Skip to main content
Cellular and Molecular Neurobiology logoLink to Cellular and Molecular Neurobiology
. 2025 Apr 7;45:33. doi: 10.1007/s10571-025-01550-4

The STING Signaling: A Novel Target for Central Nervous System Diseases

Min Song 1,#, Jianxun Ren 1,#, Zhipeng Zhu 1,2,#, Zhaohui Yi 1, Chengyun Wang 1, Lirong Liang 1, Jiahui Tian 1, Guofu Mao 1, Guohua Mao 1,, Min Chen 1,
PMCID: PMC11977075  PMID: 40195137

Abstract

The canonical cyclic GMP-AMP (cGAMP) synthase (cGAS)-Stimulator of Interferon Genes (STING) pathway has been widely recognized as a crucial mediator of inflammation in many diseases, including tumors, infections, and tissue damage. STING signaling can also be activated in a cGAS- or cGAMP-independent manner, although the specific mechanisms remain unclear. In-depth studies on the structural and molecular biology of the STING pathway have led to the development of therapeutic strategies involving STING modulators and their targeted delivery. These strategies may effectively penetrate the blood–brain barrier (BBB) and target STING signaling in multiple central nervous system (CNS) diseases in humans. In this review, we outline both canonical and non-canonical pathways of STING activation and describe the general mechanisms and associations between STING activity and CNS diseases. Finally, we discuss the prospects for the targeted delivery and clinical application of STING agonists and inhibitors, highlighting the STING signaling pathway as a novel therapeutic target in CNS diseases.

Graphical Abstract

graphic file with name 10571_2025_1550_Figa_HTML.jpg

Keywords: STING, cGAS-STING, Central nervous system, STING modulators, Drug delivery

STING Signaling Pathways

The cGAS-STING Pathway

The cGAS, first reported in 2012 (Sun et al. 2013), is a cytosolic DNA sensor that identifies exogenous DNA (from dying cells, exosomes, DNA viruses, retroviruses, and bacteria) and mitochondrial DNA (mtDNA) (Galluzzi et al. 2018). In mammals, cGAS contains a long amino-terminal unstructured region followed by a nucleotidyltransferase domain, which is activated by the binding of the sugar-phosphate backbone of double-stranded DNA (Sun and Hornung 2022). Once activated, cGAS induces conformational changes that lead to the cyclization of ATP and GTP into the second messenger, 2′3′- cGAMP (Gui et al. 2019; Zhang et al. 2019). Subsequently, the second messenger cGAMP translocates to the endoplasmic reticulum (ER) to activate the stimulator of interferon genes (STING). Then STING transfers to ER-Golgi intermediate compartments and forms tetramers by a higher-order oligomerization (Kwon and Bakhoum 2020). Activated STING plays a crucial role in engaging both the canonical and non-canonical NF-κB signaling pathways. In the canonical signaling pathway, STING recruits the IκB kinase (IKK) complex, which consists of IKKα, IKKβ, and NEMO. This complex phosphorylates IκBα, leading to the release of the RELA (p65)/p50 dimer, which then translocates to the nucleus to initiate the transcription of pro-inflammatory genes (Abe and Barber 2014). On the other hand, in the non-canonical signaling pathway, STING activates NF-κB-inducing kinase (NIK, mitogen-activated protein kinase kinase kinase 14,MAP3K14). NIK phosphorylates IKKα, which processes p100 into p52. This processing allows the RELB/p52 dimer to activate different transcriptional programs (Hou et al. 2018). STING is also phosphorylated by tank-binding kinase 1 (TBK1), which recruits interferon (IFN) regulatory factor 3 (IRF3) to induce expression of type I IFN (IFN-I) and other cytokines (Liu et al. 2015; Sun and Hornung 2022). ER withdrawal is necessary for trafficking-mediated STING degradation. STING is transported to the vesicles and rapidly turns off downstream signaling, and then STING vesicles are assigned to Rab7-positive endolysosomes to degrade (Gonugunta et al. 2017) (Fig. 1).

Fig. 1.

Fig. 1

The cGAS-STING signaling pathway. The cGAS is activated following identifying exogenous or endogenous DNA, which catalyzes ATP and GTP into cGAMP. cGAMP binds to the endoplasmic reticulum adaptor protein STING to initiate the signaling cascade. Subsequently, activated STING transfers to endoplasmic reticulum-Golgi intermediate compartments and to the Golgi apparatus, where it recruits IKK to mediate canonical NF-κB targets, and also recruits TBK1 in parallel, which phosphorylates STING and IRF3 to induce type I IFN and other cytokines. Meanwhile, STING also activates NF-κB-inducing kinase (NIK, mitogen-activated protein kinase kinase kinase 14,MAP3K14) mediated non-canonical NF-κB pathway. Finally, STING is transported to the vesicles and speedily turns off downstream signaling, subsequently STING vesicles are assigned to endolysosomes to degrade

The cGAS- or cGAMP-Independent STING Pathway

The cGAS—STING pathway is the canonical mode of STING activation. However, not all STING signaling is activated by cGAS. Following cGAMP—triggered translocation, Armadillo—like helical domain—containing protein 3 interacts with STING at the Golgi and mediates the recruitment of phosphatidylinositol 4—kinase beta (PI4KB) to synthesize PI4P, which is involved by PI4P—binding proteins in the trafficking of STING from the Golgi to endosomes. Abnormally elevated cellular PI4P has been shown to lead to cGAS-independent STING activation (Fang et al. 2023). STING interacts with the lysosomal membrane protein Niemann–Pick C1 and is recruited to the lysosome for degradation. However, in the absence of Niemann–Pick C1, the lysosomal degradation of STING is blocked leading to increased STING signaling, which occurs independently of cGAS (Chu et al. 2021). Beyond NPC1, additional factors regulate lysosomal STING degradation. For example, the E3 ubiquitin ligase TRIM29 directly ubiquitinates STING, promoting its lysosomal degradation, while TRIM29 deficiency stabilizes STING and amplifies its signaling (Zhang et al. 2020; Xie et al. 2023). These findings highlight that dysregulated lysosomal homeostasis broadly impacts STING activity through multiple mechanisms.

