Skip to main content

The neuroimmune nexus: unraveling the role of the mtDNA-cGAS-STING signal pathway in Alzheimer’s disease

Abstract

The relationship between Alzheimer's disease (AD) and neuroimmunity has gradually begun to be unveiled. Emerging evidence indicates that cyclic GMP-AMP synthase (cGAS) acts as a cytosolic DNA sensor, recognizing cytosolic damage-associated molecular patterns (DAMPs), and inducing the innate immune response by activating stimulator of interferon genes (STING). Dysregulation of this pathway culminates in AD-related neuroinflammation and neurodegeneration. A substantial body of evidence indicates that mitochondria are involved in the critical pathogenic mechanisms of AD, whose damage leads to the release of mitochondrial DNA (mtDNA) into the extramitochondrial space. This leaked mtDNA serves as a DAMP, activating various pattern recognition receptors and immune defense networks in the brain, including the cGAS-STING pathway, ultimately leading to an imbalance in immune homeostasis. Therefore, modulation of the mtDNA-cGAS-STING pathway to restore neuroimmune homeostasis may offer promising prospects for improving AD treatment outcomes. In this review, we focus on the mechanisms of mtDNA release during stress and the activation of the cGAS-STING pathway. Additionally, we delve into the research progress on this pathway in AD, and further discuss the primary directions and potential hurdles in developing targeted therapeutic drugs, to gain a deeper understanding of the pathogenesis of AD and provide new approaches for its therapy.

Background

Alzheimer’s disease (AD) is the most common form of chronic neurodegenerative disease, posing significant health risks to the elderly [1]. Clinically, it primarily manifests as a progressive impairment of memory and cognitive function [2]. Analysis of epidemiological data has indicated that the global prevalence of dementia is expected to triple by 2050, imposing heavy economic and psychological challenges on communities and households [3,4,5]. The primary pathological features of AD involve the widespread amyloid plaques and neurofibrillary tangles (NFT) within the brain, triggering irreversible neuronal damage and cognitive decline [6]. Despite significant global financial investments for tackling AD, effective treatment strategies are still lacking [7]. One possible reason for this is that the etiology of AD is multifactorial [8], and the precise molecular mechanisms underlying its onset and progression remain largely elusive. As such, there is an urgent need to explore new disease-modifying therapies that target the critical processes of AD pathogenesis [9], with the aim of obtaining better treatment outcomes. Recent studies have identified the involvement of maladaptive inflammatory responses and immune dysregulation in AD development [10, 11], highlighting the importance of understanding the potential molecular mechanisms underlying immune-inflammatory reactions.

The innate immune, serving as the first line of defense in the body’s immune system, can distinguish between “self” and “non-self” effectively [12]. Endogenous damage-associated molecular patterns (DAMPs) released after cellular damage, such as self-proteins or nucleic acids, can also be recognized by the innate immune system, thereby eliciting a swift and non-specific immune response [13]. This recognition process is mediated by pattern recognition receptors (PRRs), the majority of which possess sequence- and cell-type- specificity [14]. Notably, as a unique type of PRRs, cyclic GMP-AMP synthase (cGAS) lacks DNA sequence specificity, and cannot effectively distinguish between self and foreign DNA [15]. Consequently, mitochondrial DNA (mtDNA) and other double-stranded DNA (dsDNA) can act as DAMPs to bind cGAS upon entry into the cytoplasm, thus activating the downstream stimulator of interferon genes (STING) [16]. Typically, the cGAS-STING pathway serves as a protective mechanism against invading pathogens, and its activation by external nucleic acids, such as those from viruses, can effectively protect the cells from potential threats [17]. However, overactivation of this pathway, triggered by the accumulation of ectopic dsDNA in the central nervous system (CNS), may exacerbate neuroinflammatory responses. This dysregulated inflammatory environment likely further impairs neuronal cells, causing damage to healthy tissues and potentially triggering multiple neurological diseases that have been reported to be associated with abnormal inflammation, including AD, Parkinson’s disease (PD) [18], amyotrophic lateral sclerosis (ALS) [19], postoperative cognitive dysfunction (POCD) [20], and traumatic brain encephalopathy (TBE) [21], although the pathogenesis of these diseases shows high heterogeneity. Recent studies particularly highlight its critical role in AD.

In this review, we summarize how mtDNA is released into extramitochondrial space under cellular stress, and subsequently recognized by the cGAS-STING signal pathway. Furthermore, we discuss the pivotal role of the mtDNA-cGAS-STING pathway in AD and the prospects of targeting this pathway for AD treatment. We believe that an in-depth exploration of the mechanisms related to this cascade pathway will provide greater hope for delaying AD progression.

Mechanisms of mtDNA release

In mammalian cells, mtDNA is a double-stranded, circular DNA comprising 16,569 base pairs and encoding 37 genes [22]. mtDNA is distributed throughout the mitochondrial network, and the proteins it encodes are involved in a wide range of physiological functions [23], including energy supply, Ca2+ homeostasis maintenance, synaptic plasticity [24], and heme and cholesterol synthesis [25]. Notably, mtDNA itself exerts an endogenous pro-inflammatory effect, which is largely attributed to its unmethylated CpG islands, akin to bacterial genomes [26, 27]. As such, mtDNA released outside of mitochondria can act as an immune stimulant to mediate immune-inflammatory processes. Nevertheless, mtDNA possesses an intricate set of self-protection systems that protect it from recognition by the immune surveillance system under physiological conditions, thus maintaining its relatively stable existence within cells. These mechanisms encompass the barrier of the mitochondrial “double-layer” membrane, balanced mitochondrial dynamics, coordinated regulation of various nuclear genes, mitochondrial autophagy, and interplay with other cellular organelles [28,29,30]. Abnormalities in these mechanisms may lead to mtDNA mutations or relocation outside the mitochondria, although the specific release process and related regulatory mechanisms remain unclear. Interestingly, the mechanisms underlying mtDNA releasing to extramitochondrial space differ between apoptotic and living cells; in cells under apoptotic stress, mtDNA escape predominantly occurs through macropores formed by apoptotic regulators (BAK/BAX) [31, 32], including in senescent cells that remain resistant to apoptosis, where mtDNA can also be released via BAK/BAX-dependent minority MOMP (miMOMP) [33]. Whereas in living cells, mtDNA primarily depends on voltage-dependent anion channel (VDAC) oligomerization pores in the outer mitochondrial membrane (OMM) that promote mitochondrial outer membrane permeabilization (MOMP) [34, 35]. Besides, gasdermin pores, mitochondrial dynamics, extracellular vesicles (EVs), and neutrophil extracellular traps (NETs) also contribute to mtDNA release [36]. As such, mtDNA escape is an exceedingly complex process involving an interplay of multiple mechanisms. In the following sections, we systematically summarize these mechanisms of mtDNA release to better understand the role of mtDNA in inflammatory diseases and immune regulation.

The mechanism of mtDNA release into the extramitochondrial space

BAK/BAX pores

The prevailing view supports the notion that MOMP is the primary trigger of mtDNA release. When apoptotic signals occur within cells, the pro-apoptotic factors BAX and BAK are activated and recruited to oligomerize on the OMM, forming macropores that induce MOMP [37]. The growth rate of these macropores and the relative kinetics of mtDNA are influenced by the relative availability of Bax and Bak, while their interactions can regulate the dynamics of mtDNA release [38, 39]. Unlike non-inflammatory mitochondrial apoptosis, which relies on caspase activation, mtDNA release through these macropores does not require activated caspases [32]. The absence of the latter is crucial for mtDNA-mediated inflammation. If caspases are activated, cell death occurs after MOMP, without triggering the type I interferon (IFN) response [40, 41]. In other words, caspase activation can sequester the apoptotic process of cells from the immune system, thus avoiding unnecessary inflammatory responses. This precise regulatory mechanism ensures the orderly progression of apoptosis, which is important for maintaining immune homeostasis in organisms.

Following MOMP, the inner mitochondrial membrane (IMM) protrudes into the cytoplasm through BAK/BAX macropores to form mitochondrial herniations. Then, the mitochondrial matrix contents, including the mitochondrial genome and other mitochondrial components, are translocated outside the mitochondria [31], subsequently activating caspase-independent cell death (CICD)-associated inflammation. This process may further result in a loss of IMM integrity, culminating in mitochondrial inner membrane permeabilization (MIMP) [32]. Additionally, studies have indicated that mtDNA is released into the cytoplasm in the form of entire nucleoids. However, whether these released nucleoids remain intact or fragmented remains unclear [42]. Clarifying the fate and effects of these released nucleoids within the cytoplasm is crucial, as they may be closely linked to the potential of mtDNA as an effective biomarker.

Notably, MOMP may also be associated with the release of mitochondrial double-stranded RNA (mt-dsRNA) [43]. Dhir et al. found that the release of mt-dsRNA is almost entirely blocked by the downregulation of BAX and BAK, indicating that mt-dsRNA release may predominantly occur via MOMP. These released mt-dsRNAs can also be recognized by dsRNA sensors and activate the IFN-I pathway [44]. Furthermore, they may act as mitochondrial self-antigens involved in the development of autoimmune diseases [45]. Nevertheless, the mechanisms underlying mt-dsRNA release remain poorly understood, and their precise role in immune responses requires further investigation.

VDAC oligomer pores

In living cells, the mechanism by which mtDNA is released is not uniform. Multiple studies have indicated that oxidative stress, when insufficient to activate BAK and BAX, can induce VDAC oligomerization and subsequently form pores in the OMM, thus promoting MOMP in living cells and facilitating mtDNA release [34]. Notably, mtDNA translocation through VDAC requires the coordination of other proteins, particularly the adenine nucleotide translocator (ANT) located in the IMM, and Cyprin D in the matrix. These proteins are considered key components of the mitochondrial permeability transition pore (mPTP), a diverse protein complex located between the IMM and OMM [46, 47]. García et al. provided evidence to support this view, showing that cyclosporin A, a Cyprin D inhibitor, could affect the permeability transition pore and inhibit mtDNA release into the cytoplasm by 52% [48]. Unlike the BAK/BAX pores, mtDNA fragments, which are components of specific genes, are released via VDAC pores [49]. In addition, mPTP pores are expected to allow only mtDNA fragments with a molecular weight of less than 1.5 kDa to pass through [50], enriching evidence that the mtDNA released via VDAC pores are fragments rather than intact nucleoids.

The formation of VDAC oligomer pores is a complex process regulated by several factors. Upon entry into the mitochondrial intermembrane space, mtDNA fragments themselves interact with residues at the N-terminus of the VDAC1 subtype, thus promoting VDAC oligomerization and forming a feed-forward loop [34]. The virus-related kinase 2 (VRK2) protein, a member of the vaccinia virus-related kinase family and a serine/threonine kinase with catalytic activity [51, 52], is considered another important regulator of VDAC1 oligomerization during mtDNA release. VRK2 was recently shown to induce cGAS-mediated innate immune response [53], indicating its potential as a therapeutic target for infectious and autoimmune diseases associated with mtDNA release. Additionally, Baik et al. demonstrated that the dissociation of hexokinase 2 (HK2) from VDAC triggers calcium release from the endoplasmic reticulum (ER), which is subsequently taken up by the mitochondria, leading to VDAC oligomerization and mtDNA escape [54]. However, these studies only partly elucidated the regulatory mechanisms governing mtDNA release through VDAC oligomeric pores, and their findings are likely limited to specific disease contexts. Further research is therefore needed to investigate whether there are differences in the regulation of mtDNA release through VDAC oligomeric pores under various disease conditions such as infection, autoimmune diseases, and cancer, which will help to identify specific therapeutic targets to provide more possibilities for the treatment of these related diseases.

Gasdermin pores

Gasdermins (GSDMs) are a class of pore-forming proteins that represent another channel for mtDNA escape from mitochondria [55]. As a pivotal effector molecule in pyroptosis, gasdermin undergoes cleavage by activated caspases to generate an N-terminal domain, that inserts into the plasma membrane and oligomerizes to form large pores through its pore-forming activity [56], allowing the release of inflammatory substances like interleukin-1β (IL-1β) [57]. Huang et al. indicated that mtDNA can be released into the cytosol through gasdermin pores to activate downstream signaling pathways during inflammatory injury [58]. In detail, several members of the gasdermins, including gasdermin A3, gasdermin D (GSDMD), and gasdermin E, can facilitate mtDNA release during apoptosis or pyroptosis, and inhibit the pore activity of GSDMD significantly impedes this process, consequently ameliorating mitochondrial network structure damage and rescuing mitochondrial dysfunction [55, 59]. Additionally, oxidized mtDNA can directly interact with the GSDMD-N domain to enhance its oligomerization during the pore-forming process [60], which may exacerbate mitochondrial membrane rupture. Furthermore, membrane damage induced by gasdermin could precipitate the collapse of the mitochondrial network system, potentially providing alternative pathways for mtDNA release, such as NETs [61], and ultimately resulting in the displacement of mtDNA outside the mitochondria or even extracellularly. These studies indicate a plausible interplay between gasdermin and damaged mitochondria, both promoting cell death. Overall, the formation of gasdermin pores is a significant determinant of mtDNA escape, extending its importance beyond pyroptosis.

The mechanism and significance of mtDNA release into the extracellular space

Diverse mtDNA forms have been detected in different biological fluids, such as plasma [62], serum [63], cerebrospinal fluid (CSF) [64], and synovial fluid [65], indicating the further release of mtDNA from the cytoplasmic compartment into the extracellular environment. Currently, the active release of mtDNA is believed to occur primarily through transportation via extracellular EVs or as components of NETs, whereas its passive release typically occurs during cell injury and death processes like apoptosis, necrosis, and pyroptosis [25]. Recent studies have suggested that the level of cell-free mtDNA (cf-mtDNA) released into the extracellular fluid is a significant biomarker that partly reflects the severity and prognosis of various inflammation-related diseases. For example, a correlation has been observed between higher mtDNA copy numbers (mtDNAcn) in circulation and better cognitive function [66]. Similarly, patients with coronary heart disease have lower mtDNA levels than the control group [67]. Interestingly, evidence suggests that only cf-mtDNA, and not mtDNA present in intact circulating mitochondria, can elicit an inflammatory response [68]. Understanding the mechanisms underlying mtDNA releasing into extracellular space and the pathophysiological functions of its various forms is essential to advance our knowledge of mitochondrial biology and its roles in health and disease.

Activation of cGAS-STING by Leaked mtDNA

As mentioned above, under physiological circumstances, mtDNA is encapsulated within the “double membrane” structure of mitochondria and does not disturb the immune system. Upon release into the cytoplasm or circulation, it acts as a potential immune stimulus, eliciting an inherent immune response in the body. Although the precise mechanisms by which mtDNA triggers inflammation have not yet been fully elucidated, it is widely recognized that mtDNA primarily affects immune responses through three important signaling pathways: the toll-like receptor-9 (TLR-9), cGAS-STING, and nucleotide-binding oligomerization domain (NOD), leucine-rich repeat (LRR), and pyrin domain-containing protein 3 (NLRP3) inflammasome pathways [69] (Fig. 1). Given that the association of TLR-9 and NLRP3 with mtDNA has been extensively summarized in many other excellent reviews [25, 70], herein, we primarily focused on the crosstalk between mtDNA and the emerging immune pathway cGAS-STING, which is increasingly being recognized for its role in AD.

Fig. 1
figure 1

Overview of the mechanisms of mtDNA release and its effect on the innate immune response. Under normal circumstances, mtDNA is encapsulated within mitochondria without disturbing the immune system. However, when exposed to various adverse factors such as ROS, cellular stress, and aging, mitochondria inevitably incur a certain degree of damage. At this point, mtDNA is released into the extramitochondrial space through mechanisms involving BAK/BAX, VDAC1, GSDMD, and others. Subsequently, the leaked mtDNA acts as DAMPs, recognized by diverse PRRs, primarily activating the body’s innate immune response through three different inflammatory pathways: TLR9, NLPR3, and cGAS-STING

Molecular mechanisms of the cGAS-STING pathway

Activation of the cGAS-STING pathway is an extremely complex process (Fig. 2). The molecular weight of cGAS is approximately 60 kDa, and it comprises an N-terminal region that mediates binding to cell membrane, a highly conserved nucleotidyltransferase core domain, and a male abnormal gene family 21 (Mab21) nucleotidyltransferase domain [71, 72]. The Mab21 domain is primarily involved in dsDNA binding, and structural defects in it cause cGAS to lose the ability to induce IFN-β expression [73]. Thus, maintaining structural integrity is especially important for the physical function of cGAS. Unlike other PRRs, cGAS binds to negatively charged dsDNA through electrostatic interactions and hydrogen bonds, which partly explains its lack of sequence-specific recognition [74, 75]. In other words, it can bind to various types of nucleic acids, such as dsDNA, RNA:DNA hybrids, ssDNA, and dsRNA, but the latter two lack the ability to rearrange the catalytic pocket of cGAS to activate it [76, 77]. In addition, the intracellular localization of cGAS remains controversial. Although most studies consider it a “cytoplasmic sensor” [78], recent research indicated that cGAS is present in the cell nucleus and membrane as well [79,80,81,82]. For example, the localization of cGAS is cell-cycle dependent [83, 84]; it is typically located in the cytoplasm of non-dividing cells but translocates into the nucleus during mitosis in proliferating cells, where it binds to chromatin DNA [85]. In summary, its subcellular localization is tightly related to the cell types and biological contexts, allowing it to function more effectively.

