AD pathogenesis, focusing on the evidence from two conflicting hypotheses.

Mitochondrial dysfunction as an outcome of AD pathogenesis: The majority of researchers contend that Aβ-induced ROS generation and impaired calcium homeostasis lead to mitochondrial lesions, which are known as the secondary mitochondrial cascade. Overexpression of mutant APP in HT22 mouse hippocampal cell line results in defective mitochondrial dynamics and changes mitochondrial structure and function in neurons (33). APP can accumulate in mitochondrial import channels of AD brains and cause mitochondrial dysfunction (34). It has also been reported that Aβ directly disrupts mitochondrial function and inhibits key enzymatic activities. Lustbader et al. reported that alcohol dehydrogenase (ABAD) interacts with Aβ and mediates Aβ-induced apoptosis and free-radical generation in neurons (35). AD brains express higher levels of voltage-dependent anion-selective channel 1 (VDAC1), which interacts with Aβ and phosphorylated tau to block mitochondrial pores, precipitating mitochondrial dysfunction (36).

In addition, there are reports showing that mitochondrial fusion and fission factors are affected by Aβ. Aβ induces oxidative stress that triggers mitochondrial fragmentation through decreased mitofusin-2 (Mfn2) expression by activating cyclin-dependent kinase 5 (Cdk5)-mediated peroxidase 2 (Prx2) phosphorylation (37). Aβ also mediates dynamin-related protein 1 (Drp1) phosphorylation via AKT activation, promoting excessive mitochondrial fission and leading to neuronal apoptosis (38). Collectively, the results suggest that the accumulation of mitochondrial APP and Aβ contributes to the defective energy metabolism and mitochondrial abnormalities seen in AD.

Mitochondrial defects as a causative factor of AD: In stark contrast to the above, several studies have also implied that mitochondrial failure drives disease progression, which is known as the primary mitochondrial hypothesis. Mitochondrial loss leads to changes in ROS generation, altered calcium homeostasis, failure of mitochondrial homeostasis, and cell death. Neuron-specific deficiency of cytochrome C oxidase (COX) leads to a decrease in amyloid plaques, Aβ42 levels, β-secretase activity, and oxidative damage in the mouse model expressing mutant APP and PS1 (39). Other groups also claimed that high levels of mitochondrial DNA (mtDNA) deletion could cause COX deficiency (40). In the same context, an ultrasensitive next-generation sequencing analysis revealed an increase in mtDNA mutation frequency in AD brains (41, 42). Furthermore, injection of the mitochondrial complex I inhibitor rotenone into rats triggers tauopathy in the striatum (43). Whereas molecular mechanisms that corroborate the secondary mitochondrial hypothesis have been identified, there is only phenomenological evidence for the primary mitochondrial hypothesis that mitochondrial impairments dictate AD pathology. Nonetheless, based on the above observations, mitochondrial failure possibly facilitates AD pathogenesis.

RELATIONSHIP BETWEEN MITOCHONDRIA AND INFLAMMATION

Mitochondrial quality control

Because mitochondria in eukaryotic cells are the major organelles that provide ATP through the electron-transport chain (ETC) and ETC inevitably generates ROS, mitochondrial DNA, proteins, and lipids are damaged first, causing mitochondrial dysfunction (44). As mentioned earlier, however, MQC preserves the normal function of mitochondria. At the molecular level, mitochondria have a specific DNA polymerase subunit gamma (PolG) for mtDNA repair (45) and chaperones, such as Hsp60/70, to repair misfolded proteins (46). Mitochondrial AAA protease in the mitochondrial intermembrane space, Chip in the mitochondrial inner membrane, and LON protease in the mitochondrial matrix decompose damaged and misfolded proteins (47). Damaged proteins in the mitochondrial envelope are also ubiquitinated by the E3 ubiquitin-protein ligase MARCH5 and degraded by proteasomes (48). Antioxidants can directly quench ROS to prevent ROS-mediated damages from occurring (45). At the organelle level, mitochondrial biogenesis responds to a variety of stress conditions, such as calorie restriction, exercise, NO, CO, and ROS. This process creates new mitochondria (49) and promotes mitochondrial fusion through MFN and optic atrophy 1 (OPA1) to compensate for the deficient components. Damaged mitochondria are separated by the fission process including Drp1 or by the budding from the mitochondrial membrane. Damaged mitochondria are wrapped in autophagosomes and then eventually degraded within lysosomes (2).

Mitochondria in inflammation

Inflammation involves pathogen-associated molecular patterns (PAMPs) presented by pathogens or external ligands and damage-associated molecular patterns (DAMPs), which are endogenous molecules released into the extracellular space because of tissue damage. PAMPs and DAMPs are recognized by pattern recognition receptors (PRRs), which generate innate immunity-related substances through intracellular signaling pathways. Mitochondria have many similarities to bacteria, so the escape of mitochondrial content into the cytosol or the extracellular space serves as a PAMP or DAMP, invoking PRR signaling (50). Hence, mitochondria act as regulators of inflammatory signaling.

Inflammation triggered by the leakage of mitochondrial components

Mitochondrial DNA: Mitochondria originated from α-proteobacteria through endosymbiosis in ancient eukaryotes (51) and have circular DNA and CpG motifs similar to those of bacteria (52). Circular DNA with unmethylated CpG motifs interacts with Toll-like receptor 9 (TLR9) and activates NF-κB (53). In addition, oxidized mtDNA binds to the leucine-rich repeat pyrin domain containing 3 (NLRP3) to activate the NLRP3 inflammasome and increase interleukin-1 beta (IL-1β) during cell death (54). The cyclic GMP-AMP synthase (cGAS) stimulator of interferon genes (STING) pathway also recognizes mtDNA in the cytosol and produces IFN-1 through the interferon regulatory factor 3 (IRF3) and NF-κB (55).

N-formyl peptides: Mitochondria synthesize bacteria-like peptides using 22 tRNAs and 2 mitochondrial ribosomes, and like those of prokaryotes, mitochondrial proteins are characterized by the presence of formyl-methionine (formyl-Met) at the N-terminal (56). The mitochondrial formyl-Met acts as a chemoattractant for neutrophils when exposed outside of the cells (57) and formyl-peptides bind to the formyl peptide receptors (FPRs) to activate the FPR signaling pathway in neutrophils (58). In experiments with formyl methionine-leucine-phenylalanine (fMLP), fMLP induces IL-1β production through a rapid increase in PI3K- NF-κB activity (59).

Cardiolipin: Cardiolipin is a phospholipid that constitutes about 20% of the mitochondrial inner membrane. This phospholipid is common in bacteria but is found only in the mitochondrial inner membrane in eukaryotic cells. Cardiolipin regulates mitochondrial dynamics, apoptotic signaling, mitophagy, and ROS generation by noncovalent interactions (60). Exposure of cardiolipin on the mitochondrial outer membrane activates NLRP3 inflammasome by directly binding to NLRP3, resulting in the production of IL-1β (61). Externalized cardiolipin also induces mitophagy by binding to light-chain 3 (LC3), a receptor for autophagy (62).

ATP: Under normal conditions, extracellular ATP is rapidly degraded by the nucleotidases CD39 and CD73 (63). However, an acute increase in extracellular ATP induces IL-18 by binding to P2X7 receptor and activating NLRP3 inflammasome in macrophages (64). In the acute lung-injury model, LPS-mediated inflammation leads to a temporary accumulation of ATP in the airways of mice. Treatment with apyrase to degrade extracellular ATP reduces LPS-mediated inflammation (65).

Cytochrome C: Cytochrome C (Cyt C) is a protein that transports electrons from complex III to complex IV of ETC in the mitochondrial inner membrane. The release of Cyt C into the cytosol induces apoptosis by binding to Apaf-1, but the role of extracellular Cyt C is not clear. Some reports have shown that Cyt C may affect inflammation (66). Cyt C injected into CD8+ dendritic cells reduces IL-12 production (67). Extracellular Cyt C promotes lymphocyte death and leucine-rich alpha-2-glycoprotein-1 (LRG1) binds to Cyt C and reduces its toxicity (68). Additionally, it has been shown that extracellular Cyt C increases NF-κB activity and cytokine production in mouse spleen cells (69).