Other DNA sensors that detect lysosomal-localized DNA and activate STING signaling have been described (Motani et al. 2015). For example, DNA binding protein IFI16 and DNA damage response factors ATM and PARP-1 can also induce cGAS-independent STING activation (Dunphy et al. 2018), while DNA-dependent protein kinase initiates cGAS-independent IFN-I responses (Taffoni et al. 2023). Furthermore, the STING/IRF7 pathway has been proved to mediate DNA vaccine immunogenicity independent of cGAS (Suschak et al. 2016). Effector memory T cells trigger dsDNA breaks and induce cGAS-independent STING signaling to drive innate inflammation (Meibers et al. 2023). Functionally, Brucella abortus, influenza A virus, and streptococcal M protein have all been shown to trigger STING but not cGAS to induce host protection (Holm et al. 2016; Costa Franco et al. 2018; Movert et al. 2018). To date, activation of STING in a cGAS- and cGAMP-independent manner remains poorly understood.

STING in Brain Tumors

Anti-tumor Effects

STING signaling plays an important role in brain tumors, and functional cytoplasmic DNA sensing via cGAS-STING pathway is lacking in the vast majority of neuroblastoma cell lines (Wolpaw et al. 2022). STING mainly enhances the signal of IFN-I in the tumor microenvironment and exerts anti-tumor effects. IFN-I mainly comes from CD11b+ brain-infiltrating leukocytes in de novo gliomas in mice (Ohkuri et al. 2014; Chen et al. 2022). Both PP2Ac/STRN4 and PTEN regulates STING/IFN-I signaling in the tumor microenvironment (Ho et al. 2023; Mondal et al. 2023; Dogan et al. 2024). Hypoxia promotes the release of extracellular vesicles in glioblastoma cells and miR-25/93 expression in the extracellular vesicle, subsequently, extracellular vesicle carrying miR-25/93 were absorbed by macrophages, resulting in impaired STING/IFN-I signaling (Tankov et al. 2024), Manganese can exacerbate ionizing radiation-induced signaling damage (He et al. 2023b).

Adaptive immune cell infiltration (T cells, dendritic cells, and natural killer cells) is also an important pathway through which STING exerts its effects. Temozolomide enhances DNA damage repair by upregulating GBP3 and STING and its function is enhanced in the presence of PTEN protein (Xu et al. 2022; Yildirim et al. 2024). The combination of CD47 blockade and temozolomide enhances the antigen cross-presentation and activation of STING in antigen-presenting cells, leading to more effective T-cell initiation (von Roemeling et al. 2020). Not only N-Myc inhibition and Tumor Treating Fields (Layer et al. 2017; Chen et al. 2022) but also beta- FAM131B-AS2 inhibition, mangostin and HA-MSA2, can also increase T cell activation and recruitment through STING signaling (Chellen et al. 2024; Wang et al. 2024c; Yang et al. 2024b). In addition, OMA1, GNB4 silencing, and H3.3-G34 mutations can inhibit tumor proliferation, migration, and invasion by activating STING signaling (Haase et al. 2022; Gao and Yang 2024; Zhu et al. 2024).

In animal experiments, STING agonists (IACS-8779 and 8803) demonstrate improved efficacy, tolerability, and extended median survival. Clinically, STING agonists such as ADU-S100 (MIW815) and MK-1454 have advanced into phase I/II trials for advanced solid tumors. However, monotherapy showed limited efficacy, likely due to immunosuppressive tumor microenvironments and poor intratumoral drug delivery. Recent combinatorial strategies (e.g., STING agonists with PD-1 inhibitors or radiotherapy) achieved enhanced antitumor responses in trials (NCT03172936, NCT03010176), underscoring the need for optimized delivery systems and rational therapeutic combinations (Wang et al. 2024a). In humans, STING is markedly increased in high-grade gliomas with high recurrence risk (Zhong et al. 2024), PCBP2 upregulation suppresses cGAS/STING signaling in glioma patients(Chen et al. 2022). However, STING regulators may exhibit species-specific effects (Boudreau et al. 2021; Najem et al. 2024). For instance, murine STING has been recognized as a molecular target of ASA404 (also known as 5,6-dimethylxanthenone-4-acetic acid (DMXAA)) and binds directly to ASA404 resulting in potent anti-tumor activity through activation of TBK1 and IRF3 signaling pathways. However, human STING does not bind to and interact with ASA404 to activate TBK1–IRF3 signaling (Conlon et al. 2013, Bähr et al. 2017). Besides, ASA404 as a vascular disrupting agent has been shown to cause necrosis and hemorrhage, as well as reduce tumor growth and proliferation in subcutaneous glioma, but has no relevant activity in situ brain tumors (Bähr et al. 2017). These findings highlight the potential of STING as an anti-tumor target but underscore the challenges in translating preclinical results to human therapies.

Targeted Delivery System

The BBB is a major obstacle limiting the distribution and targeting efficiency of therapeutic drugs to the brain. Studies addressing this issue have demonstrated that nanoparticles loaded with STING agonists significantly enhance STING-mediated remodeling of the immune microenvironment, potentiate anti-tumor immunity, and inhibit tumor growth in gliomas, glioblastoma multiforme, and neuroblastoma (Wang-Bishop et al. 2020; Bielecki et al. 2021; Wang et al. 2022a; Yang et al. 2022). Currently, various targeted delivery systems have been developed to improve brain tumor therapy, including dendritic cell-tumor hybrid cell-derived chimeric exosomes (Bao et al. 2024), Trojan horse nanocapsule (Zhou et al. 2022), antimiR-25/cGAMP nano complexes (Petrovic et al. 2024), copper-coordination driven brain-targeting nano assembly (Chen et al. 2024), vitamin D3-inserted lipid hybrid neutrophil membrane biomimetic multimodal nanoplatforms (He et al. 2024), a manganese-based nanodriver (Zhang et al. 2024), a pH-responsive and guanidinium-rich nanoadjuvant (Lu et al. 2025), sequential release hydrolipo system (Yu et al. 2025), and bridging-lipid nanoparticle (Zhang et al. 2023a).