Fig. 2
figure 2

Overview of the molecular mechanisms of the cGAS-STING signal pathway. The activation of the cGAS-STING signaling begins with the detection of abnormal dsDNA within the cell. Initially, dsDNA binds to cGAS, triggering its enzymatic activity and the production of 2’3’-cGAMP. Subsequently, cGAMP binds to STING on the ER membrane, inducing conformational changes that result in STING oligomerization and translocation to the ERGIC and Golgi apparatus. This change leads to the activation of signaling molecules such as TBK1 and IRF3, which enter the nucleus and initiate the transcription of immune factors like interferons, ultimately initiating the body’s immune response to combat infection and damage

Indeed, cGAS possesses catalytic inertness. When it binds to dsDNA, a conformational change occurs, converting the catalytic pocket of cGAS from an inactive “closed” conformation to an active “open” conformation, forming a 2:2 dimeric structure composed of cGAS and dsDNA [75, 86]. Subsequently, this complex catalyzes the formation of 2’3’-cyclic GMP-AMP (2’3’-cGAMP) from guanosine triphosphate (GTP) and adenosine triphosphate (ATP), which serves as a second messenger, binding and activating the adaptor protein STING located on the ER membrane, initiating a cascade of intracellular signaling events crucial for the immune response [87].

Ishikawa et al. first reported the importance of STING in the innate immune response to antiviral immunity. As an important component of nonspecific immunity against aberrant cytoplasmic DNA, STING is widely expressed in the ER across various cell types [88]. Upon binding to 2’3’-cGAMP, STING oligomerizes and is trafficked from the ER to the Golgi apparatus [89, 90]. During this process, the C-terminal tail (CCT) of STING recruits and activates TANK-binding kinase 1 (TBK1), which phosphorylates STING and interferon regulatory factor 3 (IRF3) [91]. Subsequently, phosphorylated IRF3 forms dimers and is translocated to the nucleus to bind IFN response elements, inducing extensive transcription of downstream type I interferons (IFN-I) and interferon-stimulated genes (ISGs), thus triggering IFN-I-mediated immune responses [92]. Additionally, STING can also phosphorylate IκB kinase (IKK), causing the release of nuclear factor kappa-B (NF-κB) into the nucleus, activating the classical NF-κB signaling pathway, and inducing the expression of genes such as tumor necrosis factor-alpha (TNF-α), interleukin-6 (IL-6), and IL-1β, triggering a broad immune response [93].

cGAS-STING-mediated autophagy represents the primitive and highly conserved function of this pathway [94]. Upon binding with cGAMP, STING buds from the ER to form COPII vesicles, forming the ER-Golgi intermediate compartment (ERGIC). The ERGIC-containing STING serves as a membrane source for microtubule-associated protein 1 light chain 3 (LC3) lipidation, promoting the formation of autophagosomes [95, 96]. In contrast, autophagy can feedback-regulate the activity of this pathway as well [97]. In addition to negatively regulating STING signaling by interfering with the assembly of STING-TBK1-IRF3 or STING degradation, autophagy prevents STING activation by delivering cytoplasmic DNA to lysosomal degradation [95, 98, 99]. The reciprocal regulation between the cGAS-STING pathway and autophagy helps clear damaged organelles, abnormal proteins, and accumulated dsDNA in the cytoplasm, suppressing unnecessary inflammatory responses and supporting cellular homeostasis.

Crosstalk between cGAS-STING and mtDNA

The interaction between cGAS-STING and mtDNA and its involvement in disease pathogenesis has long been a focal point of research. A pivotal discovery linking mtDNA to the cGAS-STING pathway demonstrates that mtDNA stress is an intrinsic trigger of antiviral responses. Specifically speaking, viral infection can lead to abnormal mtDNA escape into the cytosol, where it binds to cGAS and promotes STING-IRF3-dependent signaling, mediating antiviral innate immunity [100]. Other studies have also provided evidence of mtDNA colocalization with cGAS and triggering inflammatory damage under specific conditions [40, 101]. Sliter et al. presented compelling findings connecting mtDNA-STING signaling to neurodegenerative diseases, showing that mitochondrial stress induced by mtDNA mutations triggers a STING-IFN-I response in a PD mouse model lacking Parkin or PINK1 [102]. Furthermore, the release of mtDNA is related to the mitochondrial localization of neuropathological proteins, which may impair the integrity and function of the mitochondrial membrane. A critical study has reported that the mitochondrial localization of tau protein triggers mtDNA leakage and cGAS activation in microglia treated with tau fibrils, diminishing cognitive resilience through decreasing the neuronal transcriptional network of myocyte enhancer factor 2c (MEF2C) [103]. A similar mechanism was revealed in a parallel study, where the mitochondrial translocation of transactive response DNA binding protein of 43 kDa (TDP-43) drives mtDNA release into the cytoplasm, activating the cGAS-STING pathway in ALS models [19]. These studies together demonstrate the interaction of mtDNA with the cGAS-STING and its downstream pathways, providing persuasive evidence that mtDNA escaping into the extramitochondrial space can be sensed by cGAS.

Further in-depth research has established the link between mitochondrial stress in neurons and neuroinflammation. After ischemia–reperfusion injury, ox-mtDNA activated the cGAS-STING pathway within neurons, and restricting the release of ox-mtDNA into the cytoplasm downregulated p-STING expression levels in both neurons and microglia [104]. Other research has reported that neurons could release vesicles containing mtDNA into the extracellular space, which can be engulfed by neighboring microglial cells [105,106,107,108]. Additionally, transfection of extracted mtDNA into microglia upregulated the expression of IFN-β which was significantly inhibited upon cGAS knockout [109], thus cGAS in microglia may also be activated by mtDNA released from other cells. Based on these results, we cautiously propose that stressed neurons can release mtDNA and other pro-inflammatory factors into the extracellular space under various harmful conditions, and this displaced mtDNA may then be recognized by nearby microglia or astrocytes, consequently activating cGAS-STING signaling in these immune cells and exacerbating neuroinflammation. Unfortunately, direct evidence for this process is still lacking in AD. If further validated, targeting this pathway may hold even greater promise.

The role of the mtDNA-cGAS-STING pathway in AD

AD is a neurodegenerative disease with an insidious onset, caused by the interaction of genetic and environmental factors [110]. Despite extensive research, its pathogenesis remains elusive. Emerging evidence suggests that mitochondrial disorders and innate immune responses play crucial roles in the development of AD [7, 111]. Specifically, numerous adverse factors, including elevated levels of pathogenic proteins in AD, could destabilize the neuronal mitochondrial genome [112, 113], leading to mitochondrial dysfunction, ultimately triggering the release of disrupted mtDNA into the extramitochondrial space to initiate a series of immune responses [114]. These maladaptive neuroinflammatory activities in the CNS are generally considered to exacerbate pathological changes and accelerate the rate of AD-related cognitive decline [115, 116]. Furthermore, several AD-related risk genes have been implicated in inflammation. As described by He et al., among the 50 AD-related risk loci identified through genome-wide association analysis (GWAS), more than half were significantly enriched or specifically expressed in immune cells, particularly microglia and macrophages, such as Age and apolipoprotein E (APOE), triggering receptors expressed on myeloid cells 2 (TREM2), and ATP-binding cassette subfamily A (ABCA) [117]. More importantly, the cGAS-STING signaling, a pivotal DNA-sensing pathway, links cytoplasmic mtDNA to sterile inflammation and the body’s innate immune system, and its activation has been identified across multiple cell types in AD brains [118]. Overall, mtDNA-cGAS-STING activation in AD highlights the intricate interplay between mitochondrial dysfunction and neuroinflammation (Fig. 3). Elucidating the molecular mechanisms of this immune-inflammatory signaling pathway holds immense promise for developing novel therapeutic interventions to mitigate the devastating impact of AD on both individuals and society as a whole.

Fig. 3
figure 3

mtDNA-cGAS-STING signal pathway and neuroinflammation in AD. A. In the AD brain, the neurotoxic effects of primary pathological proteins, such as Aβ plaques and neurofibrillary tangles. The overwhelmed mitochondria of injured neurons release mtDNA into the extramitochondrial space, activating immune cells and forming a neuroinflammatory microenvironment in the CNS. B. The activation of the mtDNA-cGAS-STING pathway in the brain of AD. Firstly, various adverse factors in AD, such as Aβ plaques, hyperphosphorylated tau, ROS, and aging, stimulate the release of mtDNA from mitochondria in neurons. This mtDNA, along with AD pathological proteins, initiates the cGAS-STING pathway through multiple mechanisms. Secondly, cGAMP spreads through gap junctions in neurons and various glial cells, leading to the release of a series of cytokines by activated glial cells, and modulating neuronal inflammation. Ultimately, the activation of cGAS-STING would lead to complex outcomes. Various cell types in the brain play a role in the activation of this pathway, which, on one hand, can alleviate brain inflammation to some extent, while, on the other hand, its excessive activation forms a widespread neuroinflammatory network, exacerbating brain damage and promoting disease progression

mtDNA disturbances in AD

Endogenous neuroprotection and repair within the body rely on the health of the mtDNA, whose turbulence inevitably leads to various AD pathological changes, including the abnormal aggregation of pathological amyloid-β (Aβ) plaques [119, 120] and hyperphosphorylated tau protein [121], impaired synaptic transmission and plasticity [122], cholinergic dysfunction [123], and neuroinflammation [124]. For example, mtDNA mutations in mice lead to an increase in Aβ42 levels and Aβ42 plaque density, and the accumulation of amyloidosis is caused by a decrease in the content of Aβ clearance enzymes, rather than an increase in Aβ production [125]. This decrease in Aβ clearance is likely due to energy supply disruption connected to mtDNA damage. Consistently, Scheffler et al. confirmed that mtDNA polymorphisms, such as mutations affecting oxidative phosphorylation or the tricarboxylic acid cycle, can cause reduced ATP levels and ATP-driven microglial activity, resulting in increased Aβ aggregation [126]. These studies suggested that mtDNA disruption exacerbates AD pathology by impairing mitochondrial function. In turn, mtDNA is susceptible to attacks from these destructive changes, further exacerbating mitochondrial injury [127, 128]. Specifically, Aβ can be transported to mitochondria through the translocase of the outer membrane (TOM) and localize to the mitochondrial cristae [129, 130], impairing oxidative phosphorylation pathways, causing mtDNA oxidative damage and DNA double-strand breaks (DSBs) [131]. This vicious cycle between mitochondrial defects and AD pathological impairment [132, 133] accelerates the rate of cognitive decline.

Several AD-related studies have provided abundant evidence to indicate the importance of mtDNA disturbances, primarily oxidative damage, mutations, and methylation transfer [134]. In the brain tissue samples from APPswe/PS1dE9 (APP/PS1) transgenic mouse model and AD patients, expression levels of genes necessary to mitochondrial replication and energy metabolism, including the peroxisome proliferator-activated receptor gamma coactivator 1 alpha (PGC-1α), mitochondrial transcription factor A (TFAM), and neurogenic differentiation factor-6 (NEUROD6), are significantly downregulated [135, 136], indicating a pronounced mtDNA maintenance defect in AD. Similarly, the oxidized base level of mtDNA in the brains of postmortem AD individuals has been shown to be markedly higher than that in an age-matched control group, and is approximately tenfold that of nuclear DNA (nDNA) [137]. Additionally, Chang et al. found a greater frequency of mtDNA point mutations in the hippocampal region of patients with AD, whereas the mtDNA (4977) and deletion mutation did not increase [138]. However, Hamblet et al. indicated that mtDNA delta 4977 in the temporal lobe cortex of postmortem AD brains was elevated 6.5 folds [139]. Notably, lower levels of mtDNA mutations have also been observed in patients with AD [124], which may be related to neuronal death and loss during AD progression. In addition, the degree of mtDNA methylation in AD remains highly uncertain [140, 141], possibly due to methodological differences among studies. In conclusion, current research on mtDNA abnormalities in AD has not reached a consensus, and mtDNA disorders exhibit significant heterogeneity. In the future, it will be necessary to interpret the role of mtDNA in AD pathogenesis using improved detection techniques, increased sample sizes, and other feasible approaches.

Alterations in mtDNA content in AD remain controversial [142,143,144] (Table 1). Increased levels of cytoplasmic mtDNA have been observed in the brains of 5xFAD mice compared with age-matched wild-type mice [145]. In line with this, Laura et al. reported elevated mtDNA counts in the CSF of patients with AD compared to those in normal individuals, albeit with considerable inter-individual heterogeneity [146]. Nevertheless, some studies have documented a reduction in mtDNAcn in the brain of AD patients [143, 144], as well as decreased levels of free mtDNA in the CSF of patients with preclinical AD [147]. A possible explanation is reduced energy metabolism or abnormal mitochondrial function in AD, which is reflected by mtDNAcn to some extent. While most studies have indicated a close relationship between abnormal mtDNA levels and AD pathology, determining whether changes in mtDNA content are causally related to the onset of AD and the specific mechanisms by which mtDNA levels affect AD pathogenesis remain significant challenges.

Table 1 Changes in mtDNA levels in clinical studies of AD

Direct evidence of the activation of cGAS-STING in AD

Although studies on the relationship between the cGAS-STING pathway and AD are currently limited, evidence of the activation of this pathway has been found in patients with AD and multiple AD models (Table 2). Increased interactions between dsDNA, RNA–DNA hybrids, and cGAS have been observed in AD patient-derived iPSCs [166], 5xFAD mice [145], or human AD fibroblasts [167]. Ferecskó et al. discovered that the expression levels of STING in neurons and endothelial cells were significantly elevated in CNS tissues extracted from AD patients [168]. Consistently, the phosphorylation levels of STING, TBK1, p-65, and IRF3 were found to be upregulated in the prefrontal cortex of AD patients [145]. Further studies demonstrated that cGAS gene deletion in 5xFAD mice alleviated cognitive impairment, Aβ aggregation, neuroinflammation, and cholinergic neuron damage; STING inhibitors also sustainably ameliorated AD pathogenesis, which may be partially attributable to the inhibition of neurotoxic A1 astrocytes [117, 145, 167].

Table 2 Research evidence on the correlation between the cGAS-STING pathway and AD

Interestingly, two studies based on the APP/PS1 mouse model revealed reduced activity of the cGAS-STING pathway, whereas most research reports its activation (Table 2). Wang et al. found that the expression levels of cGAS and STING were significantly downregulated in the cardiomyocytes of APP/PS1 mice [175], despite the large aggregation of cytoplasmic mtDNA. Another study also observed a downward trend in STING expression levels in the APP/PS1 model compared to wild-type mice, although the differences were insignificant [179]. These contradictory results may be potentially due to differences in genetic backgrounds, stages of disease progression, and specific experimental conditions. Findings on this pathway derived from studies based on the APP/PS1 model should be carefully evaluated. In addition, STING in the Golgi apparatus was found to be nearly depleted in AD patients in a study by Nelson et al. [172]. A possible reason for this may be the prolonged activation of the cGAS-STING in the brain tissue, which has exhausted related components, and such prolonged activation may be mediated by the accumulated mtDNA and other dsDNA. However, it remains unclear whether the prolonged activation of this pathway truly exists in AD. Additionally, the threshold dose of dsDNA that triggers neuroinflammation and cellular damage, as well as the activation effects of different types of dsDNA, has yet to be fully defined. Overall, these changes reflect the plasticity of the pathway under different conditions to some extent and the potential protective effects of modulating this pathway in AD. Based on the existing evidence, understanding the role of the cGAS-STING in AD requires considering the composite effects of these factors, and how to balance the pathway to develop more effective therapeutic strategies.