Mitochondrial ROS: In general, ROS induces functional errors by oxidizing proteins, lipids, and DNA. Mitochondria produce energy through ETC, which also generates mitochondrial ROS (mtROS) during electron transfer at the complex I and III in the mitochondrial inner membrane (70). The mtROS is released into the cytoplasm when high concentrations of Ca2+ and cyclophilin D convert the ATP synthase into a non-specific pore or when ROS opens mitochondrial permeability transition pores (71) and drives proinflammatory cytokine production. Inhibition of mtROS reduces IL-6 levels produced by LPS treatment (72), whereas mtROS increases IL-1β by activating NLRP3 inflammasomes and induces IL-6 production through inflammasome-independent transcriptional regulation (73).

Mitochondrial transcription factor A: The primary role of mitochondrial transcription factor A (TFAM) is to regulate nucleoids, a condensed form of mitochondrial DNA. TFAM deficiency induces mtDNA mutations and mtDNA escapes into the cytosol, where it induces Type I IFN production through the cGAS-STING pathway (74). Additionally, extracellular TFAM is inactive, but has structural homology with HMGB1, which binds to DNA and induces inflammation (75). Similarly, binding of TFAM to mtDNA activates Type 1 IFN in plasmacytoid dendritic cells through the RAGE-TLR9 signaling pathway (76).

Regulators of inflammatory signaling pathway in mitochondria

Mitochondrial antiviral signaling protein: Retinoic acid-inducible gene 1 receptor (RIG1)-like receptors RIG-I and MDA5 recognize different types of viral RNA (vRNA) in the cytosol and bind to mitochondrial antiviral signaling protein (MAVS) on the mitochondrial outer membrane or peroxisome by interacting via caspase activation and recruitment domains (CARDs) (77). MAVS present on the mitochondrial outer membrane activates NF-κB and IRF3 (78) or recruits NLRP3 inflammasomes to mitochondria for IL-1β production (79). In addition, MAVS is activated by mitochondrial dynamics; mitochondrial elongation induces MAVS activation, but its fission decreases MAVS expression (80). MFN2, which is required for mitochondrial fusion, directly binds to MAVS and inhibits its activity (81). Mitochondrial dynamics affect MAVS, probably because of the need for self-oligomerization for activation (82).

Evolutionarily conserved signaling intermediate in Toll pathway: Evolutionarily conserved signaling intermediate in Toll pathway (ECSIT) was identified as a TRAF-6 binding protein and is an E3 ligase involved in the TLR signaling pathway. ECSIT is a cytosolic protein, but interacts with the chaperone NDUFAF1 and traffics to the mitochondria to regulate complex I assembly (83). In the TLR signaling pathway, ECSIT binds to TRAF6 to recruit mitochondria to the phagosomal membrane and produce mtROS (84). An increase in constitutive mtROS production in ECSIT-deleted macrophages prevents further TLR-induced mtROS production, demonstrating the key role of ECSIT in mtROS production and mitophagy-dependent MQC (85).

Membrane-associated ring finger (C3HC4) 5:Membrane-associated ring finger (C3HC4) 5 (MARCH5), an E3 ligase present in the mitochondrial outer membrane, increases inflammation by poly-ubiquitinating and attenuating TANK, a TRAF-interacting protein. TANK inhibits TRAF6 in the TLR7 signaling pathway, revealing a role of mitochondria in modulating innate immunity and linking mitochondria to the TLR signaling pathway (86).

MITOCHONDRIAL-INFLAMMATION AXIS IN AD PATHOLOGY

Evidence for mitochondrial DAMPs and PAMPs in AD pathology

As discussed, mitochondrial DAMPs and PAMPs can activate inflammation. In the central nervous system (CNS), they initiate pro-inflammatory immune responses in glial cells, thereby leading to chronic neuroinflammation and accelerating the pathology of neurodegenerative diseases, including AD (8789).

Mitochondrial DNA: It has been shown that mtDNA induces neuroinflammation in vivo. Injection of mitochondrial lysates or mtDNA into the hippocampal dentate gyri triggers pro-inflammatory signaling (90). Hippocampal injection of mitochondria or mtDNA leads to NF-κB phosphorylation, induction of TNFα mRNA, and a decrease in TREM2 expression, all of which are closely associated with AD pathology (9193) and are involved in anti-inflammatory and phagocytic pathways (94, 95). Simultaneously, hippocampal injection of mtDNA increases astrocyte proliferation with elevated levels of cortical colony-stimulating factor 1 receptor (CSF1R) and GFAP proteins. Interestingly, mitochondrial lysates also upregulate endogenous APP and Aβ (90), strongly supporting the correlation between mtDNA and AD pathology. Moreover, the relevance of mtDNA levels to AD pathology was reported (96). Circulating cell-free mtDNA is profoundly downregulated in the cerebrospinal fluid (CSF) of patients with sporadic AD as well as asymptomatic subjects at risk (96). Notably, preclinical subjects with pathogenic mutations in PSEN1 exhibit a reduction in the mtDNA concentration in CSF before other AD-related biomarkers in CSF can be detected, highlighting the use of mtDNA as a potential AD biomarker. A recent study using post-mortem brain tissues reported the regional differences in mtDNA levels in human brains; the mtDNA levels in the parietal cortex is lower in non-diabetic AD subjects, but not in diabetic AD patients than in non-cognitively impaired controls (97).

Cardiolipin: It has been described that aged brains, in addition to elevated ROS production, have lower levels of cardiolipin (98, 99). In contrast, the proportion of peroxidized cardiolipin is higher in the brains of aged rodents (100), which results in various mitochondrial defects, such as low respiratory chain efficiency and elevated ROS production (99101). This excessive level of ROS may contribute to chronic inflammation. Activation of the NLRP3 inflammasome is attenuated by pharmacological inhibitors of ROS production (61). Therefore, age-related cardiolipin oxidation results in mitochondrial dysfunction and aberrant ROS production that subsequently provokes chronic inflammation.

Mitochondrial transcription factor A: TFAM is implicated in the inflammation of the CNS in neurodegenerative diseases (89). TFAM upregulates secretion of IL-6 and cytotoxins in primary microglia that were obtained from post-mortem human samples or THP-1 human monocytic cells, a model of human microglia (102). Administration of TFAM into the cisterna magna in the rodent model increases levels of pro-inflammatory cytokines, including IL-1β, IL-6, and TNF-α, in the hippocampus and frontal cortex (103), which are the predominant regions affected by AD (89). These results underscore the ability of extracellular TFAM to induce pro-inflammatory responses in microglia. Furthermore, when combined with CpG-rich mtDNA, TFAM can activate RAGE to mediate a pro-inflammatory immune response and promote the production of TNF-α via the PI3K/AKT and ERK pathways (104). Accordingly, blocking RAGE with antagonistic antibodies inhibits the secretion of monocyte chemotactic protein-1 (MCP-1) in TFAM-stimulated THP-1 cells (103). Considering that RAGE binds to Aβ (105, 106) and that microglia express high levels of RAGE in patients with AD (107), the TFAM-RAGE pathway may potentially play a role in AD pathogenesis (87).

Cytochrome C: Given that Cyt C is critical for the regulation of apoptosis, it has been implicated in the excessive cell death observed during the progression of AD. Reports showed that CSF Cyt C levels are increased in patients with MCI (87, 108). Whereas the release of Cyt C from mitochondria is considered to be a mediator of cell death in AD (109), Cyt C released into the extracellular space may be able to provoke PAMP responses (87). Mouse splenocytes exposed to Cyt C show pro-inflammatory activity, including the release of inflammatory mediators such as chemokine ligand 5 (CCL5)/RANTES, CCL3/MIP-1α, and MCP-1 (69). As circulating Cyt C is increased in many chronic inflammatory diseases, such as liver injury, SIRS, and myocardial infarction (110), extracellular Cyt C could also activate microglia-like cells to exacerbate inflammatory damage, probably by interacting with TLR4 on microglia (111).