These findings indicate that nanoparticles show promising efficacy as targeted delivery systems for brain tumor therapy (Fig. 2). Owing to the BBB, many drugs and modulators fail to reach the brain at therapeutic concentrations, leading to suboptimal treatment outcomes. By contrast, nanoparticles loaded with therapeutic agents can effectively cross the BBB and deliver precise treatment to affected areas. While further research is needed to develop safer and more effective nanomaterials, potential toxicity concerns—such as reproductive toxicity, metabolic dysregulation, and abnormal gene expression—must also be thoroughly investigated. The mechanisms of STING agonists are discussed in detail in "Microglia and Neuroinflammation".

Fig. 2.

Fig. 2

Schematic diagram of nanoparticles penetrating the BBB. The pathway of nanoparticles passing through BBB to reach the brain: (a) transcellular pathway, (b) paracellular pathway, (c) endocytosis, and (d) receptor-mediated endocytosis

Tumor Brain Metastasis

Protocadherin 7 promotes the assembly of connexin 43-based carcinoma–astrocyte gap junctions, which allow metastatic cancer cells in the brain to transfer cGAMP into astrocytes to activate the STING pathway and release inflammatory cytokines including IFN-I and tumor necrosis factor (TNF). These cytokines subsequently activate NF-κB and STAT1 signaling in brain metastatic cells to promote cancer cell survival, growth, and proliferation (Chen et al. 2016, 2017). Chromosome segregation errors lead to chromosomal instability and a preponderance of micronuclei. Micronuclear rupture gives rise to the release of genomic DNA into the cytosol, and subsequent activation of the cGAS-STING pathway and downstream non-canonical NF-κB pathway. Tumor cells displaying chromosomal instability may chronically activate innate immune pathways to boost invasion and metastasis of cancer cells via disruption of the disruption of epithelial responses to cytosolic DNA (Bakhoum et al. 2018). Perhaps, whether STING activation leads to tumor inhibition or promotion may be related to activation intensity or cell types (Gulen et al. 2017). Cancer metastasis is the principal reason for cancer-associated deaths due to the lack of effective treatments. Thus, there is a critical need to increase the survival rates of patients with cancer metastasis, for which STING may hold therapeutic potential.

STING in Stroke

Microglia and Neuroinflammation

Microglia, the primary resident immune cells in the CNS, perform continuous immune surveillance, even in a resting state (M0). Upon stimulation, they polarize into pro-inflammatory (M1) or anti-inflammatory (M2) phenotypes (Li et al. 2022). In the CNS, STING is predominantly expressed in microglia. Following subarachnoid hemorrhage, cGAMP is transferred from neuron to microglia, activatING microglial IFN-I responses (Chang et al. 2024). Microglial STING expression is markedly upregulated after ischemic stroke and subarachnoid hemorrhage. Mechanistically, STING activation polarizes microglia toward the M1 phenotype, whereas inhibition of cGAS or STING (e.g., using A151, C-176, or sulfasalazine) promotes an M2 shift, exerting anti-inflammatory effects (Kong et al. 2022; Shi et al. 2022; Li et al. 2024b; Liu et al. 2024c).

After a subarachnoid hemorrhage, microglial STING recruits and activates TBK1. TBK1 subsequently undergoes autophosphorylation and phosphorylates STING, forming a feedforward loop that sustains TBK1-STING signaling. This axis inhibits AMPK activity, driving microglial activation and M1 polarization (Peng et al. 2020). Interestingly, miR-340-5p directly inhibits STING expression and TBK1 phosphorylation to attenuate neuroinflammation (Song et al. 2022). Studies have also indicated that inhibiting STING signaling after cerebral ischemic stroke not only leads to a microglial polarization switch to the M2 phenotype, microglia-mediated synapse engulfment and improves damaged neurological function (Jiang et al. 2021; Wu et al. 2024) but also alleviates NLRP3 inflammasome-mediated brain injury and microglial pyroptosis (Li et al. 2023).

The mtDNA released into the microglial cytoplasm following cerebral ischemia/reperfusion injury has been demonstrated to activate STING and subsequently facilitate microglial polarization towards the M1 phenotype by downstream pathways, including IRF3 and NF-κB (Kong et al. 2022; Li et al. 2024a). Beyond mtDNA, histone deacetylase 3 and ceramide have also been shown to activate the cGAS-STING pathway to promote the formation of a pro-inflammatory microenvironment. Mechanistically, the former promotes the transcription of cGAS through deacetylation of NF-κB p65 in the microglia, leading to the upregulation of cGAS-STING-mediated neuroinflammation (Liao et al. 2020; Huang et al. 2025). Conversely, α-lipoic acid, FGF2, and iron Q-pretreated mesenchymal stem cells suppress neuroinflammation by inhibiting cGAS-STING (Lin et al. 2024; Ma et al. 2024; Yang et al. 2024a). Downregulating cGAS/STING/NF-κB p65 axis through targeting the JAK2-STAT3 pathway ameliorates neuroinflammation and neuronal senescence (Zhang et al. 2023b). Furthermore, m6A modification increases cGAS mRNA stability to upregulate the STING/NF-κB pathway resulting in aggravation of microglia-mediated neuroinflammation, while fat mass and obesity-associated protein can abrogate these detrimental effects in an m6A-dependent manner (Yu et al. 2023). Collectively, microglia are central to neuroinflammation and post-stroke neuroprotection, making them promising therapeutic targets.