Activation of cGAS-STING is associated with mtDNA in AD

Xie et al. previously linked the activation of the cGAS-STING pathway to mtDNA in AD. In their research, mtDNA and 2’3’-cGAMP levels in primary microglia, neurons, and astrocytes were found to be significantly increased following treatment with oligomeric human Aβ42 peptides [145]. Similarly, Wilkins et al. demonstrated that mitochondrial lysates or mtDNA injected into the mouse hippocampus could act as a DAMP to induce inflammatory responses and affect AD-related biomarkers [182]. Furthermore, Hou et al. found that nicotinamide riboside (NR) supplementation can rescue abnormal mitochondrial autophagy and reduce cytoplasmic DNA levels in the APP/PS1 mouse model, thereby reducing the cGAS perception of mtDNA [167]. In addition, Zhao et al. suggested that the mtDNA released by neurons activates the cGAS-STING pathway in adjacent microglia, astrocytes, and other neuronal cells [183], but further research is needed to confirm this opinion in AD. Overall, these studies provide preliminary evidence that extracellularly released mtDNA likely facilitates communication between various types of neuronal cells, promotes the propagation of neuroinflammation, and exacerbates the inflammatory microenvironment.

The relationship between cGAS-STING and the core pathological mechanism of AD

Hyperactivation of cGAS-STING and the aggregation of Aβ proteins are adverse factors in AD, and their interactions accelerate the pathological progression of AD [176]. Acker et al. found that the activation of cGAS-STING upregulated autophagy and induced amyloid precursor protein-C-terminal fragment (APP-CTF) accumulation [173]. Furthermore, p-STING primarily colocalized with the activated microglia marker CD68 around Aβ plaques in 5 × FAD mice [145], which may be related to ER and mitochondrial stress induced by the accumulation of neurotoxic proteins. In contrast, inhibition of the cGAS-STING and its downstream signaling could significantly alleviate Aβ toxicity [89, 117, 173]. These studies emphasize the importance of the cGAS-STING pathway in the neurotoxic process induced by Aβ aggregation and enrich the pathogenic mechanisms of Aβ as well.

The interaction between cGAS-STING and aberrantly aggregated hyperphosphorylated tau protein also contributes to AD development. Sequencing results from hippocampal tissue in a tau-induced disease model revealed elevated levels of cGAS and STING, while genetic ablation of cGAS in mice with tauopathy alleviated the activation of IFN-I response in microglia, without altering the pathological tau load in the brain. Additionally, pharmacological inhibition using TDI-6570, a non-toxic, brain-penetrant cGAS inhibitor, enhanced the neuronal MEF2C transcriptional network in the tauopathy mouse model, restoring synaptic integrity, plasticity, and memory [103]. Another recent study demonstrated that pharmacological inhibition of STING significantly reduces various pathological tau phosphorylation events in the AppNL-G-F/hTau double-knockin mouse model, further supporting the role of the cGAS-STING pathway in tau pathologies [180]. These studies revealed the critical role of the cGAS-STING pathway in AD, especially in its interaction with Aβ and tau. The modulation of this pathway may reduce the deposition of these pathological proteins, inhibit neuroinflammation, and protect neuronal and synaptic functions.

The relationship between the cGAS-STING pathway and APOE genotype

APOE ε4 is the strongest genetic risk factor for late-onset sporadic AD, exacerbating neurodegeneration induced by tau [184]. This process is partially attributed to a crucial immune hub, which involves the activation of the interferon pathway in microglia and interactions with cytotoxic T cells [185, 186]. Recent studies indicate that cGAS-STING plays a significant role in this context as well, further linking innate immunity to neuroinflammatory responses in AD. Chung et al. found that APOE ε4 allele, like Aβ and tau, upregulated cGAS and STING expression in microglia. The knockout or pharmacological inhibition of STING reversed the reactivity of APOE ε4 human iPSC-derived microglia, significantly reducing the Aβ burden and tau hyperphosphorylation while preserving memory functions in AppNL-G-F/hTau-dKI mice [180]. In addition, the R47H variant of the TREM2 and female sex are risk factors for sporadic AD. A study by Carling et al. found that APOE ε4 and R47H amplify the tau-induced microglial cGAS-STING-IFN-I responses in female mice, worsening neurodegeneration through upregulated cGAS and BAX-dependent microglial senescence, suggesting that enhanced IFN-I signaling under APOE ε4 and R47H backgrounds could be an important pathological mechanism in AD [181]. Specific APOE mutations, such as the R136S mutation on APOE3, could reduce tau-related pathological burden by inhibiting the cGAS-STING-IFN signaling pathway, and treatment with cGAS inhibitor prevented tau-induced synaptic loss in E3/P301S mice [185, 187]. These findings emphasize not only the importance of the APOE-activated cGAS-STING pathway in tau pathology but also the role of microglia in this inflammatory process. Modulating the cGAS-STING pathway may provide new strategies for developing therapies against AD, especially in carriers of the APOE ε4 allele.

Neuroinflammation induced by cGAS-STING and AD

Neuroinflammation is another significant feature of AD pathogenesis, involving aberrant activation of the innate immune system [188]. This effect is to some extent dependent on the disruption of the cGAS-STING pathway in different cells within the brain, especially in immune cells [189]. Although several types of neural cells can express cGAS and STING, microglia have been found to be the primary contributors to this pathway [117, 190, 191], which partially explains why they have been identified as major participants in neuroinflammation. Overall, abnormal activation of the cGAS-STING axis in diverse neuroglial cells is significant in the neuroinflammatory and neurodegenerative processes, particularly in microglia. Notably, moderate activation of this pathway may exert neuroprotective effects by suppressing inflammation. Evidence suggests that cGAMP treatment could activate the STING-IRF3 pathway to upregulate TREM2 expression, alleviating cognitive deficits and pathological changes in APP/PS1 transgenetic mice [179]. Another research has reported that ganciclovir can exert anti-inflammatory effects by mediating low therapeutic levels of IFNs, and this effect is STING-dependent [192]. Thus, the effects of cGAS-STING pathway activation vary, potentially depending on the differences in cell types and the extent of its activation. However, how to appropriately activate this pathway to achieve similar protective effects remains unclear. Further research is required to explore the balance between the anti-inflammatory and pro-inflammatory responses of this pathway in AD to achieve optimal neuroprotection.

The cGAS-STING pathway primarily induces neuroinflammation by regulating the expression of ISGs in inflammatory diseases [193, 194], including AD. Although the basal level of ISG mRNA in the CNS is generally much lower than in peripheral tissues, the brain is highly sensitive to IFN effects [195, 196]. In AD mouse models, pathological Aβ protein leads to a significant increase in ISG mRNA expression levels in the brain, and selective IFN receptor blockade effectively reduces persistent microgliosis and synaptic loss. Recent studies have also found that innate immune stimulation in neurons can drive seeded tau aggregation through the IFN-I response [197]. In clinical AD patients, the IFN pathway is also markedly upregulated and significantly correlated with disease severity and complement activation [198]. Additionally, the IFN-I produced by cGAS-STING pathway can induce a series of inflammatory responses by binding to its respective receptors in microglia, astrocytes, and neurons [199]. Then, these activated microglia secrete cytokines and chemokines, such as IL-1 and TNF-α, causing neurotoxicity through communicating with neurons [200]. Concisely, the cGAS-STING-IFN-I axis serves as an important mediator of neuroinflammation, forming a sophisticated regulatory network that bridges neuroglia and neurons in the CNS, leading to neuronal loss, and ultimately accelerating the onset of AD.

Interaction between Cellular Senescence and cGAS-STING in AD

The interplay between cGAS-STING and cellular senescence is considered another crucial pathological process that accelerates AD progression [201, 202]. A recent study has shown that aged microglia, compared to young microglia, exhibit an increased mtDNA burden in the cytosol. This triggers the activation of the cGAS-STING pathway and subsequent aging-related reactive microglial transcriptional states, suggesting that the cGAS-STING pathway is a driving force behind age-related inflammation [203]. Consistently, the accumulation of senescent cells, along with cGAS activation and STING homo-dimer formation, has been observed in various types of neural cells during aging [204,205,206,207]. In the APP/PS1 mouse model, cGAS and STING expression levels were higher in aged mice compared to younger mice [167]. After cGAS knockout in mouse embryonic fibroblast cells, the rate of cellular senescence significantly slowed and progressed toward immortalization [85]. Another research has shown that blocking cGAS or STING suppressed etoposide-induced cellular senescence [167]. As such, dsDNA released during aging signals through the cGAS-STING pathway, and targeting the cGAS-STING pathway holds considerable promise for slowing AD development by inhibiting the aging process.

One of the important mechanisms through which the interplay of the cGAS-STING pathway and cellular senescence contribute to AD is the activation of the senescence-associated secretory phenotype (SASP) [202], which includes a variety of cytokines, chemokines, and growth factors secreted by senescent cells [208, 209]. These SASPs can recruit immune cells and regulate their activity [207], altering intercellular communication levels, and thus accelerating the aging of senescent cells and their neighboring cells [210, 211]. During aging, the process by which the cGAS-STING induces SASP has been considered to involve LINE-1 (L1), a key component of sterile inflammation and a hallmark of aging. According to research by De Cecco et al., the transcriptional derepression of L1 activates IFN-I responses, sustaining SASP and exacerbating inflammatory senescent phenotypes during cellular senescence, and knocking out cGAS or STING suppresses SASP gene expression in late senescent cells [212]. Other studies have also shown that cGAS or STING inhibition downregulates SASP gene expression in both mouse and human cells [85, 213].

cGAS-STING-mediated senescence is also involved in the release of mtDNA. Specifically, mitochondria exhibit structural abnormalities and accumulate mtDNA in the cytoplasm in aged microglia, which subsequently triggers a cGAS-dependent inflammatory response [203]. Notably, Victorelli et al. discovered an intriguing phenomenon during cellular aging in which a subset of mitochondria undergoes a process called miMOMP, leading to mtDNA translocation into the cytosol to trigger cGAS-STING activation [33]. These findings suggested that cGAS-STING is a crucial molecular link between various types of DNA damage, mitochondrial dysfunction, and aging. More research is needed to fully understand the intricate relationships between them and the precise molecular mechanisms underlying these connections, thus providing valuable evidence for the development of more effective targeted drugs for AD.

The involvement of cGAS-STING in AD depends on microglia

Microglia, traditionally regarded as the resident macrophages of the brain, are involved in various physiological functions, ranging from immune surveillance to synaptic pruning [214]. In AD, microglia play a dual role, that is closely related to their involvement in the metabolic processes of neurotoxic proteins such as Aβ. On one hand, they can release degrading enzymes to degrade Aβ and further clear them through phagocytosis [215]. On the other hand, they further serve as carriers to seed Aβ, promoting its pathological spread throughout the CNS [216]. In turn, Aβ accumulation within the brain leads to the phenotypic shift of microglia, releasing neuroinflammatory substances and further contributing to neurotoxicity [176, 217]. Currently, the aberrant activation of cGAS-STING in microglia is believed to ultimately lead to neuronal dysfunction [117]. Modulating the activity of this pathway in microglia may aid in balancing the neuroprotective and neurotoxic effects, thereby slowing the progression of AD.

In recent studies, microglia have been regarded as the primary contributors to the cGAS-STING activation in AD, which is largely attributed to the relatively high expression levels of these two proteins [103, 170, 218], despite controversies regarding their expression levels across diverse cell types. Although Aβ oligomers can trigger cGAS activation in many types of neural cells, a more specific STING-IFN response is triggered in microglia, rather than in neurons and astrocytes [145]. Additionally, moderate expression levels of cGAS in neurons and oligodendrocytes were observed in another study, with low levels in astrocytes and endothelial cells [117]. Overall, cGAS-STING is largely not limited to a single cell type but encompasses a spectrum of cellular responses within the brain during AD. In addition to focusing on the complex role of this pathway in microglia, further exploration is required to understand the specific mechanisms in different cell types and how these cells communicate with each other, thus collectively slowing the development of AD.

Microglial neuroprotection or neurotoxicity depends on their phenotype and function to some extent (Fig. 4) [188], which can be regulated by the cGAS-STING pathway. AD-related research has focused mainly on traditional M1 and M2 microglial phenotypes currently [169, 176, 179], although evidence challenging this dichotomy exists. Single-nucleus RNA sequencing has identified other specific activation states in microglia related to cGAS, including interferon-responsive microglia (IRM), disease-associated microglia (DAM), and neurodegenerative microglia (MGnD). IRMs, a subset of microglia enriched with IFN response genes such as Ifits, Stat1, Sp100, Trim30a, and Parp14 [181, 203], play a crucial role in AD-related neuroinflammation. According to a recent study, IRMs are significantly enriched in APOE4-R47H tauopathy mice [181]. Another research found that the proportion of IRMs increases with age, a process further exacerbated by Aβ deposition [219]. Importantly, the phenotypic transition to IRMs is closely linked to DAMPs and the activation of the cGAS-STING pathway [220]. In the mg-CgasR241E microglial population, IFN signatures were broadly upregulated, indicating that cGAS activation induces microglia to transition into a distinct IFN-activated state, which in turn promotes neuroinflammation and aging processes [203]. The increase in DAM is another prominent feature in AD [221], and cGAS activation has been found to facilitate the transition of DAM subtypes from a low-activation state (DAM-1) to a high-activation state (DAM-2) [203]. In 5xFAD mice, the absence of cGAS significantly eliminates Aβ pathology-induced DAM markers [117]. However, these findings mainly rely on bulk transcriptomics and single-nucleus RNA sequencing, which may overlook the spatial differences in microglial activation states across different brain regions. The integration of spatial transcriptomics can better reveal the heterogeneity of cGAS-STING-driven microglial phenotypes across different brain regions in AD.

Fig. 4
figure 4

Microglia and the cGAS-STING pathway in AD. A The dual role of microglia in the CNS. Under physiological conditions, ramified microglia survey the brain microenvironment through their processes, sensing various damage signals. These highly dynamic cells perform immune surveillance, provide neurotrophic support, promote the establishment and maturation of neural circuits, and clear pathogens or debris, maintaining brain homeostasis. When the brain is challenged by pathogens or subjected to injury, activated microglia release various factors, including pro-inflammatory cytokines and chemokines. These factors help clear pathogens or toxins but may also lead to neuronal dysfunction and damage. B Activation of the cGAS-STING pathway in microglia in AD: Microglia are the primary cell type that activate cGAS-STING signaling in the brain. Pathological aggregates, such as Aβ plaques and tau proteins, cause mitochondrial damage and mtDNA release, leading to the activation of the cGAS-STING pathway in microglia, as well as astrocytes. Additionally, microglia secrete cytokines that induce the infiltration of peripheral immune cells, collectively modulating inflammation to alleviate neurodegeneration. However, overactivation of this system results in neuroinflammation, which can lead to neuronal dysfunction

Various receptors expressed in microglia are linked to the activation of this pathway. Polyglutamine-binding protein 1 (PQBP1), an intracellular receptor capable of binding to HIV cDNA [222], has emerged as a participant in tau-mediated cGAS activation. Upon recognition of tau3R/4R, PQBP1 activates cGAS-STING, leading to the induction of NF-κB nuclear translocation. In contrast, depletion of PQBP1 markedly attenuates the recruitment of cGAS and NF-κB-dependent transcription of inflammation genes, thus mitigating inflammation in the brain and cognitive impairment [223]. As such, PQBP1 is a promising novel therapeutic target for AD and other tau proteinopathies. Another relevant receptor is TREM2, a transmembrane immune receptor predominantly expressed in microglia [224]. Xu et al. demonstrated that stimulation of the cGAMP-STING-IRF3 pathway can induce TREM2 expression, which promotes the phagocytosis of Aβ by microglia, and reduces Aβ deposition and neuronal loss, simultaneously attenuating the pathological morphology and cognitive deficits in AD. Therefore, upregulating TREM2 may exert a regulatory effect on the cGAS-STING pathway, potentially mitigating its neurotoxic effects to some extent.