STRATEGIES FOR TARGETING MITOINFLAMMATION PATHWAYS

Targeting mtDAMP and mtPAMP

Mitochondrial ROS: Oxidative stress mediates mitochondrial damages during aging, particularly by damaging mtDNA and peroxidizing cardiolipin, and generates excessive ROS in turn as a byproduct. In this regard, the mitochondria-targeted antioxidants (MTAs) that specifically curtail oxidative stress within mitochondria have greater advantages than do untargeted cellular antioxidants (112). MTAs can cross the mitochondrial phospholipid bilayer and sequester ROS where it is generated. MTAs, such as MitoQ and MitoVitE, are more efficient in alleviating the damage caused by excessive ROS levels and blocking apoptosis than are untargeted antioxidants (113). Particularly, MitoQ is protective in the aged rodent model of neurodegenerative diseases, such as PD (114, 115), and suppresses the NLRP3 inflammasome-mediated production of inflammatory cytokines in THP-1 cells (116). In addition, the release of metals can further exacerbate the oxidative damage mediated by high ROS levels (112). Tiron, one of the MTAs, can confer marked protection against mtROS, because it targets not just ROS but also free intracellular metals that are released as a consequence of oxidative stress (112). Since iron accumulation and mtROS synergistically contribute to neurodegenerative pathology in AD and PD (117, 118), and chronic inflammation in microglia is characterized by an increase in intracellular iron levels (119), Tiron may mitigate chronic inflammation by reducing iron-mediated ROS stress in neurodegenerative disorders.

Mitochondrial DNA: mtDNA is relatively unstable and vulnerable to oxidative insults because they lack histones and have a limited enzymatic repair system. As a result, mtDNA mutations accumulate during aging (120, 121) and are a significant risk factor for AD (41, 122). Mutated mtDNA can be revised via genome-editing technologies, such as clustered regularly interspaced short palindromic repeats/associated protein 9 (CRISPR/Cas9) and transcription activator-like effector nucleases (TALENs) (42, 123). This strategy to revise the mutated mtDNA involves expressing a gRNA targeting the pathogenic mtDNA and mitoCas9 that is localized to the mitochondrial matrix and specifically cleaves the mtDNA. In addition, mitoTALENs were used to eliminate pathogenic mtDNA and thus recover respiratory capacity and improve oxidative phosphorylation (124).

Mitochondrial transcription factor A: Overexpression of mitochondrial TFAM exerted beneficial effects in model systems for aging-related hearing loss (125), memory loss (126), and AD (127, 128). In TFAM TG mice, age-related symptoms, such as mitochondrial deficits in the brain, motor learning memory, working memory, and hippocampal long-term potentiation (LTP), are alleviated (126). Remarkably, IL-1β was significantly reduced in aged TFAM TG mice, indicating compensatory suppression of the TFAM-mediated aberrant inflammatory response. TFAM overexpression also exhibits a protective effect in the 3xTg AD mouse model (PS1M146V, APPswe, and MAPT P301L triple TG), reducing cognitive dysfunction, mtDNA oxidative stress, and Aβ accumulation (128).

Targeting the inflammasome

Byproducts of mitochondrial dysfunction, such as mtROS and mtPAMP, can regulate the pro-inflammatory response by activating the inflammasome (123). Using Nlrp3 knockout and Caspase-1 knockout mice, the NLRP3/caspase-1 axis was shown to play an important role in the pathogenesis of AD (129). In agreement, inhibitors of the NLRP3 inflammasome ameliorate AD pathology in animal models of AD (130132). MCC950, which inhibits inflammasome and microglial activation in the APP/PS1 mouse model of AD (131), might inhibit NLRP3-induced oligomerization of ASC, a key adaptor protein that is required for the activation of the inflammasome (133). In addition, several clinically approved fenamate NSAIDs inhibit the NLRP3 inflammasome via the blockade of the volume-regulated anion channels (VRAC), a Cl channel, and consequently ameliorate cognitive impairment in animal models of AD (130).

Regulating mitochondrial quality control

Mitophagy: Tight regulation of MQC by facilitating mitophagy and subsequent inhibition of chronic inflammation were suggested as a potential therapeutic strategy for AD (134). A recent study by Fang et al. showed that enhancing mitophagy prevents AD pathology, including cognitive impairment, tau hyper-phosphorylation, Aβ accumulation, and neuroinflammation (135), highlighting the importance of MQC in AD intervention. Furthermore, mitochonic acid 5 (MA-5) was shown to regulate mitophagy via BCL2/adenovirus E1B 19-kDa protein-interacting protein 3 (BNIP3), reducing mitochondrial apoptosis in BV-2 cells (136). Mitophagy may inhibit inflammation by down-regulating ROS-producing mitochondria, since blocking mitophagy results in the increase of ROS, followed by NLRP3 activation (137).

Mitochondrial dynamics: Several studies reported that an imbalance of mitochondrial dynamics induces chronic inflammatory stress and thus aggravates the pathogenesis of neurodegenerative disorders. Disruption of mitochondrial fusion by Mfn2 knockout in the hippocampus results in excessive mitochondrial fragmentation and inflammatory response, which are the characteristic features of AD pathology (138). In contrast, negative regulation of mitochondrial fission by genetic or pharmacological methods significantly alleviates inflammation. Inhibiting mitochondrial fission by Mdivi-1, a chemical inhibitor of Drp1 or Drp1 knockdown, reduces pro-inflammatory signaling in the LPS-stimulated BV-2 cells (139) and a kainic acid-injected rodent model (140). Recently, Joshi et al. demonstrated that neurotoxicity can be directly attributed to the release of neurotoxic proteins from microglia displaying Drp1 and Fis1-mediated mitochondrial fragmentation, followed by the activation of naive astrocytes to the A1 state (141). This neurotoxicity could be reversed by the treatment with a heptapeptide (P110) that blocks the Drp1-Fis1 interaction. Interestingly, AD patients show a distinct pattern of mitochondrial dynamics (142). AD mitochondria exhibit significant fragmentation in a Drp1-dependent manner, whereas MCI mitochondria have increased mitochondrial Mfn2 levels, likely promoting mitochondrial fusion. These changes in mitochondrial dynamics may contribute to the induction of pro-inflammatory signaling in microglial cells. Taken together, subtle regulation of mitochondrial dynamics during disease progression may be a possible therapeutic strategy to relieve inflammatory stress and thus alleviate AD pathology.

cGAS-STING pathway

Binding of oxidized mtDNA to cGAS results in the translocation of STING to the Golgi apparatus, leading to phosphorylation of the transcription factor IRF3 and activation of NF-κB signaling (143). The cGAS-STING pathway has also been found to be involved in autophagy in innate immune cells (55). Activation of the cGAS-STING pathway promotes mitophagy through cGAS/beclin-1 interaction, which in turn negatively regulates cGAS activity and increases cytosolic DNA degradation (144). A recent study has elucidated that aberrant mitophagy in Prkn or Pink1 knockout mice leads to a strong inflammatory phenotype, which is mitigated by genetic inactivation of STING (145). Thus, the cGAS–STING pathway may be a potent therapeutic target to counter mitoinflammation.

CONCLUSION

Mitochondrial functions and inflammatory signals are closely linked to AD symptoms and pathogenesis. In this review, we described mitochondrial components as being causative factors of inflammation, but simultaneously are suitable therapeutic targets in regulating the neuroinflammation (Fig. 1, Table 1). Indeed, inhibiting mitochondrial inflammation or maintaining functional mitochondria through MQC reverts many symptoms observed in the AD model. Thus, mitochondrial inflammation is a valuable diagnostic target and requires further study as an emerging therapeutic target for treating AD.