Oxidative Stress, Autophagy and Apoptosis

STING inhibition reduces NLRP3 inflammasome activity and inflammatory cytokine production, ameliorating neuroinflammation, oxidative stress, neuronal apoptosis, microglial pyroptosis, and neurobehavioral deficits after cerebral venous sinus thrombosis (Ding et al. 2022). The cGAS-STING pathway exacerbates inflammation, autophagy, apoptosis, and oxidative stress via NCOA4-mediated ferritinophagy, worsening cerebral ischemia/reperfusion injury (Li et al. 2023; Zhao et al. 2024; Sui et al. 2025). Ginsenoside Rd inhibits ferroptosis to alleviate early brain injury through the cGAS/STING/DHODH pathway (Jiang et al. 2025). Similarly, 25-hydroxycholesterol regulates the mTOR pathway by reducing STING activity, which inhibits autophagy, apoptosis, oxidative injury, and inflammation, and eventually attenuates cerebral ischemia/reperfusion injury (Lin et al. 2021). The neuroprotective factor, Activin A, markedly suppresses cGAS-STING-mediated autophagy rather than mTOR-dependent autophagy by activating the PI3K-PKB pathway, and thus alleviates neuronal injury after ischemic stroke (Liu et al. 2023a). In a neonatal hypoxia–ischemia rat model, activating cGAS/STING signaling via LINE-1-derived cytosolic DNA was found to contribute to delayed neurodegeneration, with cortical neurons more affected than hippocampal neurons (Gamdzyk et al. 2020). STING agonists (DMXAA and CMA) significantly decrease oxygen and glucose deprivation-induced cell death in vitro through a canonical STING-TBK1-IRF3 axis. Blocking the cGAS-STING pathway promotes remyelination, thereby facilitating post-stroke functional recovery within an extended treatment window (Maimaiti et al. 2024). Likewise, tilorone as a DNA intercalator stabilizes cytosolic DNA and induces a protective IFN-I response via the same pathway (Kundu et al. 2022).

Vascular Destruction and BBB Breakdown

Neutrophils accumulate in the brain after stroke, releasing neutrophil extracellular traps (NETs), which induce the activation of STING and downstream signaling molecules including TBK1, IRF3, and IFN-Iβ, resulting in impaired vascular remodeling and revascularization. Peptidylarginine deiminase 4 is integral for the formation of NETs, and its inhibition has been manifested to reduce the STING-mediated IFN-I response, thereby reducing the breakdown of the BBB and enhancing vascular regeneration and repair (Kang et al. 2020). NETs also promote tissue plasminogen activator-induced brain hemorrhage via activating the cGAS-STING signaling and IFN-I response, which eventually leads to the destruction of the vasculature and integrity of the BBB. These effects can be reversed following treatment with DNase I or by inhibiting PAD4 (Wang et al. 2021) Smart liposomal nanocarriers targeting NETs and cGAS-STING mitigate ischemic penumbra and improve stroke outcomes (Sun et al. 2023). Maintaining the integrity of the vasculature and BBB is essential for CNS homeostasis. Collectively, targeting NETs or the cGAS-STING signaling is a prospective therapy to protect against the destruction of blood vessels and the BBB after stroke.

STING in Degenerative Diseases

Human Neurodegenerative Diseases

Elevated STING and cGAMP expression has severally been observed in the substantia nigra pars compacta of PD patients (Hinkle et al. 2022) and in the spinal cord samples of ALS patients (Yu et al. 2020). The cGAS-STING- induced IFN-I was found to be increased together with type III IFN in the peripheral blood and plasma of A-T patients (Hartlova et al. 2015; Gul et al. 2018). In addition, the STING-IFN-β axis was shown to be significantly downregulated in patients with relapsing–remitting multiple sclerosis (Masanneck et al. 2020). Meanwhile, activation of the cGAS-STING pathway in microglia can be detected in human AD and aged mouse brain. Furthermore, studies have demonstrated that the application of STING inhibitor H-151 can effectively inhibit the activation of the cGAS-STING pathway in 5 × FAD mice (Xie et al. 2023). Interestingly, IFN-β therapy led to a reduction in IFI16 and STING expression levels in a STING-dependent manner in these patients perhaps due to a negative feedback mechanism (Helbi et al. 2020). In summary, STING and its associated signaling pathways are altered in human neurodegenerative diseases, suggesting that targeting STING has great therapeutic potential and deserves further clinical and experimental research.

Mitochondrial Dysfunction

Mitochondrial dysfunction is a hallmark of degenerative diseases. Impaired DNA repair and mitochondrial oxidative stress lead to mtDNA damage and reduced mitochondrial membrane potential during neurodegeneration. Subsequently, more mtDNA to the cytoplasm results in activating the cGAS/STING/IRF3 pathway and pathological inflammatory response, which leads to synaptic loss and neurodegeneration. Exogenous melatonin has been shown to inhibit these events (Jauhari et al. 2020). Melatonin has also been found to ameliorate myopathic changes induced by activating the ALDH2-cGAS-STING-TBK1 pathway and promoting mitophagy (Wang et al. 2020). METTL14 suppresses ferroptosis and mitochondrial dysfunction in dopaminergic neurons by regulating the m6A modification of TRAF6 through the cGAS-STING Pathway (Shao et al. 2024). In contrast, metformin restores mitochondrial function to delay astrocyte senescence by Mfn2-dependent cGAS-STING (Wang et al. 2024b).

Loss of STING activity in parkin mutant flies have been shown to suppress muscle defects and ameliorate disrupted mitochondrial morphology (Moehlman et al. 2023), while parkin and Pink1 mitigate STING-induced inflammation (Sliter et al. 2018). Lee et al. reported that loss of STING or Relish was insufficient to generate the above effects (Lee et al. 2020). therefore, in the light of current evidence, it remains unknow whether STING contributes to behavioral or mitochondrial phenotypes in parkin or Pink1 mutants.

Inflammation

Neuroinflammation is thought to accompany and accelerate degenerative processes. cGAS-STING signaling has been discovered to promote neuroinflammation and neurodegeneration in various diseases, for example, Alzheimer’s disease (AD), Huntington’s disease, and amyotrophic lateral sclerosis (ALS) (Sharma et al. 2020; Tan et al. 2022; Gulen et al. 2023; Nelson and Xu 2023). In Parkinson’s disease (PD), STING activation not only induces neuroinflammation and degeneration of dopaminergic neurons via activation of downstream IFN-I, interferon regulatory factor 7, or inflammasome but also induces α-synuclein pathology (Szego et al. 2022; Zhou et al. 2025). Microglial DNase II deficiency initiates neuroinflammation, synapse loss and increased Abeta levels and tauopathy by activating the cGAS-STING-IFN-I pathway, (Li et al. 2025). Both microglial cGAS deficiency and STING inhibition not only can effectively alleviate MPTP-induced neuroinflammation, dopaminergic neurotoxicity, and neurodegeneration in PD (Ma et al. 2023; Wang et al. 2023) but also can alleviate microglial dysfunction and preserve intercellular communication in AD (Chung et al. 2024; He et al. 2025).