The propagation of inflammatory signals triggered by STING-IFN in the microglia relies on intercellular gap junctions. Gap junction channels serve as vital conduits for intercellular communication, enabling the unrestricted flow of various small molecules and ions between neighboring cells [225]. Ablasser et al. demonstrated that the 2’3’-cGAMP produced by cGAS can exploit gap junctions to establish inflammatory crosstalk between human embryonic kidney cells and murine embryonic fibroblasts [226], and can also be transported between adjacent cells through specific anion channels [227]. In a mouse model induced by high-fat diets, the activation of cGAS-STING and microglia both occurred in the CNS; however, in their neuron-microglia co-culture system, a significant inhibition in cGAS-STING activation and inflammatory crosstalk was observed upon blocking gap junctions [228]. Accordingly, 2’3’-cGAMP produced in neurons can similarly converge into adjacent microglia, triggering the activation of STING-IFN and subsequently producing diverse inflammatory factors [229]. These inflammatory mediators not only stimulate microglia but can also influence other types of cells, such as astrocytes and neurons, thereby establishing a vast inflammatory microenvironment within the brain [230, 231]. The association between gap junctions and AD pathology described by Mei et al. is also noteworthy; in this study, they observed the enrichment of gap junction-forming proteins CX43 and CX30 near Aβ plaques, and blocking these gap junctions slowed pathological progression [232]. To conclude, gap junctions and other channels are likely involved in the exchange of cGAS-STING-mediated inflammatory signals between neurons and glial cells. Exploring the signal transduction mechanisms between neighboring neural cells may help to identify approaches to regulate this process of inflammatory communication, ultimately slowing down or even inhibiting the progression of AD.

Potential therapeutic strategies and future directions

After mtDNA release, excessive activation of cGAS-STING and subsequent neuroinflammation inevitably lead to neuronal death and loss. Although the role of the mtDNA-cGAS-STING pathway in the CNS has only been partially revealed in recent years, its potential as an emerging target for AD treatment has gradually become apparent, and its significance and prospective utility cannot be ignored. Therefore, besides alleviating and inhibiting the abnormal release of mtDNA or other dsDNA caused by mitochondrial stress and autophagy imbalance [233], blocking the transmission of cGAS-STING signaling and the activation of its downstream inflammatory signals may represent a feasible approach for AD therapy [218].

Currently, a large body of preclinical research suggests that regulating the cGAS-STING is a promising intervention strategy (Table 3) (Fig. 5). Drugs targeting this pathway that have shown beneficial effects in AD animal or cellular models including H-151, RU.521, and C-176. For instance, H-151, inhibiting the palmitoylation of STING, and RU.521, targeting the catalytic site of cGAS, both significantly alleviated Aβ pathology in 5xFAD mice [145]; C-176 is an irreversible STING inhibitor that covalently binds to the Cys91 site of STING [234], significantly preventing neuroinflammation in microglia treated with Aβ25–35, and showing even more pronounced effects with combined treatment with RU.521 [176]. In addition, certain drugs, such as NR and its metabolite NAD+, the endogenous hormone melatonin, and the antibiotic dapsone [167, 171, 174, 175], all can non-specifically modulate this pathway to alleviate AD pathology. Although no drugs targeting this pathway have yet been approved for AD treatment, these studies provide valuable insights for the development of therapeutic drugs for AD in the future.

Table 3 The inhibitors targeting the cGAS-STING pathway
Fig. 5
figure 5

Overview of the inhibitors targeting the cGAS-STING signal pathway

Despite significant progress in the research on cGAS-STING and its related modulators, there are still some issues that require clarification. Firstly, the cGAS and STING proteins are widely expressed in various tissues and cells, which may lead to significant adverse reactions when cGAS and STING modulators are used. Thus, when developing drugs targeting this pathway, it would be necessary to chemically optimize them to enhance target selectivity and deliver them selectively to the CNS, minimizing adverse reactions caused by their effects on the immune system throughout the body as much as possible. Additionally, owing to the functional diversity of cGAS and STING, and the complexity of the immune surveillance network [266, 267], complete inhibition of cGAS-STING signaling may not be a feasible approach. Specifically, prolonged inhibition of this pathway may be detrimental to the treatment of diseases requiring acute and beneficial initial neuroinflammatory responses, including stroke, spinal cord injury, and traumatic brain injury [268], although it may have relatively beneficial effects on chronic neurodegenerative diseases. At the same time, STING inhibitors may lead to excessive suppression of the body’s immune system, thus impairing the immune surveillance of tumor cells to increase the risk of tumor occurrence [269], and weakening antiviral and antibacterial responses that could result in severe infections [189]. It is also essential to identify the balance point for the regulation of this pathway to ensure the maximum alleviation of AD pathology while avoiding other adverse reactions.

Secondly, in studies related to AD treatment, the expression of cGAS and STING has been reported in multiple types of neural cells [103, 117]. However, whether this pathway mediates diverse pathophysiological responses in different cell types still remains unclear. Additionally, there is as yet no consensus regarding the cell types in the brain expressing cGAS and STING and their expression levels in different cell types; the intracellular localization of cGAS has not been fully elucidated as well [270]. Thus, necessitating further research to explore their cellular distribution to provide compelling evidence for more precise targeted therapy.

Furthermore, a comprehensive consideration of other influencing factors to improve drug treatment efficacy is equally important. In addition to cGAS, other DNA sensors, such as IFN-γ inducible protein 16 (IFI16) and DEAD-box helicase 41 (DDX41), have been found to mediate downstream signaling through STING and appear to complement cGAS in pathogen detection under certain circumstances [17, 271]. Among them, DDX41 modulates cGAS activation by regulating the homeostasis of dsDNA and ssDNA [272]. However, the mechanisms of interaction among these DNA sensors remain largely unclear. Simultaneous interference with these pathways could potentially impair immune surveillance and increase susceptibility to certain diseases. Another issue that requires attention is that existing research on this pathway is mostly based on animal or cell models, and there may be species differences in cGAS and STING [268], which could lead to poor clinical outcomes. In addition, drugs must be transported from the periphery to the CNS through the blood–brain barrier (BBB), increasing BBB permeability and realizing precise targeting therapy also needs to be given priority. However, current studies on these drugs primarily focus on their anti-inflammatory effects and are limited to peripheral administration in animal or in cell models, with insufficient research on their ability to cross the BBB, specific distribution, and metabolic processes within the CNS. Moreover, optimizing drug delivery systems, such as nanoparticle-based carriers, carrier/receptor-mediated endocytosis, and exosome-mediated drug delivery [273,274,275,276], may offer breakthroughs in targeting the cGAS-STING pathway for AD treatment.

Conclusion

As research in the field has continued to advance, the mechanisms of mtDNA release and the specific role of the cGAS-STING pathway in AD are constantly being updated. Recent studies have revealed that mtDNA release and the activation of this pathway are interconnected events that collectively drive AD progression. Due to the extensive spectrum of pathogenic mechanisms formed by the interaction of this pathway with AD pathological processes, including neuroinflammation, aging, and neurodegeneration, further exploration of their causal relationships is essential. Importantly, the development of modulators targeting this pathway may provide new therapeutic strategies for AD. In summary, a thorough investigation into the role of the mtDNA-cGAS-STING pathway in AD is crucial for understanding its pathogenesis, identifying new therapeutic targets, and providing more possibilities for alleviating symptoms and slowing disease progression in patients with AD.

Data availability

Not applicable.

Abbreviations

AD:

Alzheimer’s disease

NFT:

Neurofibrillary tangles

DAMPs:

Damage-associated molecular patterns

PRRs:

Pattern recognition receptors

cGAS:

Cyclic GMP-AMP synthase

mtDNA:

Mitochondrial DNA

dsDNA:

Double-stranded DNA

STING:

Stimulator of interferon genes

CNS:

Central nervous system

PD:

Parkinson’s disease

ALS:

Amyotrophic lateral sclerosis

POCD:

Postoperative cognitive dysfunction

TBE:

Traumatic brain encephalopathy

miMOMP:

Minority MOMP

VDAC:

Voltage-dependent anion channel

OMM:

Outer mitochondrial membrane

MOMP:

Mitochondrial outer membrane permeabilization

EVs:

Extracellular vesicles

NETs:

Neutrophil extracellular traps

IFN:

Interferon

IMM:

Inner mitochondrial membrane

CICD:

Caspase-independent cell death

MIMP:

Mitochondrial inner membrane permeabilization

mt-dsRNA:

Mitochondrial double-stranded RNA

ANT:

Adenine nucleotide translocator

mPTP:

Mitochondrial permeability transition pore

VRK2:

Virus-related kinase 2

HK2:

Hexokinase 2

ER:

Endoplasmic reticulum

GSDMs:

Gasdermins

IL-1β:

Interleukin-1β

GSDMD:

Gasdermin D

CSF:

Cerebrospinal fluid

cf-mtDNA:

Cell-free mtDNA

mtDNAcn:

MtDNA copy numbers

TLR-9:

Toll-like receptor-9

NLRP3:

Nucleotide-binding oligomerization domain (NOD), leucine-rich repeat (LRR), and pyrin domain-containing protein 3

Mab21:

Male abnormal gene family 21

2 ’3’-cGAMP:

2’3’-Cyclic GMP-AMP

GTP:

Guanosine triphosphate

ATP:

Adenosine triphosphate

CCT:

C-terminal tail

TBK1:

TANK-binding kinase 1

IRF3:

Interferon regulatory factor 3

IFN-I:

Type I interferons

ISGs:

Interferon-stimulated genes

IKK:

IκB kinase

NF-κb:

Nuclear factor kappa-B

TNF-α:

Tumor necrosis factor-alpha

IL-6:

Interleukin-6

ERGIC:

ER-Golgi intermediate compartment

LC3:

Microtubule-associated protein 1 light chain 3

MEF2C:

Myocyte enhancer factor 2c

TDP-43:

Transactive response DNA binding protein of 43 kDa

GWAS:

Genome-wide association analysis

APOE:

Age and apolipoprotein E

TREM2:

Triggering receptors expressed on myeloid cells 2

ABCA:

ATP-binding cassette subfamily A

Aβ:

β-Amyloid

TOM:

Translocase of the outer membrane

DSBs:

DNA double-strand breaks

APP/PS1:

APPswe/PS1dE9

PGC-1α:

Peroxisome proliferator-activated receptor gamma coactivator 1 alpha

TFAM:

Mitochondrial transcription factor A

NEUROD6:

Neurogenic differentiation factor-6

APP-CTF:

Amyloid precursor protein-C-terminal fragment

SASP:

Senescence-associated secretory phenotype

L1:

LINE-1

IRM:

Interferon-responsive microglia

DAM:

Disease-associated microglia

MGnD:

Neurodegenerative microglia

PQBP1:

Polyglutamine-binding protein 1

IFI16:

Interferon inducible protein 16

DDX41:

DEAD-box helicase 41

BBB:

Blood-brain barrier

References

  1. Mangialasche F, Solomon A, Winblad B, Mecocci P, Kivipelto M. Alzheimer’s disease: clinical trials and drug development. Lancet Neurol. 2010;9:702–16.

    Article  PubMed  CAS  Google Scholar 

  2. DeTure MA, Dickson DW. The neuropathological diagnosis of Alzheimer’s disease. Mol Neurodegener. 2019;14:32.

    Article  PubMed  PubMed Central  Google Scholar 

  3. Jia L, Du Y, Chu L, Zhang Z, Li F, Lyu D, et al. Prevalence, risk factors, and management of dementia and mild cognitive impairment in adults aged 60 years or older in China: a cross-sectional study. Lancet Public Health. 2020;5:e661–71.

    Article  PubMed  Google Scholar 

  4. Scheltens P, De Strooper B, Kivipelto M, Holstege H, Chételat G, Teunissen CE, et al. Alzheimer’s disease. Lancet. 2021;397:1577–90.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  5. Alzheimer’s disease facts and figures. Alzheimers Dement. 2023;19:1598–695.

    Article  Google Scholar 

  6. Busche MA, Hyman BT. Synergy between amyloid-β and tau in Alzheimer’s disease. Nat Neurosci. 2020;23:1183–93.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  7. Self WK, Holtzman DM. Emerging diagnostics and therapeutics for Alzheimer disease. Nat Med. 2023;29:2187–99.

    Article  PubMed  CAS  Google Scholar 

  8. Munoz DG, Feldman H. Causes of Alzheimer’s disease. CMAJ. 2000;162:65–72.

    PubMed  PubMed Central  CAS  Google Scholar 

  9. Monteiro AR, Barbosa DJ, Remião F, Silva R. Alzheimer’s disease: Insights and new prospects in disease pathophysiology, biomarkers and disease-modifying drugs. Biochem Pharmacol. 2023;211: 115522.

    Article  PubMed  CAS  Google Scholar 

  10. van Olst L, Kamermans A, Halters S, van der Pol SMA, Rodriguez E, Verberk IMW, et al. Adaptive immune changes associate with clinical progression of Alzheimer’s disease. Mol Neurodegener. 2024;19:38.

    Article  PubMed  PubMed Central  Google Scholar 

  11. Chen X, Holtzman DM. Emerging roles of innate and adaptive immunity in Alzheimer’s disease. Immunity. 2022;55:2236–54.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  12. Gong T, Liu L, Jiang W, Zhou R. DAMP-sensing receptors in sterile inflammation and inflammatory diseases. Nat Rev Immunol. 2020;20:95–112.

    Article  PubMed  CAS  Google Scholar 

  13. Zindel J, Kubes P. DAMPs, PAMPs, and LAMPs in Immunity and Sterile Inflammation. Annu Rev Pathol. 2020;15:493–518.

    Article  PubMed  CAS  Google Scholar 

  14. Chow J, Franz KM, Kagan JC. PRRs are watching you: Localization of innate sensing and signaling regulators. Virology. 2015;479–480:104–9.

    Article  PubMed  Google Scholar 

  15. Ablasser A, Chen ZJ. cGAS in action: Expanding roles in immunity and inflammation. Science. 2019;363:eaat8657.

  16. Kim J, Kim H-S, Chung JH. Molecular mechanisms of mitochondrial DNA release and activation of the cGAS-STING pathway. Exp Mol Med. 2023;55:510–9.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  17. Cheng Z, Dai T, He X, Zhang Z, Xie F, Wang S, et al. The interactions between cGAS-STING pathway and pathogens. Signal Transduct Target Ther. 2020;5:91.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  18. Liu Y, Duan R, Li P, Zhang B, Liu Y. 3-N-butylphthalide attenuates neuroinflammation in rotenone-induced Parkinson’s disease models via the cGAS-STING pathway. Int J Immunopathol Pharmacol. 2024;38:3946320241229041.

    Article  PubMed  CAS  Google Scholar 

  19. Yu C-H, Davidson S, Harapas CR, Hilton JB, Mlodzianoski MJ, Laohamonthonkul P, et al. TDP-43 Triggers Mitochondrial DNA Release via mPTP to Activate cGAS/STING in ALS. Cell. 2020;183:636-649.e18.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  20. Yang N-S-Y, Zhong W-J, Sha H-X, Zhang C-Y, Jin L, Duan J-X, et al. mtDNA-cGAS-STING axis-dependent NLRP3 inflammasome activation contributes to postoperative cognitive dysfunction induced by sevoflurane in mice. Int J Biol Sci. 2024;20:1927–46.

  21. Fritsch LE, Ju J, Gudenschwager Basso EK, Soliman E, Paul S, Chen J, et al. Type I Interferon Response Is Mediated by NLRX1-cGAS-STING Signaling in Brain Injury. Front Mol Neurosci. 2022;15: 852243.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  22. Gustafsson CM, Falkenberg M, Larsson N-G. Maintenance and Expression of Mammalian Mitochondrial DNA. Annu Rev Biochem. 2016;85:133–60.

    Article  PubMed  CAS  Google Scholar 

  23. Yan C, Duanmu X, Zeng L, Liu B, Song Z. Mitochondrial DNA: Distribution, Mutations, and Elimination. Cells. 2019;8:379.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  24. Wang B, Huang M, Shang D, Yan X, Zhao B, Zhang X. Mitochondrial Behavior in Axon Degeneration and Regeneration. Front Aging Neurosci. 2021;13: 650038.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  25. De Gaetano A, Solodka K, Zanini G, Selleri V, Mattioli AV, Nasi M, et al. Molecular Mechanisms of mtDNA-Mediated Inflammation Cells. 2021;10:2898.

    PubMed  Google Scholar 

  26. Collins LV, Hajizadeh S, Holme E, Jonsson I-M, Tarkowski A. Endogenously oxidized mitochondrial DNA induces in vivo and in vitro inflammatory responses. J Leukoc Biol. 2004;75:995–1000.