ACKNOWLEDGEMENTS

This work was supported by a Bio & Medical Technology Development Program of the National Research Foundation (NRF-2017M3A9G7073521) and a CRI grant (NRF-2019R1A 2B5B03070352) funded by the Ministry of Education, Science and Technology, Korea.

CONFLICTS OF INTEREST

The authors have no conflicting interests.

FIGURE
Fig. 1. Mitochondria as a regulator of inflammation. (Upper) Leakage of mitochondrial components. (a) Leaked N-formyl peptide binds to formyl peptide receptor (FPR) to activate NF-κB. (b) Leakage of mtDNA to the cytosol activates Toll-Like-Receptor (TLR), cyclic GMP-AMP synthase-simulator of interferon genes (cGAS-STING) pathways, and NLRP3 inflammasome. (c) ATP and cardiolipin activate NLRP3 inflammasome. (d) Mitochondrial ROS (mtROS) activates NLRP3 inflammasome and mitogen-activated protein kinase (MAPK) signaling. (e) Binding of mtDNA to cytosolic TFAM activates the TLR pathway. (f) Cytosolic cytochrome C (Cyt C) reduces IL-12 and increases lymphocyte cell death. (Lower) Regulation of inflammation pathway by mitochondrial factors. (g) MAVS recruits RIG-I or MDA5 to viral RNA to activate NF-κB and IRF3. (h) ESCIT generates mtROS by binding to TRAF6. (i) MARCH5 ubiquitinates TANK to enhance TRAF6 signaling.
TABLE

Strategies to modify the stress in the mitochondria-inflammation pathway

Targeting of mitoinflammation pathways Strategies Beneficial or detrimental* References
Targeting mtDAMP and mtPAMP mtROS Mitochondria-targeted antioxidant Mito Q, MitoVitE, Tiron (112116)
mtDNA Mitochondrial genomic editing mtCRISPR/Cas9, mtTALEN (42, 123125)
TFAM TFAM ectopic expression TFAM transgenic mouse (126129)
Targeting inflammasome NLRP3 inflammasome Pharmacological inhibition of NLRP3 MCC950, JC-124, Fenamate NSAIDs (131133)
Regulating mitochondrial quality control Mitophagy Pharmacological enhancement of mitophagy Nicotinamide mononucleotide (NMN), Urolithin A (UA), Actinonin (AC), Mitochonic acid 5 (MA-5) (136, 137)
Mitochondrial dynamics Mitochondrial fission Mdivi-1, Heptapeptide P110 (140142)
Mitochondrial fusion *Mfn2/mouse (139)
cGAS-STING pathway Inhibition of cGAS-STING pathway *Prkn/mouse, *Pink1/mouse (145)