STING is also regulated by upstream signaling molecules in neurodegenerative diseases. For example, C9ORF72 inhibits STING- and JAK-STAT-mediated inflammation in ALS and frontotemporal lobar degeneration (McCauley et al. 2020; Pang and Hu 2023). Conversely, Tau activates microglia to facilitate brain inflammation through the PQBP1-cGAS-STING pathway (Jin et al. 2021). Moreover, SGK1, 1-BP exposure or high-fat diet not only exerts pro-inflammatory effects via the cGAS-STING-, NLRP3- and NF-κB-mediated inflammatory pathways but also exacerbates the degeneration and necroptosis of midbrain dopaminergic neurons (Kwon et al. 2021; Song et al. 2024). IL-6 and impaired proteasomes aggravate neuroinflammation and Semen Strychni pulveratum and vomicine alleviate neuroinflammation both by regulating STING signaling (Dwamena et al. 2024; Liu et al. 2024a; Zhan et al. 2025). Taken together, the above studies manifest that regulating STING signaling to improve neuroinflammation has significant potential in the treatment of neurodegenerative diseases.

Ataxia-Telangiectasia Mutated (ATM)

Ataxia-telangiectasia (A-T), a rare recessively inherited disorder that leads to progressive neurological decline, is caused by biallelic mutations in the ATM gene, which encodes a PI3K-like kinase. ATM kinase is not only involved in regulating the response to DNA double-strand breakage but is also a potential target for radiosensitization (Chiu et al. 2023). DNA damage that is not properly repaired results in increased cytosolic DNA and subsequent cGAS-STING pathway activation, which in turn exacerbates the inflammatory microenvironment (Quek et al. 2017). ATM deficiency leads to cytoplasmic DNA accumulation, STING activation and microglia overactivation (Song et al. 2019), while inhibition of ATM has been proved to activate the STING signaling, thereby enhancing the effects of anti-tumor and cancer treatments (Hu et al. 2021; Gao et al. 2023b).

In A-T fibroblasts, it displays elevated cGAS-STING signaling, and IFN-stimulated gene expression, but normal levels of IFN gene expression (Haj et al. 2023). The rapid senescence of ATM-deficient lung fibroblasts is driven by the synergistic effect of the cGAS-STING, p53, and p38 MAPK pathways to cope with persistent DNA damage (Haj et al. 2025). Suppression of the cGAS-STING signaling can mitigate neuropathology and premature senescence in A-T brain organoids (Aguado et al. 2021). Together, these studies indicate that the cGAS-STING signaling pathway provides a new therapeutic target for managing inflammation, premature senescence, and other related symptoms in A-T patients.

Others

Glial cells have a significant impact on neurodegeneration. For example, inducing IFN-I by STING during chronic neurodegeneration influences the microglial phenotype and exacerbates disease progression (Nazmi et al. 2019), Furthermore, deletion of microglial cGAS markedly suppresses plaque formation and prevents amyloid-β-induced cognitive impairment (He et al. 2023a), and STING inhibition can normalize the Abeta phagocytic capacity of microglia and alleviate cellular senescence (Yuan et al. 2025). Increased STING expression has been reported in the striatal astrocytes of multiple system atrophy patients (Inoue et al. 2021). Mechanistically, deletion of astrocytic cGAS has been shown to ameliorate astrocyte senescence and neurodegeneration (Jiang et al. 2023). Chemerin/chemokine-like receptor 1/STING pathway is involved in astrocyte recruitment to Abeta plaques (Liu et al. 2024b).

Increased mtDNA in the cytosol and defects in the degradation of activated STING have been associated with PD pathogenesis (Hancock-Cerutti et al. 2022). In addition, the mtDNA-STING-NLRP3/IL-1β axis has been shown to initiate neutrophil infiltration into the cerebra and induce impaired cognition in AD (Xia et al. 2023). Withaferin A has been proved to exert neuroprotective effects by activating DJ1 and Nrf2, and inhibiting STING in PD (Zhao et al. 2021). Strikingly, activation of the cGAMP-STING-IRF3 pathway induces the expression of triggering receptor expressed on myeloid cells 2 (TREM2), which regulates microglial polarization and suppresses AD (Xu et al. 2019). TOLLIP is a STING stabilizing agent that can prevent the degradation of STING through direct interactions (Pokatayev et al. 2020).

Drug Therapy

To date, several drugs have been shown to play a role in treating neurodegenerative diseases through STING signaling. Their common mechanism of action is to regulate the inflammatory response. Supplementation with the nicotinamide adenine dinucleotide (NAD+) precursor nicotinamide riboside was found to normalize elevated STING levels resulting in decreased pro-inflammatory cytokines, DNA damage, and cellular aging in AD (Hou et al. 2021). Analogously, tetrahydroxy stilbene glycoside, the main active ingredient of the Chinese herb Polygonum multiflorum, was shown to reduce M1 type microglia and pro-inflammatory cytokine in AD via inhibition of the cGAS-STING signaling pathway (Gao et al. 2023a). Consistent with the STING inhibitor H-151, dimethyl fumarate can downregulate inflammatory cytokines, granzyme, and chemokines by NF-κB and cGAS-STING to target inflammation and autoimmunity in sporadic ALS (Zamiri et al. 2023). The neuroprotective effects of silibinin include ameliorating STING-mediated neuroinflammation through attenuating ferroptosis damage in sporadic AD and STING-IRF3-IFN-β pathway in PD (Liu et al. 2021, 2023b). These findings indicate that targeting STING signaling may be beneficial in the development of novel treatments for neurodegenerative diseases.