    Article  PubMed  CAS  Google Scholar 

  27. Fang C, Wei X, Wei Y. Mitochondrial DNA in the regulation of innate immune responses. Protein Cell. 2016;7:11–6.

    Article  PubMed  CAS  Google Scholar 

  28. Picca A, Calvani R, Coelho-Junior HJ, Marzetti E. Cell Death and Inflammation: The Role of Mitochondria in Health and Disease. Cells. 2021;10:537.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  29. Ng MYW, Wai T, Simonsen A. Quality control of the mitochondrion. Dev Cell. 2021;56:881–905.

    Article  PubMed  CAS  Google Scholar 

  30. Chapman J, Ng YS, Nicholls TJ. The Maintenance of Mitochondrial DNA Integrity and Dynamics by Mitochondrial Membranes. Life (Basel). 2020;10:164.

    PubMed  CAS  Google Scholar 

  31. McArthur K, Whitehead LW, Heddleston JM, Li L, Padman BS, Oorschot V, et al. BAK/BAX macropores facilitate mitochondrial herniation and mtDNA efflux during apoptosis. Science. 2018;359:eaao6047.

  32. Riley JS, Quarato G, Cloix C, Lopez J, O’Prey J, Pearson M, et al. Mitochondrial inner membrane permeabilisation enables mtDNA release during apoptosis. EMBO J. 2018;37: e99238.

    Article  PubMed  PubMed Central  Google Scholar 

  33. Victorelli S, Salmonowicz H, Chapman J, Martini H, Vizioli MG, Riley JS, et al. Apoptotic stress causes mtDNA release during senescence and drives the SASP. Nature. 2023;622:627–36.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  34. Kim J, Gupta R, Blanco LP, Yang S, Shteinfer-Kuzmine A, Wang K, et al. VDAC oligomers form mitochondrial pores to release mtDNA fragments and promote lupus-like disease. Science. 2019;366:1531–6.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  35. Yan J, Liu W, Feng F, Chen L. VDAC oligomer pores: A mechanism in disease triggered by mtDNA release. Cell Biol Int. 2020;44:2178–81.

    Article  PubMed  CAS  Google Scholar 

  36. Pérez-Treviño P, Velásquez M, García N. Mechanisms of mitochondrial DNA escape and its relationship with different metabolic diseases. Biochim Biophys Acta Mol Basis Dis. 2020;1866: 165761.

    Article  PubMed  Google Scholar 

  37. Jiang X, Jiang H, Shen Z, Wang X. Activation of mitochondrial protease OMA1 by Bax and Bak promotes cytochrome c release during apoptosis. Proc Natl Acad Sci U S A. 2014;111:14782–7.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  38. Cosentino K, Hertlein V, Jenner A, Dellmann T, Gojkovic M, Peña-Blanco A, et al. The interplay between BAX and BAK tunes apoptotic pore growth to control mitochondrial-DNA-mediated inflammation. Mol Cell. 2022;82:933-949.e9.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  39. Gillies LA, Du H, Peters B, Knudson CM, Newmeyer DD, Kuwana T. Visual and functional demonstration of growing Bax-induced pores in mitochondrial outer membranes. Mol Biol Cell. 2015;26:339–49.

    Article  PubMed  PubMed Central  Google Scholar 

  40. White MJ, McArthur K, Metcalf D, Lane RM, Cambier JC, Herold MJ, et al. Apoptotic caspases suppress mtDNA-induced STING-mediated type I IFN production. Cell. 2014;159:1549–62.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  41. Rongvaux A, Jackson R, Harman CCD, Li T, West AP, de Zoete MR, et al. Apoptotic caspases prevent the induction of type I interferons by mitochondrial DNA. Cell. 2014;159:1563–77.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  42. Newman LE, Shadel GS. Mitochondrial DNA Release in Innate Immune Signaling. Annu Rev Biochem. 2023;92:299–332.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  43. Yoon J, Kim S, Lee M, Kim Y. Mitochondrial nucleic acids in innate immunity and beyond. Exp Mol Med. 2023;55:2508–18.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  44. Dhir A, Dhir S, Borowski LS, Jimenez L, Teitell M, Rötig A, et al. Mitochondrial double-stranded RNA triggers antiviral signalling in humans. Nature. 2018;560:238–42.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  45. Becker Y, Marcoux G, Allaeys I, Julien A-S, Loignon R-C, Benk-Fortin H, et al. Autoantibodies in Systemic Lupus Erythematosus Target Mitochondrial RNA. Front Immunol. 2019;10:1026.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  46. Baines CP. The molecular composition of the mitochondrial permeability transition pore. J Mol Cell Cardiol. 2009;46:850–7.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  47. Murphy E, Steenbergen C. Mechanisms underlying acute protection from cardiac ischemia-reperfusion injury. Physiol Rev. 2008;88:581–609.

    Article  PubMed  CAS  Google Scholar 

  48. García N, García JJ, Correa F, Chávez E. The permeability transition pore as a pathway for the release of mitochondrial DNA. Life Sci. 2005;76:2873–80.

    Article  PubMed  Google Scholar 

  49. García N, Chávez E. Mitochondrial DNA fragments released through the permeability transition pore correspond to specific gene size. Life Sci. 2007;81:1160–6.

    Article  PubMed  Google Scholar 

  50. Halestrap AP, McStay GP, Clarke SJ. The permeability transition pore complex: another view. Biochimie. 2002;84:153–66.

    Article  PubMed  CAS  Google Scholar 

  51. Nezu J, Oku A, Jones MH, Shimane M. Identification of two novel human putative serine/threonine kinases, VRK1 and VRK2, with structural similarity to vaccinia virus B1R kinase. Genomics. 1997;45:327–31.

    Article  PubMed  CAS  Google Scholar 

  52. Zhu H, Li Q, Zhao Y, Peng H, Guo L, Zhu J, et al. Vaccinia-related kinase 2 drives pancreatic cancer progression by protecting Plk1 from Chfr-mediated degradation. Oncogene. 2021;40:4663–74.

    Article  PubMed  CAS  Google Scholar 

  53. He W-R, Cao L-B, Yang Y-L, Hua D, Hu M-M, Shu H-B. VRK2 is involved in the innate antiviral response by promoting mitostress-induced mtDNA release. Cell Mol Immunol. 2021;18:1186–96.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  54. Baik SH, Ramanujan VK, Becker C, Fett S, Underhill DM, Wolf AJ. Hexokinase dissociation from mitochondria promotes oligomerization of VDAC that facilitates NLRP3 inflammasome assembly and activation. Sci Immunol. 2023;8:eade7652.

  55. Miao R, Jiang C, Chang WY, Zhang H, An J, Ho F, et al. Gasdermin D permeabilization of mitochondrial inner and outer membranes accelerates and enhances pyroptosis. Immunity. 2023;56:2523-2541.e8.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  56. Ding J, Wang K, Liu W, She Y, Sun Q, Shi J, et al. Pore-forming activity and structural autoinhibition of the gasdermin family. Nature. 2016;535:111–6.

    Article  PubMed  CAS  Google Scholar 

  57. Monteleone M, Stanley AC, Chen KW, Brown DL, Bezbradica JS, von Pein JB, et al. Interleukin-1β Maturation Triggers Its Relocation to the Plasma Membrane for Gasdermin-D-Dependent and -Independent Secretion. Cell Rep. 2018;24:1425–33.

    Article  PubMed  CAS  Google Scholar 

  58. Huang LS, Hong Z, Wu W, Xiong S, Zhong M, Gao X, et al. mtDNA Activates cGAS Signaling and Suppresses the YAP-Mediated Endothelial Cell Proliferation Program to Promote Inflammatory Injury. Immunity. 2020;52:475-486.e5.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  59. Heilig R, Lee J, Tait SWG. Mitochondrial DNA in cell death and inflammation. Biochem Soc Trans. 2023;51:457–72.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  60. Miao N, Wang Z, Wang Q, Xie H, Yang N, Wang Y, et al. Oxidized mitochondrial DNA induces gasdermin D oligomerization in systemic lupus erythematosus. Nat Commun. 2023;14:872.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  61. Zhao C, Liang F, Ye M, Wu S, Qin Y, Zhao L, et al. GSDMD promotes neutrophil extracellular traps via mtDNA-cGAS-STING pathway during lung ischemia/reperfusion. Cell Death Discov. 2023;9:368.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  62. Deng X, Yang G, Zheng X, Yang Y, Qin H, Liu Z-X, et al. Plasma mtDNA copy numbers are associated with GSTK1 expression and inflammation in type 2 diabetes. Diabet Med. 2020;37:1874–8.

    Article  PubMed  CAS  Google Scholar 

  63. Liang Y, Fan S, Jiang Y, Ji T, Chen R, Xu Q, et al. Elevated serum mitochondrial DNA levels were associated with the progression and mortality in idiopathic pulmonary fibrosis. Int Immunopharmacol. 2023;123: 110754.

    Article  PubMed  CAS  Google Scholar 

  64. Leurs CE, Podlesniy P, Trullas R, Balk L, Steenwijk MD, Malekzadeh A, et al. Cerebrospinal fluid mtDNA concentration is elevated in multiple sclerosis disease and responds to treatment. Mult Scler. 2018;24:472–80.

    Article  PubMed  CAS  Google Scholar 

  65. Hajizadeh S, DeGroot J, TeKoppele JM, Tarkowski A, Collins LV. Extracellular mitochondrial DNA and oxidatively damaged DNA in synovial fluid of patients with rheumatoid arthritis. Arthritis Res Ther. 2003;5:R234-240.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  66. Zhang Y, Liu X, Wiggins KL, Kurniansyah N, Guo X, Rodrigue AL, et al. Association of Mitochondrial DNA Copy Number With Brain MRI Markers and Cognitive Function: A Meta-analysis of Community-Based Cohorts. Neurology. 2023;100:e1930–43.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  67. Chen S, Xie X, Wang Y, Gao Y, Xie X, Yang J, et al. Association between leukocyte mitochondrial DNA content and risk of coronary heart disease: a case-control study. Atherosclerosis. 2014;237:220–6.

    Article  PubMed  CAS  Google Scholar 

  68. Stephens OR, Grant D, Frimel M, Wanner N, Yin M, Willard B, et al. Characterization and origins of cell-free mitochondria in healthy murine and human blood. Mitochondrion. 2020;54:102–12.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  69. Ding W, Chen J, Zhao L, Wu S, Chen X, Chen H. Mitochondrial DNA leakage triggers inflammation in age-related cardiovascular diseases. Front Cell Dev Biol. 2024;12:1287447.

    Article  PubMed  PubMed Central  Google Scholar 

  70. Long G, Gong R, Wang Q, Zhang D, Huang C. Role of released mitochondrial DNA in acute lung injury. Front Immunol. 2022;13: 973089.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  71. Civril F, Deimling T, de Oliveira Mann CC, Ablasser A, Moldt M, Witte G, et al. Structural mechanism of cytosolic DNA sensing by cGAS. Nature. 2013;498:332–7.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  72. Bai J, Liu F. Nuclear cGAS: sequestration and beyond. Protein Cell. 2022;13:90–101.

    Article  PubMed  Google Scholar 

  73. Sun L, Wu J, Du F, Chen X, Chen ZJ. Cyclic GMP-AMP synthase is a cytosolic DNA sensor that activates the type I interferon pathway. Science. 2013;339:786–91.

    Article  PubMed  CAS  Google Scholar 

  74. Herzner A-M, Hagmann CA, Goldeck M, Wolter S, Kübler K, Wittmann S, et al. Sequence-specific activation of the DNA sensor cGAS by Y-form DNA structures as found in primary HIV-1 cDNA. Nat Immunol. 2015;16:1025–33.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  75. Li X, Shu C, Yi G, Chaton CT, Shelton CL, Diao J, et al. Cyclic GMP-AMP synthase is activated by double-stranded DNA-induced oligomerization. Immunity. 2013;39:1019–31.

    Article  PubMed  CAS  Google Scholar 

  76. Crossley MP, Song C, Bocek MJ, Choi J-H, Kousouros JN, Sathirachinda A, et al. R-loop-derived cytoplasmic RNA-DNA hybrids activate an immune response. Nature. 2023;613:187–94.

    Article  PubMed  CAS  Google Scholar 

  77. Yu L, Liu P. Cytosolic DNA sensing by cGAS: regulation, function, and human diseases. Signal Transduct Target Ther. 2021;6:170.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  78. Chen Q, Sun L, Chen ZJ. Regulation and function of the cGAS-STING pathway of cytosolic DNA sensing. Nat Immunol. 2016;17:1142–9.

    Article  PubMed  CAS  Google Scholar 

  79. Gekara NO, Jiang H. The innate immune DNA sensor cGAS: A membrane, cytosolic, or nuclear protein? Sci Signal. 2019;12:eaax3521.

  80. Barnett KC, Coronas-Serna JM, Zhou W, Ernandes MJ, Cao A, Kranzusch PJ, et al. Phosphoinositide Interactions Position cGAS at the Plasma Membrane to Ensure Efficient Distinction between Self- and Viral DNA. Cell. 2019;176:1432-1446.e11.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  81. Volkman HE, Cambier S, Gray EE, Stetson DB. Tight nuclear tethering of cGAS is essential for preventing autoreactivity. Elife. 2019;8: e47491.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  82. Lu Y, Zhao M, Chen L, Wang Y, Liu T, Liu H. cGAS: action in the nucleus. Front Immunol. 2024;15:1380517.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  83. Li T, Huang T, Du M, Chen X, Du F, Ren J, et al. Phosphorylation and chromatin tethering prevent cGAS activation during mitosis. Science. 2021;371:eabc5386.

  84. Zhong L, Hu M-M, Bian L-J, Liu Y, Chen Q, Shu H-B. Phosphorylation of cGAS by CDK1 impairs self-DNA sensing in mitosis. Cell Discov. 2020;6:26.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  85. Yang H, Wang H, Ren J, Chen Q, Chen ZJ. cGAS is essential for cellular senescence. Proc Natl Acad Sci U S A. 2017;114:E4612–20.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  86. Gao P, Ascano M, Wu Y, Barchet W, Gaffney BL, Zillinger T, et al. Cyclic [G(2’,5’)pA(3’,5’)p] is the metazoan second messenger produced by DNA-activated cyclic GMP-AMP synthase. Cell. 2013;153:1094–107.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  87. Xia P, Wang S, Gao P, Gao G, Fan Z. DNA sensor cGAS-mediated immune recognition. Protein Cell. 2016;7:777–91.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  88. Ishikawa H, Barber GN. STING is an endoplasmic reticulum adaptor that facilitates innate immune signalling. Nature. 2008;455:674–8.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  89. Chen K, Lai C, Su Y, Bao WD, Yang LN, Xu P-P, et al. cGAS-STING-mediated IFN-I Response in Host Defense and Neuroinflammatory Diseases. Curr Neuropharmacol. 2022;20:362–71.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  90. Shang G, Zhang C, Chen ZJ, Bai X-C, Zhang X. Cryo-EM structures of STING reveal its mechanism of activation by cyclic GMP-AMP. Nature. 2019;567:389–93.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  91. Zhang C, Shang G, Gui X, Zhang X, Bai X-C, Chen ZJ. Structural basis of STING binding with and phosphorylation by TBK1. Nature. 2019;567:394–8.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  92. King KR, Aguirre AD, Ye Y-X, Sun Y, Roh JD, Ng RP, et al. IRF3 and type I interferons fuel a fatal response to myocardial infarction. Nat Med. 2017;23:1481–7.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  93. Abe T, Barber GN. Cytosolic-DNA-mediated, STING-dependent proinflammatory gene induction necessitates canonical NF-κB activation through TBK1. J Virol. 2014;88:5328–41.

    Article  PubMed  PubMed Central  Google Scholar 

  94. Woznica A, Kumar A, Sturge CR, Xing C, King N, Pfeiffer JK. STING mediates immune responses in the closest living relatives of animals. Elife. 2021;10: e70436.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  95. Gui X, Yang H, Li T, Tan X, Shi P, Li M, et al. Autophagy induction via STING trafficking is a primordial function of the cGAS pathway. Nature. 2019;567:262–6.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  96. Yang J, Tang X, Nandakumar KS, Cheng K. Autophagy induced by STING, an unnoticed and primordial function of cGAS. Cell Mol Immunol. 2019;16:683–4.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  97. Hu X, Zhang H, Zhang Q, Yao X, Ni W, Zhou K. Emerging role of STING signalling in CNS injury: inflammation, autophagy, necroptosis, ferroptosis and pyroptosis. J Neuroinflammation. 2022;19:242.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  98. Saitoh T, Fujita N, Hayashi T, Takahara K, Satoh T, Lee H, et al. Atg9a controls dsDNA-driven dynamic translocation of STING and the innate immune response. Proc Natl Acad Sci U S A. 2009;106:20842–6.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  99. Liu D, Wu H, Wang C, Li Y, Tian H, Siraj S, et al. STING directly activates autophagy to tune the innate immune response. Cell Death Differ. 2019;26:1735–49.