REFERENCES
  1. Smith RA, Hartley RC, Cocheme HM and Murphy MP (2012) Mitochondrial pharmacology. Trends Pharmacol Sci 33, 341-352
    Pubmed CrossRef
  2. Yoo SM and Jung YK (2018) A Molecular Approach to Mitophagy and Mitochondrial Dynamics. Mol Cells 41, 18-26
    Pubmed KoreaMed CrossRef
  3. Suomalainen A and Battersby BJ (2018) Mitochondrial diseases: the contribution of organelle stress responses to pathology. Nat Rev Mol Cell Biol 19, 77-92
    Pubmed CrossRef
  4. Leyns CEG, Ulrich JD, Finn MB et al (2017) TREM2 deficiency attenuates neuroinflammation and protects against neurodegeneration in a mouse model of tauopathy. Proc Natl Acad Sci U S A 114, 11524-11529
    Pubmed KoreaMed CrossRef
  5. Corder EH, Saunders AM, Strittmatter WJ et al (1993) Gene dose of apolipoprotein E type 4 allele and the risk of Alzheimer's disease in late onset families. Science 261, 921-923
    Pubmed CrossRef
  6. Robert J, Button EB, Yuen B et al (2017) Clearance of beta-amyloid is facilitated by apolipoprotein E and circulating high-density lipoproteins in bioengineered human vessels. Elife 6, e29595.
    Pubmed KoreaMed CrossRef
  7. Tai LM, Ghura S, Koster KP et al (2015) APOE-modulated Abeta-induced neuroinflammation in Alzheimer's disease: current landscape, novel data, and future perspective. J Neurochem 133, 465-488
    Pubmed KoreaMed CrossRef
  8. Zhang B, Gaiteri C, Bodea LG et al (2013) Integrated Systems Approach Identifies Genetic Nodes and Networks in Late-Onset Alzheimer’s Disease. Cell 153, 707-720
    Pubmed KoreaMed CrossRef
  9. Griffin WS, Stanley LC, Ling C et al (1989) Brain interleukin 1 and S-100 immunoreactivity are elevated in Down syndrome and Alzheimer disease. Proc Natl Acad Sci U S A 86, 7611-7615
    Pubmed KoreaMed CrossRef
  10. Eikelenboom P and Stam FC (1982) Immunoglobulins and complement factors in senile plaques. An immunoperoxidase study. Acta Neuropathol 57, 239-242
    Pubmed CrossRef
  11. Eriksen JL, Sagi SA, Smith TE et al (2003) NSAIDs and enantiomers of flurbiprofen target -secretase and lower A42 in vivo. J Clin Invest 112, 440-449
    Pubmed KoreaMed CrossRef
  12. Yan Q, Zhang J, Liu H et al (2003) Anti-Inflammatory Drug Therapy Alters -Amyloid Processing and Deposition in an Animal Model of Alzheimer's Disease. J Neurosci 23, 7504-7509
    Pubmed KoreaMed CrossRef
  13. Zandi PP, Anthony JC, Hayden KM, Mehta K, Mayer L and Breitner JCS (2002) Reduced incidence of AD with NSAID but not not H2 receptor antagonists: the Cache County Study. Neurology 59, 880-886
    Pubmed CrossRef
  14. Breitner JC, Welsh KA, Helms MJ et al (1995) Delayed onset of Alzheimer's disease with nonsteroidal antiinflammatory and histamine H2 blocking drugs. Neurobiol Aging 16, 523-530
    Pubmed CrossRef
  15. Wyss-Coray T, Lin C, Yan F et al (2001) TGF-beta1 promotes microglial amyloid-beta clearance and reduces plaque burden in transgenic mice. Nat Med 7, 612-618
    Pubmed CrossRef
  16. Wyss-Coray T, Yan F, Lin AHT et al (2002) Prominent neurodegeneration and increased plaque formation in complement-inhibited Alzheimer's mice. Proc Natl Acad Sci U S A 99, 10837-10842
    Pubmed KoreaMed CrossRef
  17. Liu CC, Hu J, Zhao N et al (2017) Astrocytic LRP1 Mediates Brain Abeta Clearance and Impacts Amyloid Deposition. J Neurosci 37, 4023-4031
    Pubmed KoreaMed CrossRef
  18. Kanekiyo T, Cirrito JR, Liu CC et al (2013) Neuronal clearance of amyloid-beta by endocytic receptor LRP1. J Neurosci 33, 19276-19283
    Pubmed KoreaMed CrossRef
  19. Fu Y, Hsiao JH, Paxinos G, Halliday GM and Kim WS (2016) ABCA7 Mediates Phagocytic Clearance of Amyloid-beta in the Brain. J Alzheimers Dis 54, 569-584
    Pubmed CrossRef
  20. Chakrabarty P, Li A, Ceballos-Diaz C et al (2015) IL-10 alters immunoproteostasis in APP mice, increasing plaque burden and worsening cognitive behavior. Neuron 85, 519-533
    Pubmed KoreaMed CrossRef
  21. Grilli M, Ribola M, Alberici A, Valerio A, Memo M and Spano P (1995) Identification and characterization of a kappa B/Rel binding site in the regulatory region of the amyloid precursor protein gene. J Biol Chem 270, 26774-26777
    Pubmed CrossRef
  22. Cho HJ, Kim SK, Jin SM et al (2007) IFN-gamma-induced BACE1 expression is mediated by activation of JAK2 and ERK1/2 signaling pathways and direct binding of STAT1 to BACE1 promoter in astrocytes. Glia 55, 253-262
    Pubmed CrossRef
  23. Sy M, Kitazawa M, Medeiros R et al (2011) Inflammation induced by infection potentiates tau pathological features in transgenic mice. Am J Pathol 178, 2811-2822
    Pubmed KoreaMed CrossRef
  24. Billups B and Forsythe ID (2002) Presynaptic Mitochondrial Calcium Sequestration Influences Transmission at Mammalian Central Synapses. J Neurosci 22, 5840-5847
    Pubmed KoreaMed CrossRef
  25. Zhou B, Yu P, Lin M-Y, Sun T, Chen Y and Sheng ZH (2016) Facilitation of axon regeneration by enhancing mitochondrial transport and rescuing energy deficits. J Cell Biol 214, 103-119
    Pubmed KoreaMed CrossRef
  26. Tang FL, Liu W, Hu JX et al (2015) VPS35 Deficiency or Mutation Causes Dopaminergic Neuronal Loss by Impairing Mitochondrial Fusion and Function. Cell Rep 12, 1631-1643
    Pubmed KoreaMed CrossRef
  27. Johnson AB and Blum NR (1970) Nucleoside phosphatase activities associated with the tangles and plaques of alzheimer's disease: a histochemical study of natural and experimental neurofibrillary tangles. J Neuropathol Exp Neurol 29, 463-478
    Pubmed CrossRef
  28. Zhang L, Trushin S, Christensen TA et al (2016) Altered brain energetics induces mitochondrial fission arrest in Alzheimer’s Disease. Sci Rep 6, 18725
    Pubmed KoreaMed CrossRef
  29. Gibson GE, Sheu KF, Blass JP et al (1988) Reduced activities of thiamine-dependent enzymes in the brains and peripheral tissues of patients with Alzheimer's disease. Arch Neurol 45, 836-840
    Pubmed CrossRef
  30. Sorbi S, Bird ED and Blass JP (1983) Decreased pyruvate dehydrogenase complex activity in Huntington and Alzheimer brain. Ann Neurol 13, 72-78
    Pubmed CrossRef
  31. Mutisya EM, Bowling AC and Beal MF (1994) Cortical Cytochrome Oxidase Activity Is Reduced in Alzheimer's Disease. J Neurochem 63, 2179-2184
    Pubmed CrossRef
  32. Mecocci P, MacGarvey U and Beal MF (1994) Oxidative damage to mitochondrial DNA is increased in Alzheimer's disease. Ann Neurol 36, 747-751
    Pubmed CrossRef
  33. Reddy PH, Yin X, Manczak M et al (2018) Mutant APP and amyloid beta-induced defective autophagy, mitophagy, mitochondrial structural and functional changes and synaptic damage in hippocampal neurons from Alzheimer’s disease. Hum Mol Genet 27, 2502-2516
    Pubmed KoreaMed CrossRef
  34. Devi L, Prabhu BM, Galati DF, Avadhani NG and Anandatheerthavarada HK (2006) Accumulation of Amyloid Precursor Protein in the Mitochondrial Import Channels of Human Alzheimer’s Disease Brain Is Associated with Mitochondrial Dysfunction. J Neurosci 26, 9057-9068
    Pubmed KoreaMed CrossRef
  35. Lustbader JW, Cirilli M, Lin C et al (2004) ABAD Directly Links A to Mitochondrial Toxicity in Alzheimer's Disease. Science 304, 448-452
    Pubmed CrossRef
  36. Manczak M and Reddy PH (2012) Abnormal interaction of VDAC1 with amyloid beta and phosphorylated tau causes mitochondrial dysfunction in Alzheimer's disease. Hum Mol Genet 21, 5131-5146
    Pubmed KoreaMed CrossRef
  37. Park J, Choi H, Min JS et al (2015) Loss of mitofusin 2 links beta-amyloid-mediated mitochondrial fragmentation and Cdk5-induced oxidative stress in neuron cells. J Neurochem 132, 687-702