STING in Intracranial Infection

Herpes Simplex Virus Type 1 Infection

IFN-I can protect the brain from viral infections, and the critical regulatory factor DDOST is indispensable for producing anti-viral IFN-I mediated by STING (Tu et al. 2022). Following herpes simplex virus type 1 (HSV-1) infection, IFN-I is mainly generated by microglia in a cGAS-STING-dependent manner. Furthermore, cGAS- or STING-deficient mice display increased viral loads and are susceptible to herpes simplex encephalitis (HSE) (Reinert et al. 2016). The STING agonist DMXAA has been shown to reduce viral replication by increasing IFN-I production (Ceron et al. 2019). HSV-1 infection of microglia induced IFN-I expression at low-to-medium viral loads, but cGAS/STING-dependent apoptosis at high viral loads (Reinert et al. 2021). While IFN-I plays a central role in HSV-1 antiviral responses, emerging evidence highlights IFN-independent mechanisms of STING in viral clearance. Notably, STING activation can directly induce autophagy-mediated degradation of viral particles by recruiting autophagy-related proteins (e.g., LC3) independent of IFN signaling (Gui et al. 2019). This pathway complements IFN-I-mediated immunity and may explain residual antiviral activity observed in IFNAR-deficient models (Gui et al. 2019). These dual roles (IFN-dependent and IFN-independent) collectively position STING as a multifaceted hub for antiviral defense against neurotropic viruses like HSV-1.

STING is crucial for control of HSV-1 after central infection, but dispensable for survival after peripheral infection (Parker et al. 2015). MAM domain containing 2 (MAMDC2) interacts with STING via its first MAM domain and increases the polymerization of STING, subsequently activating downstream TBK1-IRF3 signaling to enhance the IFN-I-based innate anti-viral response (Wang et al. 2022b). In contrast, TRIM18 recruits protein phosphatase 1A to dephosphorylate and inactivate TBK1, thereby preventing TBK1 from interacting with upstream adaptors and STING, and ultimately leading to the suppression of IFN-I mediated anti-viral effects during viral infections (Fang et al. 2022). HSV-1 VP1-2 results in deubiquitination of STING, which inhibits downstream IRF3 activation and IFN-I expression, thus facilitating brain infection and potential HSE progression (Bodda et al. 2020). In addition, resveratrol, an antitoxin produced by many plants when stimulated, ameliorates HSV-1-induced neuroinflammation, microglial M1 polarization, and viral encephalitis by STING/NF-κB pathway (Huang et al. 2024).

Other Infections

Infection with the Japanese encephalitis virus leads to the upregulation of various viral sensors, IFN regulatory factors, and STING. STING then induces anti-viral immune responses mediated through the activation of IFN-I. Moreover, STING expression affects the intracellular viral load (Nazmi et al. 2012; Sharma et al. 2021). However, in Rasmussen’s encephalitis, STING, IFN-β, and IFI16 are produced at relatively lower levels, and anti-viral innate immunity is insufficiently activated (Wang et al. 2022c). Microbial activation of STING signaling requires TRIF, which interacts directly with STING to enhance STING dimerization and intermembrane translocation. Thus, STING agonists are unable to induce genes in the absence of TRIF (Wang et al. 2016).

Following West Nile virus encephalitis, ELF4 combines with STING to induce IFN-I production and anti-viral immunity (You et al. 2013). Consistent with these reports, knockout of STING in bone marrow-derived macrophages leads to reduced IFN-I expression levels, which alters the CD4+/CD8+ T cell ratio and increases viral susceptibility in the CNS to causing CNS diseases (McGuckin Wuertz et al. 2019). Furthermore, Zika virus infection has been shown to activate NF-κB signaling and downstream dSTING expression to alleviate brain infections in Drosophila (Liu et al. 2018). In general, these studies expound that targeting the STING signaling pathway and STING-mediated IFN-I response is a beneficial therapeutic strategy to attenuate intracranial viral infection.

STING in Traumatic Brain Injury

IFN-I is associated with the occurrence and development of traumatic brain injury (TBI) neuropathology, and its canonical activation by cGAS-STING signaling is a key component of neuroinflammation, cognitive impairment, neural tissue damage, and autophagy dysfunction (Abdullah et al. 2018; Barrett et al. 2021; Fritsch et al. 2022; Fryer et al. 2024; Packer et al. 2024). STING signaling in peripheral immune cells and microglia drives an early inflammatory response, but CD300LF+ microglia can reduce neuroinflammation by STING inhibition (Fritsch et al. 2024; Lu et al. 2024). Following TBI, aberrant ER stress activates neuronal STING signaling and the release of excess IFN-I, which leads to white matter injury due to T cell infiltration and microglial activation (Sen et al. 2020). Consistent with previous studies, STING agonist DMXAA has been shown to intensify IFN-I-associated inflammation and gliosis (Wangler et al. 2022), while intranasal administration of atomic-let-7i was found to effectively suppress its downstream target STING to ameliorate cognitive function (He et al. 2022).

STING signaling also affects programmed cell death following TBI. Inhibiting the formation of NETs not only attenuates neuroinflammation, neurological deficits, and neuronal apoptosis through the STING-dependent IRE1α/ASK1/JNK pathway (Shi et al. 2023) but also mitigates NLRP1-associated neuronal pyroptosis through the STING/IRE1a pathway (Cao et al. 2023). Moreover, STING also activates NLRP3-mediated pyroptosis and augments pyroptosis-related neuroinflammation (Zhang et al. 2022). Ferroptosis is a novel type of iron-dependent programmed cell death. Moderately intensive treadmill exercise has been proved to ameliorate ferroptosis, cognitive deficits, and neurodegeneration through inhibition of the STING pathway (Chen et al. 2023). Strikingly, gender differences have been reported in cases of repeated mild TBI. Female mice were reported to display higher DNA damage, and lower expression of the cGAS-STING signaling and senescence protein p21 than males (Schwab et al. 2022). In conclusion, further research should focus on the specific mechanisms underlying STING signaling to improve and treat TBI.

Discussion and Conclusion

In recent years, STING signaling has received increasing attention. As a member of the cGAS/DncV-like nucleotidyltransferase family, cGAS detects abnormal DNA signals in the cytoplasm and catalyzes the synthesis of cyclic dinucleotide cGAMP to activate the downstream adapter protein STING (Kranzusch 2019). This is the canonical pathway for STING activation. However, STING can also be activated in a cGAS- and cGAMP-independent manner, although less is known about this signaling pathway. STING is an ER transmembrane protein, and its translocation to the Golgi apparatus is a hallmark of its activation. Thus, further research is required to elucidate the specific mechanisms mediating the translocation of STING from the ER to the Golgi apparatus, as well as to understand how STING is effectively sorted into lysosomes for degradation.