    Article  PubMed  CAS  Google Scholar 

  100. West AP, Khoury-Hanold W, Staron M, Tal MC, Pineda CM, Lang SM, et al. Mitochondrial DNA stress primes the antiviral innate immune response. Nature. 2015;520:553–7.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  101. Zhang X, Wu J, Liu Q, Li X, Li S, Chen J, et al. mtDNA-STING pathway promotes necroptosis-dependent enterocyte injury in intestinal ischemia reperfusion. Cell Death Dis. 2020;11:1050.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  102. Sliter DA, Martinez J, Hao L, Chen X, Sun N, Fischer TD, et al. Parkin and PINK1 mitigate STING-induced inflammation. Nature. 2018;561:258–62.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  103. Udeochu JC, Amin S, Huang Y, Fan L, Torres ERS, Carling GK, et al. Tau activation of microglial cGAS-IFN reduces MEF2C-mediated cognitive resilience. Nat Neurosci. 2023;26:737–50.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  104. Li Q, Yang L, Wang K, Chen Z, Liu H, Yang X, et al. Oxidized mitochondrial DNA activates the cGAS-STING pathway in the neuronal intrinsic immune system after brain ischemia-reperfusion injury. Neurotherapeutics. 2024;21: e00368.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  105. Torralba D, Baixauli F, Villarroya-Beltri C, Fernández-Delgado I, Latorre-Pellicer A, Acín-Pérez R, et al. Priming of dendritic cells by DNA-containing extracellular vesicles from activated T cells through antigen-driven contacts. Nat Commun. 2018;9:2658.

    Article  PubMed  PubMed Central  Google Scholar 

  106. Gambardella S, Limanaqi F, Ferese R, Biagioni F, Campopiano R, Centonze D, et al. ccf-mtDNA as a Potential Link Between the Brain and Immune System in Neuro-Immunological Disorders. Front Immunol. 2019;10:1064.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  107. Liu Y, Zhang B, Duan R, Liu Y. Mitochondrial DNA Leakage and cGas/STING Pathway in Microglia: Crosstalk Between Neuroinflammation and Neurodegeneration. Neuroscience. 2024;548:1–8.

    Article  PubMed  CAS  Google Scholar 

  108. Tresse E, Marturia-Navarro J, Sew WQG, Cisquella-Serra M, Jaberi E, Riera-Ponsati L, et al. Mitochondrial DNA damage triggers spread of Parkinson’s disease-like pathology. Mol Psychiatry. 2023;28:4902–14.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  109. Liao Y, Cheng J, Kong X, Li S, Li X, Zhang M, et al. HDAC3 inhibition ameliorates ischemia/reperfusion-induced brain injury by regulating the microglial cGAS-STING pathway. Theranostics. 2020;10:9644–62.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  110. Guo T, Zhang D, Zeng Y, Huang TY, Xu H, Zhao Y. Molecular and cellular mechanisms underlying the pathogenesis of Alzheimer’s disease. Mol Neurodegener. 2020;15:40.

    Article  PubMed  PubMed Central  Google Scholar 

  111. Ashleigh T, Swerdlow RH, Beal MF. The role of mitochondrial dysfunction in Alzheimer’s disease pathogenesis. Alzheimers Dement. 2023;19:333–42.

    Article  PubMed  CAS  Google Scholar 

  112. Reddy PH, Oliver DM. Amyloid Beta and Phosphorylated Tau-Induced Defective Autophagy and Mitophagy in Alzheimer’s Disease. Cells. 2019;8:488.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  113. Reddy PH, Yin X, Manczak M, Kumar S, Pradeepkiran JA, Vijayan M, et al. 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. 2018;27:2502–16.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  114. Wilkins HM, Carl SM, Weber SG, Ramanujan SA, Festoff BW, Linseman DA, et al. Mitochondrial lysates induce inflammation and Alzheimer’s disease-relevant changes in microglial and neuronal cells. J Alzheimers Dis. 2015;45:305–18.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  115. VanItallie TB. Alzheimer’s disease: Innate immunity gone awry? Metabolism. 2017;69S:S41–9.

    Article  PubMed  Google Scholar 

  116. Lecca D, Jung YJ, Scerba MT, Hwang I, Kim YK, Kim S, et al. Role of chronic neuroinflammation in neuroplasticity and cognitive function: A hypothesis. Alzheimers Dement. 2022;18:2327–40.

    Article  PubMed  CAS  Google Scholar 

  117. He S, Li X, Mittra N, Bhattacharjee A, Wang H, Zhao S, et al. Microglial cGAS deletion protects against amyloid-β induced Alzheimer’s disease pathogenesis. bioRxiv. 2023;2023.08.07.552300.

  118. Li F, Wang N, Zheng Y, Luo Y, Zhang Y. cGAS- Stimulator of Interferon Genes Signaling in Central Nervous System Disorders. Aging Dis. 2021;12:1658–74.

    Article  PubMed  PubMed Central  Google Scholar 

  119. Sorrentino V, Romani M, Mouchiroud L, Beck JS, Zhang H, D’Amico D, et al. Enhancing mitochondrial proteostasis reduces amyloid-β proteotoxicity. Nature. 2017;552:187–93.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  120. Huang Z, Yan Q, Wang Y, Zou Q, Li J, Liu Z, et al. Role of Mitochondrial Dysfunction in the Pathology of Amyloid-β. J Alzheimers Dis. 2020;78:505–14.

    Article  PubMed  CAS  Google Scholar 

  121. Weidling IW, Wilkins HM, Koppel SJ, Hutfles L, Wang X, Kalani A, et al. Mitochondrial DNA Manipulations Affect Tau Oligomerization. J Alzheimers Dis. 2020;77:149–63.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  122. Verma H, Gangwar P, Yadav A, Yadav B, Rao R, Kaur S, et al. Understanding the neuronal synapse and challenges associated with the mitochondrial dysfunction in mild cognitive impairment and Alzheimer’s disease. Mitochondrion. 2023;73:19–29.

    Article  PubMed  CAS  Google Scholar 

  123. Bury AG, Pyle A, Elson JL, Greaves L, Morris CM, Hudson G, et al. Mitochondrial DNA changes in pedunculopontine cholinergic neurons in Parkinson disease. Ann Neurol. 2017;82:1016–21.

    Article  PubMed  CAS  Google Scholar 

  124. Shang D, Huang M, Wang B, Yan X, Wu Z, Zhang X. mtDNA Maintenance and Alterations in the Pathogenesis of Neurodegenerative Diseases. Curr Neuropharmacol. 2023;21:578–98.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  125. Kukreja L, Kujoth GC, Prolla TA, Van Leuven F, Vassar R. Increased mtDNA mutations with aging promotes amyloid accumulation and brain atrophy in the APP/Ld transgenic mouse model of Alzheimer’s disease. Mol Neurodegener. 2014;9:16.

    Article  PubMed  PubMed Central  Google Scholar 

  126. Scheffler K, Krohn M, Dunkelmann T, Stenzel J, Miroux B, Ibrahim S, et al. Mitochondrial DNA polymorphisms specifically modify cerebral β-amyloid proteostasis. Acta Neuropathol. 2012;124:199–208.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  127. Mao P, Reddy PH. Aging and amyloid beta-induced oxidative DNA damage and mitochondrial dysfunction in Alzheimer’s disease: implications for early intervention and therapeutics. Biochim Biophys Acta. 2011;1812:1359–70.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  128. Gezen-Ak D, Yurttaş Z, Çamoǧlu T, Dursun E. Could Amyloid-β 1–42 or α-Synuclein Interact Directly with Mitochondrial DNA? A Hypothesis ACS Chem Neurosci. 2022;13:2803–12.

    Article  PubMed  CAS  Google Scholar 

  129. Hansson Petersen CA, Alikhani N, Behbahani H, Wiehager B, Pavlov PF, Alafuzoff I, et al. The amyloid beta-peptide is imported into mitochondria via the TOM import machinery and localized to mitochondrial cristae. Proc Natl Acad Sci U S A. 2008;105:13145–50.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  130. Devi L, Prabhu BM, Galati DF, Avadhani NG, Anandatheerthavarada HK. Accumulation of amyloid precursor protein in the mitochondrial import channels of human Alzheimer’s disease brain is associated with mitochondrial dysfunction. J Neurosci. 2006;26:9057–68.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  131. Suberbielle E, Sanchez PE, Kravitz AV, Wang X, Ho K, Eilertson K, et al. Physiologic brain activity causes DNA double-strand breaks in neurons, with exacerbation by amyloid-β. Nat Neurosci. 2013;16:613–21.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  132. Mancuso M, Calsolaro V, Orsucci D, Siciliano G, Murri L. Is there a primary role of the mitochondrial genome in Alzheimer’s disease? J Bioenerg Biomembr. 2009;41:411–6.

    Article  PubMed  CAS  Google Scholar 

  133. Oka S, Leon J, Sakumi K, Ide T, Kang D, LaFerla FM, et al. Human mitochondrial transcriptional factor A breaks the mitochondria-mediated vicious cycle in Alzheimer’s disease. Sci Rep. 2016;6:37889.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  134. Antonyová V, Kejík Z, Brogyányi T, Kaplánek R, Pajková M, Talianová V, et al. Role of mtDNA disturbances in the pathogenesis of Alzheimer’s and Parkinson’s disease. DNA Repair (Amst). 2020;91–92: 102871.

    Article  PubMed  Google Scholar 

  135. Pedrós I, Petrov D, Allgaier M, Sureda F, Barroso E, Beas-Zarate C, et al. Early alterations in energy metabolism in the hippocampus of APPswe/PS1dE9 mouse model of Alzheimer’s disease. Biochim Biophys Acta. 2014;1842:1556–66.

    Article  PubMed  Google Scholar 

  136. Hokama M, Oka S, Leon J, Ninomiya T, Honda H, Sasaki K, et al. Altered expression of diabetes-related genes in Alzheimer’s disease brains: the Hisayama study. Cereb Cortex. 2014;24:2476–88.

    Article  PubMed  Google Scholar 

  137. Wang J, Xiong S, Xie C, Markesbery WR, Lovell MA. Increased oxidative damage in nuclear and mitochondrial DNA in Alzheimer’s disease. J Neurochem. 2005;93:953–62.

    Article  PubMed  CAS  Google Scholar 

  138. Chang SW, Zhang D, Chung HD, Zassenhaus HP. The frequency of point mutations in mitochondrial DNA is elevated in the Alzheimer’s brain. Biochem Biophys Res Commun. 2000;273:203–8.

    Article  PubMed  CAS  Google Scholar 

  139. Hamblet NS, Castora FJ. Elevated levels of the Kearns-Sayre syndrome mitochondrial DNA deletion in temporal cortex of Alzheimer’s patients. Mutat Res. 1997;379:253–62.

    Article  PubMed  CAS  Google Scholar 

  140. Stoccoro A, Siciliano G, Migliore L, Coppedè F. Decreased Methylation of the Mitochondrial D-Loop Region in Late-Onset Alzheimer’s Disease. J Alzheimers Dis. 2017;59:559–64.

    Article  PubMed  CAS  Google Scholar 

  141. Blanch M, Mosquera JL, Ansoleaga B, Ferrer I, Barrachina M. Altered Mitochondrial DNA Methylation Pattern in Alzheimer Disease-Related Pathology and in Parkinson Disease. Am J Pathol. 2016;186:385–97.

    Article  PubMed  CAS  Google Scholar 

  142. Lunnon K, Keohane A, Pidsley R, Newhouse S, Riddoch-Contreras J, Thubron EB, et al. Mitochondrial genes are altered in blood early in Alzheimer’s disease. Neurobiol Aging. 2017;53:36–47.

    Article  PubMed  CAS  Google Scholar 

  143. Wei W, Keogh MJ, Wilson I, Coxhead J, Ryan S, Rollinson S, et al. Mitochondrial DNA point mutations and relative copy number in 1363 disease and control human brains. Acta Neuropathol Commun. 2017;5:13.

    Article  PubMed  PubMed Central  Google Scholar 

  144. Klein H-U, Trumpff C, Yang H-S, Lee AJ, Picard M, Bennett DA, et al. Characterization of mitochondrial DNA quantity and quality in the human aged and Alzheimer’s disease brain. Mol Neurodegener. 2021;16:75.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  145. Xie X, Ma G, Li X, Zhao J, Zhao Z, Zeng J. Activation of innate immune cGAS-STING pathway contributes to Alzheimer’s pathogenesis in 5×FAD mice. Nat Aging. 2023;3:202–12.

    Article  PubMed  CAS  Google Scholar 

  146. Cervera-Carles L, Alcolea D, Estanga A, Ecay-Torres M, Izagirre A, Clerigué M, et al. Cerebrospinal fluid mitochondrial DNA in the Alzheimer’s disease continuum. Neurobiol Aging. 2017;53:192.e1-192.e4.

    Article  PubMed  CAS  Google Scholar 

  147. Podlesniy P, Figueiro-Silva J, Llado A, Antonell A, Sanchez-Valle R, Alcolea D, et al. Low cerebrospinal fluid concentration of mitochondrial DNA in preclinical Alzheimer disease. Ann Neurol. 2013;74:655–68.

    Article  PubMed  CAS  Google Scholar 

  148. Rice AC, Keeney PM, Algarzae NK, Ladd AC, Thomas RR, Bennett JP. Mitochondrial DNA copy numbers in pyramidal neurons are decreased and mitochondrial biogenesis transcriptome signaling is disrupted in Alzheimer’s disease hippocampi. J Alzheimers Dis. 2014;40:319–30.

    Article  PubMed  CAS  Google Scholar 

  149. Rodríguez-Santiago B, Casademont J, Nunes V. Is mitochondrial DNA depletion involved in Alzheimer’s disease? Eur J Hum Genet. 2001;9:279–85.

    Article  PubMed  Google Scholar 

  150. Coskun PE, Beal MF, Wallace DC. Alzheimer’s brains harbor somatic mtDNA control-region mutations that suppress mitochondrial transcription and replication. Proc Natl Acad Sci U S A. 2004;101:10726–31.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  151. Soltys DT, Pereira CPM, Rowies FT, Farfel JM, Grinberg LT, Suemoto CK, et al. Lower mitochondrial DNA content but not increased mutagenesis associates with decreased base excision repair activity in brains of AD subjects. Neurobiol Aging. 2019;73:161–70.

    Article  PubMed  CAS  Google Scholar 

  152. Podlesniy P, Llorens F, Puigròs M, Serra N, Sepúlveda-Falla D, Schmidt C, et al. Cerebrospinal Fluid Mitochondrial DNA in Rapid and Slow Progressive Forms of Alzheimer’s Disease. Int J Mol Sci. 2020;21:6298.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  153. Hirai K, Aliev G, Nunomura A, Fujioka H, Russell RL, Atwood CS, et al. Mitochondrial abnormalities in Alzheimer’s disease. J Neurosci. 2001;21:3017–23.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  154. Lv X, Zhou D, Ge B, Chen H, Du Y, Liu S, et al. Association of Folate Metabolites and Mitochondrial Function in Peripheral Blood Cells in Alzheimer’s Disease: A Matched Case-Control Study. J Alzheimers Dis. 2019;70:1133–42.

    Article  PubMed  CAS  Google Scholar 

  155. Terzioğlu G, Örmeci B, Türksoy Ö, Sayman C, Çınar N, Öztürk GA, et al. Mitochondrial depletion in CD4+ and CD19+ peripheral lymphocytes in early stage Alzheimer’s disease. Mech Ageing Dev. 2017;167:24–9.

    Article  PubMed  Google Scholar 

  156. Silzer T, Barber R, Sun J, Pathak G, Johnson L, O’Bryant S, et al. Circulating mitochondrial DNA: New indices of type 2 diabetes-related cognitive impairment in Mexican Americans. PLoS ONE. 2019;14: e0213527.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  157. Thubron EB, Rosa HS, Hodges A, Sivaprasad S, Francis PT, Pienaar IS, et al. 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. 2019;9:11386.