    Pubmed CrossRef
  38. Kim DI, Lee KH, Gabr AA et al (2016) A-Induced Drp1 phosphorylation through Akt activation promotes excessive mitochondrial fission leading to neuronal apoptosis. Biochim Biophys Acta 1863, 2820-2834
    Pubmed CrossRef
  39. Fukui H, Diaz F, Garcia S and Moraes CT (2007) Cytochrome c oxidase deficiency in neurons decreases both oxidative stress and amyloid formation in a mouse model of Alzheimer's disease. Proc Natl Acad Sci U S A 104, 14163-14168
    Pubmed KoreaMed CrossRef
  40. Krishnan KJ, Ratnaike TE, De Gruyter HLM, Jaros E and Turnbull DM (2012) Mitochondrial DNA deletions cause the biochemical defect observed in Alzheimer's disease. Neurobiol Aging 33, 2210-2214
    Pubmed CrossRef
  41. Hoekstra JG, Hipp MJ, Montine TJ and Kennedy SR (2016) Mitochondrial DNA mutations increase in early stage Alzheimer disease and are inconsistent with oxidative damage. Ann Neurol 80, 301-306
    Pubmed KoreaMed CrossRef
  42. Coskun PE, Beal MF and Wallace DC (2004) Alzheimer's brains harbor somatic mtDNA control-region mutations that suppress mitochondrial transcription and replication. Proc Natl Acad Sci U S A 101, 10726-10731
    Pubmed KoreaMed CrossRef
  43. Höglinger GU, Lannuzel A, Khondiker ME et al (2005) The mitochondrial complex I inhibitor rotenone triggers a cerebral tauopathy. J Neurochem 95, 930-939
    Pubmed CrossRef
  44. Lopez-Otin C, Blasco MA, Partridge L, Serrano M and Kroemer G (2013) The hallmarks of aging. Cell 153, 1194-1217
    Pubmed KoreaMed CrossRef
  45. Scheibye-Knudsen M, Fang EF, Croteau DL, Wilson DM and Bohr VA (2015) Protecting the mitochondrial powerhouse. Trends Cell Biol 25, 158-170
    Pubmed KoreaMed CrossRef
  46. Hammerling BC and Gustafsson AB (2014) Mitochondrial quality control in the myocardium: Cooperation between protein degradation and mitophagy. J Mol Cell Cardiol 75, 122-130
    Pubmed KoreaMed CrossRef
  47. Cenini G and Voos W (2016) Role of Mitochondrial Protein Quality Control in Oxidative Stress-induced Neurodegenerative Diseases. Curr Alzheimer Res 13, 164-173
    Pubmed CrossRef
  48. Bragoszewski P, Turek M and Chacinska A (2017) Control of mitochondrial biogenesis and function by the ubiquitin - proteasome system. Open Biol 7, 17007
    Pubmed KoreaMed CrossRef
  49. Suliman HB and Piantadosi CA (2016) Mitochondrial Quality Control as a Therapeutic Target. Pharmacol Rev 68, 20-48
    Pubmed CrossRef
  50. Meyer A, Laverny G, Bernardi L et al (2018) Mitochondria: An Organelle of Bacterial Origin Controlling Inflammation. Front Immunol 9, 536
    Pubmed KoreaMed CrossRef
  51. Archibald JM (2015) Endosymbiosis and Eukaryotic Cell Evolution. Curr Biol 25, R911-921
    Pubmed CrossRef
  52. Barbalat R, Ewald SE, Mouchess ML and Barton GM (2011) Nucleic acid recognition by the innate immune system. Annu Rev Immunol 29, 185-214
    Pubmed CrossRef
  53. Contis A, Mitrovic S, Lavie J et al (2017) Neutrophil-derived mitochondrial DNA promotes receptor activator of nuclear factor kappaB and its ligand signalling in rheumatoid arthritis. Rheumatology 56, 1200-1205
    Pubmed CrossRef
  54. Shimada K, Crother TR, Karlin J et al (2012) Oxidized mitochondrial DNA activates the NLRP3 inflammasome during apoptosis. Immunity 36, 401-414
    Pubmed KoreaMed CrossRef
  55. Bai J and Liu F (2019) The cGAS-cGAMP-STING Pathway: A Molecular Link Between Immunity and Metabolism. Diabetes 68, 1099-1108
    Pubmed KoreaMed CrossRef
  56. Dorward DA, Lucas CD, Chapman GB, Haslett C, Dhaliwal K and Rossi AG (2015) The role of formylated peptides and formyl peptide receptor 1 in governing neutrophil function during acute inflammation. Am J Pathol 185, 1172-1184
    Pubmed KoreaMed CrossRef
  57. Dahlgren C, Gabl M, Holdfeldt A, Winther M and Forsman H (2016) Basic characteristics of the neutrophil receptors that recognize formylated peptides, a dangerassociated molecular pattern generated by bacteria and mitochondria. Biochem Pharmacol 114, 22-39
    Pubmed CrossRef
  58. Raoof M, Zhang Q, Itagaki K and Hauser CJ (2010) Mitochondrial peptides are potent immune activators that activate human neutrophils via FPR-1. J Trauma 68, 1328-1332; discussion 1332-1324
    Pubmed CrossRef
  59. Pan ZK, Chen LY, Cochrane CG and Zuraw BL (2000) fMet-Leu-Phe stimulates proinflammatory cytokine gene expression in human peripheral blood monocytes: the role of phosphatidylinositol 3-kinase. J Immunol 164, 404-411
    Pubmed CrossRef
  60. Banoth B and Cassel SL (2018) Mitochondria in innate immune signaling. Transl Res 202, 52-68
    Pubmed KoreaMed CrossRef
  61. Iyer SS, He Q, Janczy JR et al (2013) Mitochondrial Cardiolipin Is Required for Nlrp3 Inflammasome Activation. Immunity 39, 311-323
    Pubmed KoreaMed CrossRef
  62. Chu CT, Bayir H and Kagan VE (2014) LC3 binds externalized cardiolipin on injured mitochondria to signal mitophagy in neurons Implications for Parkinson disease. Autophagy 10, 376-378
    Pubmed KoreaMed CrossRef
  63. Allard B, Longhi MS, Robson SC and Stagg J (2017) The ectonucleotidases CD39 and CD73: Novel checkpoint inhibitor targets. Immunol Rev 276, 121-144
    Pubmed KoreaMed CrossRef
  64. Amores-Iniesta J, Barbera-Cremades M, Martinez CM et al (2017) Extracellular ATP Activates the NLRP3 Inflammasome and Is an Early Danger Signal of Skin Allograft Rejection. Cell Rep 21, 3414-3426
    Pubmed KoreaMed CrossRef
  65. Cauwels A, Rogge E, Vandendriessche B, Shiva S and Brouckaert P (2014) Extracellular ATP drives systemic inflammation, tissue damage and mortality. Cell Death Dis 5, e1102-e1102
    Pubmed KoreaMed CrossRef
  66. Eleftheriadis T, Pissas G, Liakopoulos V and Stefanidis I (2016) Cytochrome c as a Potentially Clinical Useful Marker of Mitochondrial and Cellular Damage. Front Immunol 7, 279
    Pubmed KoreaMed CrossRef
  67. Lin ML, Zhan Y, Projetto AI et al (2008) Selective suicide of cross-presenting CD8(+) dendritic cells by cytochrome c injection shows functional heterogeneity within this subset. Proc Natl Acad Sci U S A 105, 3029-3034
    Pubmed KoreaMed CrossRef
  68. Codina R, Vanasse A, Kelekar A, Vezys V and Jemmerson R (2010) Cytochrome c-induced lymphocyte death from the outside in: inhibition by serum leucine-rich alpha-2-glycoprotein-1. Apoptosis 15, 139-152
    Pubmed CrossRef
  69. Pullerits R, Bokarewa M, Jonsson IM, Verdrengh M and Tarkowski A (2005) Extracellular cytochrome c, a mitochondrial apoptosis-related protein, induces arthritis. Rheumatology 44, 32-39
    Pubmed CrossRef
  70. Mittal M, Siddiqui MR, Tran K, Reddy SP and Malik AB (2014) Reactive Oxygen Species in Inflammation and Tissue Injury. Antioxid Redox Sign 20, 1126-1167
    Pubmed KoreaMed CrossRef
  71. Kozlov AV, Lancaster JR, Meszaros AT and Weidinger A (2017) Mitochondria-meditated pathways of organ failure upon inflammation. Redox Biol 13, 170-181
    Pubmed KoreaMed CrossRef
  72. Naik E and Dixit VM (2011) Mitochondrial reactive oxygen species drive proinflammatory cytokine production. J Exp Med 208, 417-420
    Pubmed KoreaMed CrossRef
  73. Nakahira K, Haspel JA, Rathinam VAK et al (2011) Autophagy proteins regulate innate immune responses by inhibiting the release of mitochondrial DNA mediated by the NALP3 inflammasome. Nat Immunol 12, 222-230
    Pubmed KoreaMed CrossRef
  74. West AP, Khoury-Hanold W, Staron M et al (2015) Mitochondrial DNA stress primes the antiviral innate immune response. Nature 520, 553-557