Targeting STING signaling pathways is a growing field of research. In particular, STING agonists and inhibitors (Tables 1, 2) have been the main focus of studies due to the regulatory role of STING in many diseases. STING agonists include native cyclic dinucleotides, cyclic dinucleotides derivatives, and non-nucleotide agonists (Wu et al. 2020) while STING inhibitors include nitrofurans, indole derivatives, oxo-tetrahydro-isoquinoline carboxylic acids, multisubstituted benzamides or benzenesulfonamides, cyclopeptides, dimeric benzimidazoles, heterobicyclic derivatives, miscellaneous compounds and STING degraders (Shen et al. 2022) (Fig. 3). Due to the presence of the blood–brain barrier, 98% of small-molecule drugs and almost all large-molecule drugs are restricted from entering the brain (Li et al. 2021), which limits the application of STING agonists and inhibitors in CNS diseases. Nanoparticle-based drug delivery is one of the most effective and promising ways of delivering drugs into inaccessible regions such as the brain since it protects the therapeutic drugs while simultaneously delivering them effectively to impaired areas (Saraiva et al. 2016). Previous studies have confirmed the efficacy of nanoparticles in crossing the BBB (Dong 2018). Indeed, nanoparticles loaded with STING modulators have been shown to display therapeutic effects on various diseases including cancers, SARS-CoV-2 infections, and Middle East respiratory syndrome coronavirus (Lin et al. 2019; Dosta et al. 2023; Zhang et al. 2023c). In the past few decades, researchers have explored many specific pathways leading to central nervous system diseases and developed targeted drugs and molecular formulations. Nevertheless, its clinical treatment progress is slow, and one of the main challenges is the obstruction of the BBB. Due to the presence of the BBB, many drugs cannot reach the brain or reach the brain at a very low concentration that cannot achieve effective therapeutic concentrations, resulting in limited clinical use of these drugs in the treatment of CNS diseases. Nanoparticles can assist drugs in effectively crossing the blood–brain barrier and targeting delivery into the brain. In theory, it is a promising idea that drug-loaded nanoparticles are encapsulated with cell membranes such as neutrophil and blood cell membranes, which not only can improve their targeting and precise delivery to the lesion site but also can escape immune defense to prevent clearance by the body. Although nanoparticles are an increasingly important research field in the treatment of CNS diseases, there are limited reports on the clinical application of nanoparticles in CNS diseases. Thus, further studies are necessary to evaluate their side effects and toxicity concerning CNS diseases.

Table 1.

The cGAS regulators in CNS diseases

cGAS modulators Regulatory effects Disease types Targeted delivery Effects References
RU.521 Inhibitor Cerebral venous sinus thrombosis None Beneficial Ding et al. (2022)
Cerebral ischemia–reperfusion injury None Beneficial Li et al. (2023)
Ischemic stroke None Beneficial Liu et al. (2023a); Zhao et al. (2024)
Neonatal hypoxic-ischemic encephalopathy None Beneficial Gamdzyk et al. (2020)
Huntington’s disease None Beneficial Jauhari et al. (2020)
Amyotrophic lateral sclerosis None Beneficial Yu et al. (2020); Tan et al. (2022)
Parkinson’s disease None Beneficial Ma et al. (2023); Zhou et al. (2025)
Alzheimer’s disease None Beneficial Gao et al. (2023a)
PF-06928215 Inhibitor Alzheimer’s disease None Detrimental Wang et al. (2020)

Table 2.

The STING regulators in CNS diseases

STING modulators Regulatory effects Disease types Targeted delivery Effects References
Brain tumors c-di-GMP Agonist Gliomas None Beneficial Ohkuri et al. (2014)
Agonist Glioblastoma multiforme Nanoparticles Beneficial Bielecki et al. (2021)
Agonist Glioblastoma nanocapsule Beneficial Zhou et al. (2022)
IACS8779 Agonist Glioblastoma None Beneficial Boudreau et al. (2021)
8803 Agonist Glioblastoma None Beneficial Najem et al. (2024)
DMXAA Agonist Neuroblastoma None Beneficial Layer et al. (2017)
SR717 Agonist Glioma Nanoparticles Beneficial Wang et al. (2022a)
2′3′-cGAMP Agonist Neuroblastoma Nanoparticles Beneficial Wang-Bishop et al. (2020)
CMA Agonist T cell-derived malignancies None Uncertain Gulen et al. (2017)
Stroke C-176 Inhibitor Ischemic stroke None Beneficial Peng et al. (2020); Kong et al. (2022)
CMA Agonist Subarachnoid hemorrhage None Detrimental Peng et al. (2020)
Agonist Ischemic stroke None Beneficial Kundu et al. (2022)
H151 Inhibitor Ischemic stroke None Beneficial Liu et al. (2024c); Wu et al. (2024)
2′3′-cGAMP Agonist Cerebral venous sinus thrombosis None Detrimental Ding et al. (2022)
Agonist ischemic stroke None Detrimental Liu et al. (2023a)
Agonist Neonatal hypoxic-ischemic encephalopathy None Detrimental Gamdzyk et al. (2020)
DMXAA Agonist Ischaemic stroke None Beneficial Kundu et al. (2022)
Degenerative diseases c-di-AM(PS)2 Agonist Alzheimer’s disease None Beneficial Wang et al. (2020)
Astin C Inhibitor Alzheimer’s disease None Detrimental Wang et al. (2020)
C-176 Inhibitor Parkinson’s disease None Beneficial Wang et al. (2023)
H-151 Inhibitor Amyotrophic lateral sclerosis None Beneficial Yu et al. (2020); Tan et al. (2022)
Inhibitor Ataxia- telangiectasia None Beneficial Aguado et al. (2021)
Inhibitor Alzheimer’s disease None Beneficial Hou et al. (2021); Gao et al. (2023a)
Inhibitor Sporadic amyotrophic lateral sclerosis None Beneficial Zamiri et al. (2023)
Inhibitor Parkinson’s disease None Beneficial Hinkle et al. (2022); Zhou et al. (2025)
DMXAA Agonist Parkinson’s disease None Detrimental Zhao et al. (2021); Hinkle et al. (2022)
palmitic acid Agonist Human Neurodegenerative Diseases None Detrimental Ferecskó et al. (2023)
Intracranial infection DMXAA Agonist Herpes simplex encephalitis None Beneficial Ceron et al. (2019)
2′3′-cGAMP Agonist Herpes simplex encephalitis None Beneficial Reinert et al. (2021)
2′3′-c-di-AM (PS)2(Rp, Rp) Agonist Herpes simplex encephalitis None Beneficial Wang et al. (2022b)
c-di-GMP Agonist Microbial infection None TRIF required Wang et al. (2016)
Traumatic brain injury DMXAA Agonist Traumatic brain injury None Detrimental Wangler et al. (2022)
2′3′-cGAMP Agonist Traumatic brain injury None Detrimental Shi et al. (2023)
cGAMP Agonist Traumatic brain injury None Detrimental Cao et al. (2023)
ADU- S100 Agonist Traumatic brain injury None Detrimental Zhang et al. (2022)
C-176 Inhibitor Traumatic brain injury None Beneficial Zhang et al. (2022); Cao et al. (2023); Shi et al. (2023); Fryer et al. (2024)