    Article  PubMed  PubMed Central  Google Scholar 

  158. Lee J-W, Park KD, Im J-A, Kim MY, Lee D-C. Mitochondrial DNA copy number in peripheral blood is associated with cognitive function in apparently healthy elderly women. Clin Chim Acta. 2010;411:592–6.

    Article  PubMed  CAS  Google Scholar 

  159. Lee J-Y, Kim J-H, Lee D-C. Combined Impact of Telomere Length and Mitochondrial DNA Copy Number on Cognitive Function in Community-Dwelling Very Old Adults. Dement Geriatr Cogn Disord. 2017;44:232–43.

    Article  PubMed  CAS  Google Scholar 

  160. Lynch MT, Taub MA, Farfel JM, Yang J, Abadir P, De Jager PL, et al. Evaluating genomic signatures of aging in brain tissue as it relates to Alzheimer’s disease. Sci Rep. 2023;13:14747.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  161. Gorham IK, Reid DM, Sun J, Zhou Z, Barber RC, Phillips NR. Blood-Based mtDNA Quantification Indicates Population-Specific Differences Associated with Alzheimer’s Disease-Related Risk. J Alzheimers Dis. 2024;97:1407–19.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  162. Liou C-W, Chen S-H, Lin T-K, Tsai M-H, Chang C-C. Oxidative Stress Biomarkers and Mitochondrial DNA Copy Number Associated with APOE4 Allele and Cholinesterase Inhibitor Therapy in Patients with Alzheimer’s Disease. Antioxidants (Basel). 2021;10:1971.

    Article  PubMed  CAS  Google Scholar 

  163. Harerimana NV, Paliwali D, Romero-Molina C, Bennett DA, Pa J, Goate A, et al. The role of mitochondrial genome abundance in Alzheimer’s disease. Alzheimers Dement. 2023;19:2069–83.

    Article  PubMed  CAS  Google Scholar 

  164. Coskun PE, Wyrembak J, Derbereva O, Melkonian G, Doran E, Lott IT, et al. Systemic mitochondrial dysfunction and the etiology of Alzheimer’s disease and down syndrome dementia. J Alzheimers Dis. 2010;20(Suppl 2):S293-310.

    Article  PubMed  PubMed Central  Google Scholar 

  165. Delbarba A, Abate G, Prandelli C, Marziano M, Buizza L, Arce Varas N, et al. Mitochondrial Alterations in Peripheral Mononuclear Blood Cells from Alzheimer’s Disease and Mild Cognitive Impairment Patients. Oxid Med Cell Longev. 2016;2016:5923938.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  166. Scopa C, Barnada SM, Cicardi ME, Singer M, Trotti D, Trizzino M. JUN upregulation drives aberrant transposable element mobilization, associated innate immune response, and impaired neurogenesis in Alzheimer’s disease. Nat Commun. 2023;14:8021.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  167. Hou Y, Wei Y, Lautrup S, Yang B, Wang Y, Cordonnier S, et al. NAD+ supplementation reduces neuroinflammation and cell senescence in a transgenic mouse model of Alzheimer’s disease via cGAS-STING. Proc Natl Acad Sci U S A. 2021;118: e2011226118.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  168. Ferecskó AS, Smallwood MJ, Moore A, Liddle C, Newcombe J, Holley J, et al. STING-Triggered CNS Inflammation in Human Neurodegenerative Diseases. Biomedicines. 2023;11:1375.

    Article  PubMed  PubMed Central  Google Scholar 

  169. Gao D, Hao J-P, Li B-Y, Zheng C-C, Miao B-B, Zhang L, et al. Tetrahydroxy stilbene glycoside ameliorates neuroinflammation for Alzheimer’s disease via cGAS-STING. Eur J Pharmacol. 2023;953: 175809.

    Article  PubMed  CAS  Google Scholar 

  170. Microglial cGAS-STING links innate immunity and Alzheimer’s disease. Nat Aging. 2023;3:155–6.

    Article  Google Scholar 

  171. Larrick JW, Mendelsohn AR. Modulation of cGAS-STING Pathway by Nicotinamide Riboside in Alzheimer’s Disease. Rejuvenation Res. 2021;24:397–402.

    Article  PubMed  CAS  Google Scholar 

  172. Nelson TJ, Xu Y. Sting and p53 DNA repair pathways are compromised in Alzheimer’s disease. Sci Rep. 2023;13:8304.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  173. Van Acker ZP, Perdok A, Hellemans R, North K, Vorsters I, Cappel C, et al. Phospholipase D3 degrades mitochondrial DNA to regulate nucleotide signaling and APP metabolism. Nat Commun. 2023;14:2847.

    Article  PubMed  PubMed Central  Google Scholar 

  174. Lee JH, Kanwar B, Lee CJ, Sergi C, Coleman MD. Dapsone is an anticatalysis for Alzheimer’s disease exacerbation. iScience. 2022;25:104274.

  175. Wang S, Wang L, Qin X, Turdi S, Sun D, Culver B, et al. ALDH2 contributes to melatonin-induced protection against APP/PS1 mutation-prompted cardiac anomalies through cGAS-STING-TBK1-mediated regulation of mitophagy. Signal Transduct Target Ther. 2020;5:119.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  176. Wu Z, Tang W, Ibrahim FEEM, Chen X, Yan H, Tao C, et al. Aβ Induces Neuroinflammation and Microglial M1 Polarization via cGAS-STING-IFITM3 Signaling Pathway in BV-2 Cells. Neurochem Res. 2023;48:2881–94.

    Article  PubMed  CAS  Google Scholar 

  177. Xia X, He X, Zhao T, Yang J, Bi Z, Fu Q, et al. Inhibiting mtDNA-STING-NLRP3/IL-1β axis-mediated neutrophil infiltration protects neurons in Alzheimer’s disease. Cell Prolif. 2024;57: e13529.

    Article  PubMed  CAS  Google Scholar 

  178. Liu P, Chen W, Kang Y, Wang C, Wang X, Liu W, et al. Silibinin ameliorates STING-mediated neuroinflammation via downregulation of ferroptotic damage in a sporadic Alzheimer’s disease model. Arch Biochem Biophys. 2023;744: 109691.

    Article  PubMed  CAS  Google Scholar 

  179. Xu Q, Xu W, Cheng H, Yuan H, Tan X. Efficacy and mechanism of cGAMP to suppress Alzheimer’s disease by elevating TREM2. Brain Behav Immun. 2019;81:495–508.

    Article  PubMed  CAS  Google Scholar 

  180. Chung S, Jeong J-H, Park J-C, Han JW, Lee Y, Kim J-I, et al. Blockade of STING activation alleviates microglial dysfunction and a broad spectrum of Alzheimer’s disease pathologies. Exp Mol Med. 2024;56:1936–51.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  181. Carling GK, Fan L, Foxe NR, Norman K, Wong MY, Zhu D, et al. Alzheimer’s disease-linked risk alleles elevate microglial cGAS-associated senescence and neurodegeneration in a tauopathy model. Neuron. 2024;S0896–6273(24):00654–8.

    Google Scholar 

  182. Wilkins HM, Koppel SJ, Weidling IW, Roy N, Ryan LN, Stanford JA, et al. Extracellular Mitochondria and Mitochondrial Components Act as Damage-Associated Molecular Pattern Molecules in the Mouse Brain. J Neuroimmune Pharmacol. 2016;11:622–8.

    Article  PubMed  PubMed Central  Google Scholar 

  183. Zhao Y, Liu B, Xu L, Yu S, Fu J, Wang J, et al. ROS-Induced mtDNA Release: The Emerging Messenger for Communication between Neurons and Innate Immune Cells during Neurodegenerative Disorder Progression. Antioxidants (Basel). 2021;10:1917.

    Article  PubMed  CAS  Google Scholar 

  184. Shi Y, Yamada K, Liddelow SA, Smith ST, Zhao L, Luo W, et al. ApoE4 markedly exacerbates tau-mediated neurodegeneration in a mouse model of tauopathy. Nature. 2017;549:523–7.

    Article  PubMed  PubMed Central  Google Scholar 

  185. Nelson MR, Liu P, Agrawal A, Yip O, Blumenfeld J, Traglia M, et al. The APOE-R136S mutation protects against APOE4-driven Tau pathology, neurodegeneration and neuroinflammation. Nat Neurosci. 2023;26:2104–21.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  186. Chen X, Firulyova M, Manis M, Herz J, Smirnov I, Aladyeva E, et al. Microglia-mediated T cell infiltration drives neurodegeneration in tauopathy. Nature. 2023;615:668–77.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  187. Naguib S, Lopez-Lee C, Torres ER, Lee S-I, Zhu J, Zhu D, et al. APOE3 - R136S mutation confers resilience against tau pathology via cGAS-STING-IFN inhibition. bioRxiv. 2025;2024.04.25.591140.

  188. Leng F, Edison P. Neuroinflammation and microglial activation in Alzheimer disease: where do we go from here? Nat Rev Neurol. 2021;17:157–72.

    Article  PubMed  Google Scholar 

  189. Guo X, Yang L, Wang J, Wu Y, Li Y, Du L, et al. The cytosolic DNA-sensing cGAS-STING pathway in neurodegenerative diseases. CNS Neurosci Ther. 2024;30: e14671.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  190. Cox DJ, Field RH, Williams DG, Baran M, Bowie AG, Cunningham C, et al. DNA sensors are expressed in astrocytes and microglia in vitro and are upregulated during gliosis in neurodegenerative disease. Glia. 2015;63:812–25.

    Article  PubMed  PubMed Central  Google Scholar 

  191. Jeffries AM, Marriott I. Human microglia and astrocytes express cGAS-STING viral sensing components. Neurosci Lett. 2017;658:53–6.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  192. Mathur V, Burai R, Vest RT, Bonanno LN, Lehallier B, Zardeneta ME, et al. Activation of the STING-Dependent Type I Interferon Response Reduces Microglial Reactivity and Neuroinflammation. Neuron. 2017;96:1290-1302.e6.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  193. Reinert LS, Lopušná K, Winther H, Sun C, Thomsen MK, Nandakumar R, et al. Sensing of HSV-1 by the cGAS-STING pathway in microglia orchestrates antiviral defence in the CNS. Nat Commun. 2016;7:13348.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  194. Dai J, Huang Y-J, He X, Zhao M, Wang X, Liu Z-S, et al. Acetylation Blocks cGAS Activity and Inhibits Self-DNA-Induced Autoimmunity. Cell. 2019;176:1447-1460.e14.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  195. Paul BD, Snyder SH, Bohr VA. Signaling by cGAS-STING in Neurodegeneration, Neuroinflammation, and Aging. Trends Neurosci. 2021;44:83–96.

    Article  PubMed  CAS  Google Scholar 

  196. Ida-Hosonuma M, Iwasaki T, Yoshikawa T, Nagata N, Sato Y, Sata T, et al. The Alpha/Beta Interferon Response Controls Tissue Tropism and Pathogenicity of Poliovirus. J Virol. 2005;79:4460.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  197. Sanford SAI, Miller LVC, Vaysburd M, Keeling S, Tuck BJ, Clark J, et al. The type-I interferon response potentiates seeded tau aggregation and exacerbates tau pathology. Alzheimers Dement. 2024;20:1013–25.

    Article  PubMed  CAS  Google Scholar 

  198. Roy ER, Wang B, Wan Y, Chiu G, Cole A, Yin Z, et al. Type I interferon response drives neuroinflammation and synapse loss in Alzheimer disease. J Clin Investig. 2020;130:1912.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  199. Roy ER, Chiu G, Li S, Propson NE, Kanchi R, Wang B, et al. Concerted type I interferon signaling in microglia and neural cells promotes memory impairment associated with amyloid β plaques. Immunity. 2022;55:879-894.e6.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  200. Harry GJ, Kraft AD. Neuroinflammation and microglia: considerations and approaches for neurotoxicity assessment. Expert Opin Drug Metab Toxicol. 2008;4:1265–77.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  201. Ren C, Jin J, Li C, Xiang J, Wu Y, Zhou Y, et al. Metformin inactivates the cGAS-STING pathway through autophagy and suppresses senescence in nucleus pulposus cells. J Cell Sci. 2022;135:jcs259738.

  202. Carling GK, Fan L, Foxe NR, Norman K, Ye P, Wong MY, et al. Alzheimer’s disease-linked risk alleles elevate microglial cGAS-associated senescence and neurodegeneration in a tauopathy model. bioRxiv. 2024;2024.01.24.577107.

  203. Gulen MF, Samson N, Keller A, Schwabenland M, Liu C, Glück S, et al. cGAS-STING drives ageing-related inflammation and neurodegeneration. Nature. 2023;620:374–80.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  204. Liu R-M. Aging, Cellular Senescence, and Alzheimer’s Disease. Int J Mol Sci. 2022;23:1989.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  205. Matsudaira T, Nakano S, Konishi Y, Kawamoto S, Uemura K, Kondo T, et al. Cellular senescence in white matter microglia is induced during ageing in mice and exacerbates the neuroinflammatory phenotype. Commun Biol. 2023;6:665.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  206. Chinta SJ, Woods G, Rane A, Demaria M, Campisi J, Andersen JK. Cellular senescence and the aging brain. Exp Gerontol. 2015;68:3–7.

    Article  PubMed  CAS  Google Scholar 

  207. Dou Z, Ghosh K, Vizioli MG, Zhu J, Sen P, Wangensteen KJ, et al. Cytoplasmic chromatin triggers inflammation in senescence and cancer. Nature. 2017;550:402–6.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  208. Carreno G, Guiho R, Martinez-Barbera JP. Cell senescence in neuropathology: A focus on neurodegeneration and tumours. Neuropathol Appl Neurobiol. 2021;47:359–78.

    Article  PubMed  PubMed Central  Google Scholar 

  209. Si Z, Sun L, Wang X. Evidence and perspectives of cell senescence in neurodegenerative diseases. Biomed Pharmacother. 2021;137: 111327.

    Article  PubMed  CAS  Google Scholar 

  210. Ma X, Xin D, She R, Liu D, Ge J, Mei Z. Novel insight into cGAS-STING pathway in ischemic stroke: from pre- to post-disease. Front Immunol. 2023;14:1275408.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  211. Tchkonia T, Zhu Y, van Deursen J, Campisi J, Kirkland JL. Cellular senescence and the senescent secretory phenotype: therapeutic opportunities. J Clin Invest. 2013;123:966–72.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  212. De Cecco M, Ito T, Petrashen AP, Elias AE, Skvir NJ, Criscione SW, et al. L1 drives IFN in senescent cells and promotes age-associated inflammation. Nature. 2019;566:73–8.

    Article  PubMed  PubMed Central  Google Scholar 

  213. Aguado J, Chaggar HK, Gómez-Inclán C, Shaker MR, Leeson HC, Mackay-Sim A, et al. Inhibition of the cGAS-STING pathway ameliorates the premature senescence hallmarks of Ataxia-Telangiectasia brain organoids. Aging Cell. 2021;20: e13468.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  214. Shippy DC, Ulland TK. Microglial Immunometabolism in Alzheimer’s Disease. Front Cell Neurosci. 2020;14: 563446.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  215. Heneka MT. Inflammasome activation and innate immunity in Alzheimer’s disease. Brain Pathol. 2017;27:220–2.

    Article  PubMed  PubMed Central  Google Scholar 

  216. d’Errico P, Ziegler-Waldkirch S, Aires V, Hoffmann P, Mezö C, Erny D, et al. Microglia contribute to the propagation of Aβ into unaffected brain tissue. Nat Neurosci. 2022;25:20–5.

    Article  PubMed  Google Scholar 

  217. Xie L, Zhang N, Zhang Q, Li C, Sandhu AF, Iii GW, et al. Inflammatory factors and amyloid β-induced microglial polarization promote inflammatory crosstalk with astrocytes. Aging (Albany NY). 2020;12:22538–49.

    PubMed  CAS  Google Scholar 

  218. Huang Y, Liu B, Sinha SC, Amin S, Gan L. Mechanism and therapeutic potential of targeting cGAS-STING signaling in neurological disorders. Mol Neurodegener. 2023;18:79.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  219. Sala Frigerio C, Wolfs L, Fattorelli N, Thrupp N, Voytyuk I, Schmidt I, et al. The Major Risk Factors for Alzheimer’s Disease: Age, Sex, and Genes Modulate the Microglia Response to Aβ Plaques. Cell Rep. 2019;27:1293-1306.e6.