    Pubmed KoreaMed CrossRef
  75. Tian J, Avalos AM, Mao SY et al (2007) Toll-like receptor 9-dependent activation by DNA-containing immune complexes is mediated by HMGB1 and RAGE. Nat Immunol 8, 487-496
    Pubmed CrossRef
  76. Julian MW, Shao GH, Bao SY et al (2012) Mitochondrial Transcription Factor A Serves as a Danger Signal by Augmenting Plasmacytoid Dendritic Cell Responses to DNA. J Immunol 189, 433-443
    Pubmed KoreaMed CrossRef
  77. Jacobs JL and Coyne CB (2013) Mechanisms of MAVS Regulation at the Mitochondrial Membrane. J Mol Biol 425, 5009-5019
    Pubmed KoreaMed CrossRef
  78. Seth RB, Sun LJ, Ea CK and Chen ZJJ (2005) Identification and characterization of MAVS, a mitochondrial antiviral signaling protein that activates NF-kappa B and IRF3. Cell 122, 669-682
    Pubmed CrossRef
  79. Subramanian N, Natarajan K, Clatworthy MR, Wang Z and Germain RN (2013) The Adaptor MAVS Promotes NLRP3 Mitochondrial Localization and Inflammasome Activation. Cell 153, 348-361
    Pubmed KoreaMed CrossRef
  80. Castanier C, Garcin D, Vazquez A and Arnoult D (2010) Mitochondrial dynamics regulate the RIG-I-like receptor antiviral pathway. EMBO Rep 11, 133-138
    Pubmed KoreaMed CrossRef
  81. Yasukawa K, Oshiumi H, Takeda M et al (2009) Mitofusin 2 Inhibits Mitochondrial Antiviral Signaling. Sci Signal 2, ra47
    Pubmed CrossRef
  82. Tang ED and Wang CY (2009) MAVS Self-Association Mediates Antiviral Innate Immune Signaling. J Virol 83, 3420-3428
    Pubmed KoreaMed CrossRef
  83. Vogel RO, Janssen RJRJ, van den Brand MAM et al (2007) Cytosolic signaling protein Ecsit also localizes to mitochondria where it interacts with chaperone NDUFAF1 and functions in complex I assembly. Gene Dev 21, 615-624
    Pubmed KoreaMed CrossRef
  84. Geng J, Sun XF, Wang P et al (2015) Kinases Mst1 and Mst2 positively regulate phagocytic induction of reactive oxygen species and bactericidal activity. Nat Immunol 16, 1142-1152
    Pubmed KoreaMed CrossRef
  85. Carneiro FRG, Lepelley A, Seeley JJ, Hayden MS and Ghosh S (2018) An Essential Role for ECSIT in Mitochondrial Complex I Assembly and Mitophagy in Macrophages. Cell Rep 22, 2654-2666
    Pubmed KoreaMed CrossRef
  86. Shi HX, Liu X, Wang Q et al (2011) Mitochondrial Ubiquitin Ligase MARCH5 Promotes TLR7 Signaling by Attenuating TANK Action. PLoS Pathog 7, e1002057
    Pubmed KoreaMed CrossRef
  87. Wilkins HM, Carl SM, Greenlief ACS, Festoff BW and Swerdlow RH (2014) Bioenergetic Dysfunction and Inflammation in Alzheimer’s Disease: A Possible Connection. Front Aging Neurosci 6, 311
    Pubmed KoreaMed CrossRef
  88. Wilkins HM, Weidling IW, Ji Y and Swerdlow RH (2017) Mitochondria-Derived Damage-Associated Molecular Patterns in Neurodegeneration. Front Immunol 8, 508
    Pubmed KoreaMed CrossRef
  89. Bajwa E, Pointer CB and Klegeris A (2019) The Role of Mitochondrial Damage-Associated Molecular Patterns in Chronic Neuroinflammation. Mediators Inflammation 2019, 4050796
    Pubmed KoreaMed CrossRef
  90. Wilkins HM, Koppel SJ, Weidling IW et al (2016) Extracellular Mitochondria and Mitochondrial Components Act as Damage-Associated Molecular Pattern Molecules in the Mouse Brain. J Neuroimmune Pharmacol 11, 622-628
    Pubmed KoreaMed CrossRef
  91. Guerreiro R, Wojtas A, Bras J et al (2013) TREM2 variants in Alzheimer's disease. N Engl J Med 368, 117-127
    Pubmed KoreaMed CrossRef
  92. Korvatska O, Leverenz JB, Jayadev S et al (2015) R47H Variant of TREM2 Associated With Alzheimer Disease in a Large Late-Onset Family: Clinical, Genetic, and Neuropathological Study. JAMA Neurol 72, 920-927
    Pubmed KoreaMed CrossRef
  93. Wang Y, Cella M, Mallinson K et al (2015) TREM2 lipid sensing sustains the microglial response in an Alzheimer's disease model. Cell 160, 1061-1071
    Pubmed KoreaMed CrossRef
  94. Turnbull IR, Gilfillan S, Cella M et al (2006) Cutting edge: TREM-2 attenuates macrophage activation. J Immunol 177, 3520-3524
    Pubmed CrossRef
  95. Jiang T, Zhang YD, Chen Q et al (2016) TREM2 modifies microglial phenotype and provides neuroprotection in P301S tau transgenic mice. Neuropharmacology 105, 196-206
    Pubmed CrossRef
  96. Podlesniy P, Figueiro-Silva J, Llado A et al (2013) Low cerebrospinal fluid concentration of mitochondrial DNA in preclinical Alzheimer disease. Ann Neurol 74, 655-668
    Pubmed CrossRef
  97. Thubron EB, Rosa HS, Hodges A et al (2019) Regional mitochondrial DNA and cell-type changes in post-mortem brains of non-diabetic Alzheimer’s disease are not present in diabetic Alzheimer’s disease. Sci Rep 9, 11386
    Pubmed KoreaMed CrossRef
  98. Ruggiero FM, Cafagna F, Petruzzella V, Gadaleta MN and Quagliariello E (1992) Lipid composition in synaptic and nonsynaptic mitochondria from rat brains and effect of aging. J Neurochem 59, 487-491
    Pubmed CrossRef
  99. Pointer CB and Klegeris A (2017) Cardiolipin in Central Nervous System Physiology and Pathology. Cell Mol Neurobiol 37, 1161-1172
    Pubmed CrossRef
  100. Petrosillo G, Matera M, Casanova G, Ruggiero FM and Paradies G (2008) Mitochondrial dysfunction in rat brain with aging Involvement of complex I, reactive oxygen species and cardiolipin. Neurochem Int 53, 126-131
    Pubmed CrossRef
  101. Perier C, Tieu K, Guegan C et al (2005) Complex I deficiency primes Bax-dependent neuronal apoptosis through mitochondrial oxidative damage. Proc Natl Acad Sci U S A 102, 19126-19131
    Pubmed KoreaMed CrossRef
  102. Little JP, Simtchouk S, Schindler SM et al (2014) Mitochondrial transcription factor A (Tfam) is a proinflammatory extracellular signaling molecule recognized by brain microglia. Mol Cell Neurosci 60, 88-96
    Pubmed CrossRef
  103. Schindler SM, Frank MG, Annis JL, Maier SF and Klegeris A (2018) Pattern recognition receptors mediate pro-inflammatory effects of extracellular mitochondrial transcription factor A (TFAM). Mol Cell Neurosci 89, 71-79
    Pubmed CrossRef
  104. Julian MW, Shao G, Vangundy ZC, Papenfuss TL and Crouser ED (2013) Mitochondrial transcription factor A, an endogenous danger signal, promotes TNF release via RAGE- and TLR9-responsive plasmacytoid dendritic cells. PLoS One 8, e72354-e72354
    Pubmed KoreaMed CrossRef
  105. Verdier Y, Zarandi M and Penke B (2004) Amyloid beta-peptide interactions with neuronal and glial cell plasma membrane: binding sites and implications for Alzheimer's disease. J Pept Sci 10, 229-248
    Pubmed CrossRef
  106. Xie J, Mendez JD, Mendez-Valenzuela V and Aguilar-Hernandez MM (2013) Cellular signalling of the receptor for advanced glycation end products (RAGE). Cell Signal 25, 2185-2197
    Pubmed CrossRef
  107. Lue LF, Walker DG, Brachova L et al (2001) Involvement of microglial receptor for advanced glycation endproducts (RAGE) in Alzheimer's disease: identification of a cellular activation mechanism. Exp Neurol 171, 29-45
    Pubmed CrossRef
  108. Papaliagkas V, Anogeianakis G, Tsolaki M, Koliakos G and Kimiskidis V (2009) Prediction of Conversion from Mild Cognitive Impairment to Alzheimer’s Disease by CSF Cytochrome c Levels and N200 Latency. Curr Alzheimer Res 6, 279-284
    Pubmed CrossRef
  109. Takuma K, Yan SS, Stern DM and Yamada K (2005) Mitochondrial dysfunction, endoplasmic reticulum stress, and apoptosis in Alzheimer's disease. J Pharmacol Sci 97, 312-316
    Pubmed CrossRef
  110. Krysko DV, Agostinis P, Krysko O et al (2011) Emerging role of damage-associated molecular patterns derived from mitochondria in inflammation. Trends Immunol 32, 157-164
    Pubmed CrossRef
  111. Gouveia A, Bajwa E and Klegeris A (2017) Extracellular cytochrome c as an intercellular signaling molecule regulating microglial functions. Biochim Biophys Acta Gen Subj 1861, 2274-2281
    Pubmed CrossRef
  112. Oyewole AO and Birch-Machin MA (2015) Mitochondriatargeted antioxidants. FASEB J 29, 4766-4771
    Pubmed CrossRef
  113. Jauslin ML, Meier T, Smith RA and Murphy MP (2003) Mitochondria-targeted antioxidants protect Friedreich Ataxia fibroblasts from endogenous oxidative stress more effectively than untargeted antioxidants. FASEB J 17, 1972-1974
    Pubmed CrossRef
  114. Gioscia-Ryan RA, LaRocca TJ, Sindler AL, Zigler MC, Murphy MP and Seals DR (2014) Mitochondria-targeted antioxidant (MitoQ) ameliorates age-related arterial endothelial dysfunction in mice. J Physiol 592, 2549-2561
    Pubmed KoreaMed CrossRef
  115. Jin H, Kanthasamy A, Ghosh A, Anantharam V, Kalyanaraman B and Kanthasamy AG (2014) Mitochondriatargeted antioxidants for treatment of Parkinson's disease: preclinical and clinical outcomes. Biochimica et biophysica acta 1842, 1282-1294
    Pubmed KoreaMed CrossRef
  116. Dashdorj A, Jyothi KR, Lim S et al (2013) Mitochondriatargeted antioxidant MitoQ ameliorates experimental mouse colitis by suppressing NLRP3 inflammasomemediated inflammatory cytokines. BMC Med 11, 178
    Pubmed KoreaMed CrossRef
  117. Asano T, Koike M, Sakata S et al (2015) Possible involvement of iron-induced oxidative insults in neurodegeneration. Neurosci Lett 588, 29-35
    Pubmed CrossRef
  118. Mena NP, Urrutia PJ, Lourido F, Carrasco CM and Nunez MT (2015) Mitochondrial iron homeostasis and its dysfunctions in neurodegenerative disorders. Mitochondrion 21, 92-105
    Pubmed CrossRef
  119. Thomsen MS, Andersen MV, Christoffersen PR, Jensen MD, Lichota J and Moos T (2015) Neurodegeneration with inflammation is accompanied by accumulation of iron and ferritin in microglia and neurons. Neurobiol Dis 81, 108-118
    Pubmed CrossRef
  120. Smigrodzki RM and Khan SM (2005) Mitochondrial microheteroplasmy and a theory of aging and age-related disease. Rejuvenation Res 8, 172-198
    Pubmed CrossRef
  121. Casoli T, Spazzafumo L, Di Stefano G and Conti F (2015) Role of diffuse low-level heteroplasmy of mitochondrial DNA in Alzheimer's disease neurodegeneration. Front Aging Neurosci 7, 142-142
    Pubmed KoreaMed CrossRef
  122. Onyango IG (2018) Modulation of mitochondrial bioenergetics as a therapeutic strategy in Alzheimer's disease. Neural Regen Res 13, 19-25
    Pubmed KoreaMed CrossRef
  123. Jo A, Ham S, Lee GH et al (2015) Efficient Mitochondrial Genome Editing by CRISPR/Cas9. Biomed Res Int 2015, 305716
    Pubmed KoreaMed CrossRef
  124. Hashimoto M, Bacman SR, Peralta S et al (2015) MitoTALEN: A General Approach to Reduce Mutant mtDNA Loads and Restore Oxidative Phosphorylation Function in Mitochondrial Diseases. Mol Ther 23, 1592-1599
    Pubmed KoreaMed CrossRef
  125. Zhong Y, Hu YJ, Chen B et al (2011) Mitochondrial transcription factor A overexpression and base excision repair deficiency in the inner ear of rats with D-galactose-induced aging. FEBS J 278, 2500-2510
    Pubmed CrossRef
  126. Hayashi Y, Yoshida M, Yamato M et al (2008) Reverse of age-dependent memory impairment and mitochondrial DNA damage in microglia by an overexpression of human mitochondrial transcription factor a in mice. J Neurosci 28, 8624-8634
    Pubmed KoreaMed CrossRef
  127. Xu S, Zhong M, Zhang L et al (2009) Overexpression of Tfam protects mitochondria against beta-amyloidinduced oxidative damage in SH-SY5Y cells. FEBS J 276, 3800-3809
    Pubmed CrossRef
  128. Oka S, Leon J, Sakumi K et al (2016) Human mitochondrial transcriptional factor A breaks the mitochondria-mediated vicious cycle in Alzheimer's disease. Sci Rep 6, 37889
    Pubmed KoreaMed CrossRef
  129. Heneka MT, Kummer MP, Stutz A et al (2013) NLRP3 is activated in Alzheimer's disease and contributes to pathology in APP/PS1 mice. Nature 493, 674-678
    Pubmed KoreaMed CrossRef
  130. Daniels MJ, Rivers-Auty J, Schilling T et al (2016) Fenamate NSAIDs inhibit the NLRP3 inflammasome and protect against Alzheimer's disease in rodent models. Nat Commun 7, 12504
    Pubmed KoreaMed CrossRef
  131. Dempsey C, Rubio Araiz A, Bryson KJ et al (2017) Inhibiting the NLRP3 inflammasome with MCC950 promotes non-phlogistic clearance of amyloid-beta and cognitive function in APP/PS1 mice. Brain Behav Immun 61, 306-316
    Pubmed CrossRef
  132. Yin J, Zhao F, Chojnacki JE et al (2018) NLRP3 Inflammasome Inhibitor Ameliorates Amyloid Pathology in a Mouse Model of Alzheimer's Disease. Mol Neurobiol 55, 1977-1987
    Pubmed KoreaMed CrossRef
  133. Yang Y, Wang H, Kouadir M, Song H and Shi F (2019) Recent advances in the mechanisms of NLRP3 inflammasome activation and its inhibitors. Cell Death Dis 10, 128
    Pubmed KoreaMed CrossRef
  134. Lautrup S, Lou G, Aman Y, Nilsen H, Tao J and Fang EF (2019) Microglial mitophagy mitigates neuroinflammation in Alzheimer's disease. Neurochem Int 129, 104469
    Pubmed CrossRef
  135. Fang EF, Hou Y, Palikaras K et al (2019) Mitophagy inhibits amyloid- and tau pathology and reverses cognitive deficits in models of Alzheimer’s disease. Nat Neurosci 22, 401-412
    Pubmed KoreaMed CrossRef
  136. Lei Q, Tan J, Yi S, Wu N, Wang Y and Wu H (2018) Mitochonic acid 5 activates the MAPK-ERK-yap signaling pathways to protect mouse microglial BV-2 cells against TNFalpha-induced apoptosis via increased Bnip3-related mitophagy. Cell Mol Biol Lett 23, 14
    Pubmed KoreaMed CrossRef
  137. Zhou R, Yazdi AS, Menu P and Tschopp J (2011) A role for mitochondria in NLRP3 inflammasome activation. Nature 469, 221-225
    Pubmed CrossRef
  138. Jiang S, Nandy P, Wang W et al (2018) Mfn2 ablation causes an oxidative stress response and eventual neuronal death in the hippocampus and cortex. Mol Neurodegener 13, 5
    Pubmed KoreaMed CrossRef
  139. Park J, Choi H, Min JS et al (2013) Mitochondrial dynamics modulate the expression of pro-inflammatory mediators in microglial cells. J Neurochem 127, 221-232
    Pubmed CrossRef
  140. Kim H, Lee JY, Park KJ, Kim W-H and Roh GS (2016) A mitochondrial division inhibitor, Mdivi-1, inhibits mitochondrial fragmentation and attenuates kainic acid-induced hippocampal cell death. BMC Neurosci 17, 33
    Pubmed KoreaMed CrossRef
  141. Joshi AU, Minhas PS, Liddelow SA et al (2019) Fragmented mitochondria released from microglia trigger A1 astrocytic response and propagate inflammatory neurodegeneration. Nat Neurosci 22, 1635-1648
    Pubmed KoreaMed CrossRef
  142. Akhter F, Chen D, Yan SF and Yan SS (2017) Mitochondrial Perturbation in Alzheimer's Disease and Diabetes. Prog Mol Biol Transl Sci 146, 341-361
    Pubmed KoreaMed CrossRef
  143. Barber GN (2014) STING-dependent cytosolic DNA sensing pathways. Trends Immunol 35, 88-93
    Pubmed CrossRef
  144. Liang Q, Seo GJ, Choi YJ et al (2014) Crosstalk between the cGAS DNA sensor and Beclin-1 autophagy protein shapes innate antimicrobial immune responses. Cell Host Microbe 15, 228-238
    Pubmed KoreaMed CrossRef
  145. Sliter DA, Martinez J, Hao L et al (2018) Parkin and PINK1 mitigate STING-induced inflammation. Nature 561, 258-262
    Pubmed KoreaMed CrossRef


This Article


Cited By Articles

Funding Information

Collections

Services
Social Network Service

e-submission

Archives