    Article  PubMed  CAS  Google Scholar 

  220. Escoubas CC, Dorman LC, Nguyen PT, Lagares-Linares C, Nakajo H, Anderson SR, et al. Type-I-interferon-responsive microglia shape cortical development and behavior. Cell. 2024;187:1936-1954.e24.

    Article  PubMed  CAS  Google Scholar 

  221. Deczkowska A, Keren-Shaul H, Weiner A, Colonna M, Schwartz M, Amit I. Disease-Associated Microglia: A Universal Immune Sensor of Neurodegeneration. Cell. 2018;173:1073–81.

    Article  PubMed  CAS  Google Scholar 

  222. Yoh SM, Schneider M, Seifried J, Soonthornvacharin S, Akleh RE, Olivieri KC, et al. PQBP1 is a Proximal Sensor of the cGAS-dependent Innate Response to HIV-1. Cell. 2015;161:1293–305.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  223. Jin M, Shiwaku H, Tanaka H, Obita T, Ohuchi S, Yoshioka Y, et al. Tau activates microglia via the PQBP1-cGAS-STING pathway to promote brain inflammation. Nat Commun. 2021;12:6565.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  224. Yeh FL, Hansen DV, Sheng M. TREM2, Microglia, and Neurodegenerative Diseases. Trends Mol Med. 2017;23:512–33.

    Article  PubMed  CAS  Google Scholar 

  225. Villanelo F, Escalona Y, Pareja-Barrueto C, Garate JA, Skerrett IM, Perez-Acle T. Accessing gap-junction channel structure-function relationships through molecular modeling and simulations. BMC Cell Biol. 2017;18:5.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  226. Ablasser A, Schmid-Burgk JL, Hemmerling I, Horvath GL, Schmidt T, Latz E, et al. Cell intrinsic immunity spreads to bystander cells via the intercellular transfer of cGAMP. Nature. 2013;503:530–4.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  227. Zhou C, Chen X, Planells-Cases R, Chu J, Wang L, Cao L, et al. Transfer of cGAMP into Bystander Cells via LRRC8 Volume-Regulated Anion Channels Augments STING-Mediated Interferon Responses and Anti-viral Immunity. Immunity. 2020;52:767-781.e6.

    Article  PubMed  CAS  Google Scholar 

  228. Elzinga SE, Henn R, Murdock BJ, Kim B, Hayes JM, Mendelson F, et al. cGAS/STING and innate brain inflammation following acute high-fat feeding. Front Immunol. 2022;13:1012594.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  229. Chang H, Li Z, Zhang W, Lin C, Shen Y, Zhang G, et al. Transfer of cGAMP from neuron to microglia activates microglial type I interferon responses after subarachnoid hemorrhage. Cell Commun Signal. 2024;22:3.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  230. Koulakoff A, Mei X, Orellana JA, Sáez JC, Giaume C. Glial connexin expression and function in the context of Alzheimer’s disease. Biochim Biophys Acta. 2012;1818:2048–57.

    Article  PubMed  CAS  Google Scholar 

  231. Koulakoff A, Ezan P, Giaume C. Neurons control the expression of connexin 30 and connexin 43 in mouse cortical astrocytes. Glia. 2008;56:1299–311.

    Article  PubMed  Google Scholar 

  232. Mei X, Ezan P, Giaume C, Koulakoff A. Astroglial connexin immunoreactivity is specifically altered at β-amyloid plaques in β-amyloid precursor protein/presenilin1 mice. Neuroscience. 2010;171:92–105.

    Article  PubMed  CAS  Google Scholar 

  233. Kalani K, Chaturvedi P, Chaturvedi P, Kumar Verma V, Lal N, Awasthi SK, et al. Mitochondrial mechanisms in Alzheimer’s disease: Quest for therapeutics. Drug Discov Today. 2023;28: 103547.

    Article  PubMed  CAS  Google Scholar 

  234. Haag SM, Gulen MF, Reymond L, Gibelin A, Abrami L, Decout A, et al. Targeting STING with covalent small-molecule inhibitors. Nature. 2018;559:269–73.

    Article  PubMed  CAS  Google Scholar 

  235. Steinhagen F, Zillinger T, Peukert K, Fox M, Thudium M, Barchet W, et al. Suppressive oligodeoxynucleotides containing TTAGGG motifs inhibit cGAS activation in human monocytes. Eur J Immunol. 2018;48:605–11.

    Article  PubMed  CAS  Google Scholar 

  236. Li Q, Cao Y, Dang C, Han B, Han R, Ma H, et al. Inhibition of double-strand DNA-sensing cGAS ameliorates brain injury after ischemic stroke. EMBO Mol Med. 2020;12: e11002.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  237. Wang M, Sooreshjani MA, Mikek C, Opoku-Temeng C, Sintim HO. Suramin potently inhibits cGAMP synthase, cGAS, in THP1 cells to modulate IFN-β levels. Future Med Chem. 2018;10:1301–17.

    Article  PubMed  CAS  Google Scholar 

  238. An J, Woodward JJ, Sasaki T, Minie M, Elkon KB. Cutting edge: Antimalarial drugs inhibit IFN-β production through blockade of cyclic GMP-AMP synthase-DNA interaction. J Immunol. 2015;194:4089–93.

    Article  PubMed  CAS  Google Scholar 

  239. An J, Woodward JJ, Lai W, Minie M, Sun X, Tanaka L, et al. Inhibition of Cyclic GMP-AMP Synthase Using a Novel Antimalarial Drug Derivative in Trex1-Deficient Mice. Arthritis Rheumatol. 2018;70:1807–19.

    Article  PubMed  CAS  Google Scholar 

  240. Wang X, Wang Y, Cao A, Luo Q, Chen D, Zhao W, et al. Development of cyclopeptide inhibitors of cGAS targeting protein-DNA interaction and phase separation. Nat Commun. 2023;14:6132.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  241. Padilla-Salinas R, Sun L, Anderson R, Yang X, Zhang S, Chen ZJ, et al. Discovery of Small-Molecule Cyclic GMP-AMP Synthase Inhibitors. J Org Chem. 2020;85:1579–600.

    Article  PubMed  CAS  Google Scholar 

  242. Green JP, El-Sharkawy LY, Roth S, Zhu J, Cao J, Leach AG, et al. Discovery of an inhibitor of DNA-driven inflammation that preferentially targets the AIM2 inflammasome. iScience. 2023;26:106758.

  243. Vincent J, Adura C, Gao P, Luz A, Lama L, Asano Y, et al. Small molecule inhibition of cGAS reduces interferon expression in primary macrophages from autoimmune mice. Nat Commun. 2017;8:750.

    Article  PubMed  PubMed Central  Google Scholar 

  244. Hall J, Brault A, Vincent F, Weng S, Wang H, Dumlao D, et al. Discovery of PF-06928215 as a high affinity inhibitor of cGAS enabled by a novel fluorescence polarization assay. PLoS ONE. 2017;12: e0184843.

    Article  PubMed  PubMed Central  Google Scholar 

  245. Lama L, Adura C, Xie W, Tomita D, Kamei T, Kuryavyi V, et al. Development of human cGAS-specific small-molecule inhibitors for repression of dsDNA-triggered interferon expression. Nat Commun. 2019;10:2261.

    Article  PubMed  PubMed Central  Google Scholar 

  246. Zhao W, Xiong M, Yuan X, Li M, Sun H, Xu Y. In Silico Screening-Based Discovery of Novel Inhibitors of Human Cyclic GMP-AMP Synthase: A Cross-Validation Study of Molecular Docking and Experimental Testing. J Chem Inf Model. 2020;60:3265–76.

    Article  PubMed  CAS  Google Scholar 

  247. Tan J, Wu B, Chen T, Fan C, Zhao J, Xiong C, et al. Synthesis and Pharmacological Evaluation of Tetrahydro-γ-carboline Derivatives as Potent Anti-inflammatory Agents Targeting Cyclic GMP-AMP Synthase. J Med Chem. 2021;64:7667–90.

    Article  PubMed  CAS  Google Scholar 

  248. Li J, Xiong M, Liu J, Zhang F, Li M, Zhao W, et al. Discovery of novel cGAS inhibitors based on natural flavonoids. Bioorg Chem. 2023;140: 106802.

    Article  PubMed  CAS  Google Scholar 

  249. Liu Z-S, Cai H, Xue W, Wang M, Xia T, Li W-J, et al. G3BP1 promotes DNA binding and activation of cGAS. Nat Immunol. 2019;20:18–28.

    Article  PubMed  CAS  Google Scholar 

  250. Yu Z, Zheng L, Geng Y, Zhang Y, Wang Y, You G, et al. FTO alleviates cerebral ischemia/reperfusion-induced neuroinflammation by decreasing cGAS mRNA stability in an m6A-dependent manner. Cell Signal. 2023;109: 110751.

    Article  PubMed  CAS  Google Scholar 

  251. Sun H, Huang Y, Mei S, Xu F, Liu X, Zhao F, et al. A Nuclear Export Signal Is Required for cGAS to Sense Cytosolic DNA. Cell Rep. 2021;34: 108586.

    Article  PubMed  CAS  Google Scholar 

  252. Liu M, Li Y, Han S, Wang H, Li J. Activin A alleviates neuronal injury through inhibiting cGAS-STING-mediated autophagy in mice with ischemic stroke. J Cereb Blood Flow Metab. 2023;43:736–48.

    Article  PubMed  CAS  Google Scholar 

  253. Chauhan C, Kaundal RK. The role of cGAS-STING signaling in ischemic stroke: From immune response to therapeutic targeting. Drug Discov Today. 2023;28: 103792.

    Article  PubMed  CAS  Google Scholar 

  254. Li S, Hong Z, Wang Z, Li F, Mei J, Huang L, et al. The Cyclopeptide Astin C Specifically Inhibits the Innate Immune CDN Sensor STING. Cell Rep. 2018;25:3405-3421.e7.

    Article  PubMed  CAS  Google Scholar 

  255. Siu T, Altman MD, Baltus GA, Childers M, Ellis JM, Gunaydin H, et al. Discovery of a Novel cGAMP Competitive Ligand of the Inactive Form of STING. ACS Med Chem Lett. 2019;10:92–7.

    Article  PubMed  CAS  Google Scholar 

  256. Hong Z, Mei J, Li C, Bai G, Maimaiti M, Hu H, et al. STING inhibitors target the cyclic dinucleotide binding pocket. Proc Natl Acad Sci U S A. 2021;118: e2105465118.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  257. Hansen AL, Buchan GJ, Rühl M, Mukai K, Salvatore SR, Ogawa E, et al. Nitro-fatty acids are formed in response to virus infection and are potent inhibitors of STING palmitoylation and signaling. Proc Natl Acad Sci U S A. 2018;115:E7768–75.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  258. Mukai K, Konno H, Akiba T, Uemura T, Waguri S, Kobayashi T, et al. Activation of STING requires palmitoylation at the Golgi. Nat Commun. 2016;7:11932.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  259. Vinogradova EV, Zhang X, Remillard D, Lazar DC, Suciu RM, Wang Y, et al. An Activity-Guided Map of Electrophile-Cysteine Interactions in Primary Human T Cells. Cell. 2020;182:1009-1026.e29.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  260. Kwon D, Park E, Sesaki H, Kang S-J. Carbonyl cyanide 3-chlorophenylhydrazone (CCCP) suppresses STING-mediated DNA sensing pathway through inducing mitochondrial fission. Biochem Biophys Res Commun. 2017;493:737–43.

    Article  PubMed  CAS  Google Scholar 

  261. Li W, Li Y, Kang J, Jiang H, Gong W, Chen L, et al. 4-octyl itaconate as a metabolite derivative inhibits inflammation via alkylation of STING. Cell Rep. 2023;42: 112145.

    Article  PubMed  CAS  Google Scholar 

  262. Prabakaran T, Troldborg A, Kumpunya S, Alee I, Marinković E, Windross SJ, et al. A STING antagonist modulating the interaction with STIM1 blocks ER-to-Golgi trafficking and inhibits lupus pathology. EBioMedicine. 2021;66: 103314.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  263. Jia M, Qin D, Zhao C, Chai L, Yu Z, Wang W, et al. Redox homeostasis maintained by GPX4 facilitates STING activation. Nat Immunol. 2020;21:727–35.

    Article  PubMed  CAS  Google Scholar 

  264. Gao J, Zheng M, Wu X, Zhang H, Su H, Dang Y, et al. CDK inhibitor Palbociclib targets STING to alleviate autoinflammation. EMBO Rep. 2022;23: e53932.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  265. Liu J, Yuan L, Ruan Y, Deng B, Yang Z, Ren Y, et al. Novel CRBN-Recruiting Proteolysis-Targeting Chimeras as Degraders of Stimulator of Interferon Genes with In Vivo Anti-Inflammatory Efficacy. J Med Chem. 2022;65:6593–611.

    Article  PubMed  CAS  Google Scholar 

  266. Liu H, Wang F, Cao Y, Dang Y, Ge B. The multifaceted functions of cGAS. J Mol Cell Biol. 2022;14:mjac031.

  267. Li Q, Wu P, Du Q, Hanif U, Hu H, Li K. cGAS-STING, an important signaling pathway in diseases and their therapy. MedComm. 2020;2024(5): e511.

    Google Scholar 

  268. Fryer AL, Abdullah A, Taylor JM, Crack PJ. The Complexity of the cGAS-STING Pathway in CNS Pathologies. Front Neurosci. 2021;15: 621501.

    Article  PubMed  PubMed Central  Google Scholar 

  269. Wang R, Hussain A, Guo Q, Ma M. cGAS-STING at the crossroads in cancer therapy. Crit Rev Oncol Hematol. 2024;193: 104194.

    Article  PubMed  Google Scholar 

  270. Joshi B, Joshi JC, Mehta D. Regulation of cGAS Activity and Downstream Signaling. Cells. 2022;11:2812.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  271. Zhang Z, Yuan B, Bao M, Lu N, Kim T, Liu Y-J. The helicase DDX41 senses intracellular DNA mediated by the adaptor STING in dendritic cells. Nat Immunol. 2011;12:959–65.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  272. Singh RS, Vidhyasagar V, Yang S, Arna AB, Yadav M, Aggarwal A, et al. DDX41 is required for cGAS-STING activation against DNA virus infection. Cell Rep. 2022;39: 110856.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  273. Zhu Z, Lu H, Jin L, Gao Y, Qian Z, Lu P, et al. C-176 loaded Ce DNase nanoparticles synergistically inhibit the cGAS-STING pathway for ischemic stroke treatment. Bioact Mater. 2023;29:230–40.

    PubMed  PubMed Central  CAS  Google Scholar 

  274. Nance E, Pun SH, Saigal R, Sellers DL. Drug delivery to the central nervous system. Nat Rev Mater. 2022;7:314–31.

    Article  PubMed  CAS  Google Scholar 

  275. Terstappen GC, Meyer AH, Bell RD, Zhang W. Strategies for delivering therapeutics across the blood-brain barrier. Nat Rev Drug Discov. 2021;20:362–83.

    Article  PubMed  CAS  Google Scholar 

  276. Cai H, Pang Y, Ren Z, Fu X, Jia L. Delivering synaptic protein mRNAs via extracellular vesicles ameliorates cognitive impairment in a mouse model of Alzheimer’s disease. BMC Med. 2024;22:138.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

Download references

Acknowledgements

Biorender was used to create the images used in Figs. 1~ 5.

Authors’ information

Not applicable.

Funding

This project was supported by the National Natural Science Foundation of China (Grant Nos. 81870825 and 82071194), the Beijing Brain Initiative from the Beijing Municipal Science & Technology Commission (Grant No. Z201100005520016), the Beijing Municipal Natural Science Foundation (Grant No. 7202061), the Capital’s Funds for Health Improvement and Research (Grant No.2022–2-2017), and the STI 2030-Major Projects (Grant No. 2022ZD0211600, 2022ZD0211605).

Author information

Authors and Affiliations

Authors

Contributions

LJ and SQ contributed to the conception and the structure of the manuscript. The manuscript was drafted by SQ, and then commented upon and edited by all authors. All authors read and approved the final manuscript.

Corresponding author

Correspondence to Longfei Jia.

Ethics declarations

Ethics approval and consent to participate

Not applicable.

Consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Additional information

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Quan, S., Fu, X., Cai, H. et al. The neuroimmune nexus: unraveling the role of the mtDNA-cGAS-STING signal pathway in Alzheimer’s disease. Mol Neurodegeneration 20, 25 (2025). https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s13024-025-00815-2

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s13024-025-00815-2

Keywords