Fig. 3.

Fig. 3

The chemical structures of cGAS and STING mediators applied in CNS diseases

Multiple alleles have been reported in the coding gene of STING (TMEM173), while noticeable diversity has been detected in TMEM173 genotypes and STING protein expression among different ethnic groups (Fryer et al. 2021). Furthermore, significant differences between mouse and human STING mean that STING agonists exhibiting significant pharmacological effects in mice may not necessarily be validated in humans (Shih et al. 2018). For example, DMXAA (ASA404), one of the earliest reported non-cyclic dinucleotide small molecule STING agonists with preclinical efficacy, binds selectively to mouse STING rather than human STING (Cherney et al. 2022). Similar effects have also been reported in brain tumors (Bähr et al. 2017). These reports highlight the challenges associated with interspecies differences in STING and indicate that future studies should focus on human STING. Clinical trials on STING agonists and inhibitors should receive more attention. In conclusion, although the study of STING signaling remains a challenging area of research, there is enormous potential and significant prospects in disease treatments by targeting the STING signaling pathway.

Author Contributions

MS, JR, and ZZ wrote the original draft; ZY, CW and LL searched and organized the literature; JT and GM drew the figures; GM and MC reviewed and edited the manuscript and figures.

Funding

This work was supported by the National Nature Science Foundation of China (Grant Number NO. 82160243), Foundation of Jiangxi health and Family Planning Commission (Grant Number No. 220210596 and No.202210572) and the Natural Science Foundation of Jiangxi Provincial Department of Science and Technology (Grant Number 20242BAB25476).

Data Availability

No datasets were generated or analysed during the current study.

Declarations

Competing interests

The authors declare that they have no competing interests.

Footnotes

Publisher's Note

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

Min Song, Jianxun Ren and Zhipeng Zhu have contributed equally as co-first authors.

Contributor Information

Guohua Mao, Email: Maoguohua1967@sina.com.

Min Chen, Email: ndefy18261@ncu.edu.cn.

References

  1. Abdullah A, Zhang M, Frugier T, Bedoui S, Taylor JM, Crack PJ (2018) STING-mediated type-I interferons contribute to the neuroinflammatory process and detrimental effects following traumatic brain injury. J Neuroinflammation 15(1):323 [DOI] [PMC free article] [PubMed] [Google Scholar]
  2. Abe T, Barber GN (2014) Cytosolic-DNA-mediated, STING-dependent proinflammatory gene induction necessitates canonical NF-kappaB activation through TBK1. J Virol 88(10):5328–5341 [DOI] [PMC free article] [PubMed] [Google Scholar]
  3. Aguado J, Chaggar HK, Gomez-Inclan C, Shaker MR, Leeson HC, Mackay-Sim A, Wolvetang EJ (2021) Inhibition of the cGAS-STING pathway ameliorates the premature senescence hallmarks of Ataxia-Telangiectasia brain organoids. Aging Cell 20(9):e13468 [DOI] [PMC free article] [PubMed] [Google Scholar]
  4. Bähr O, Gross S, Harter PN, Kirches E, Mawrin C, Steinbach JP, Mittelbronn M (2017) ASA404, a vascular disrupting agent, as an experimental treatment approach for brain tumors. Oncol Lett 14(5):5443–5451 [DOI] [PMC free article] [PubMed] [Google Scholar]
  5. Bakhoum SF, Ngo B, Laughney AM, Cavallo JA, Murphy CJ, Ly P, Shah P, Sriram RK, Watkins TBK, Taunk NK, Duran M, Pauli C, Shaw C, Chadalavada K, Rajasekhar VK, Genovese G, Venkatesan S, Birkbak NJ, McGranahan N, Lundquist M, LaPlant Q, Healey JH, Elemento O, Chung CH, Lee NY, Imielenski M, Nanjangud G, Pe’er D, Cleveland DW, Powell SN, Lammerding J, Swanton C, Cantley LC (2018) Chromosomal instability drives metastasis through a cytosolic DNA response. Nature 553(7689):467–472 [DOI] [PMC free article] [PubMed] [Google Scholar]
  6. Bao P, Gu HY, Ye JJ, He JL, Zhong Z, Yu AX, Zhang XZ (2024) Chimeric exosomes functionalized with STING activation for personalized glioblastoma immunotherapy. Adv Sci (Weinh) 11(6):e2306336 [DOI] [PMC free article] [PubMed] [Google Scholar]
  7. Barrett JP, Knoblach SM, Bhattacharya S, Gordish-Dressman H, Stoica BA, Loane DJ (2021) Traumatic brain injury induces cGAS activation and type I interferon signaling in aged mice. Front Immunol 12:710608 [DOI] [PMC free article] [PubMed] [Google Scholar]
  8. Bielecki PA, Lorkowski ME, Becicka WM, Atukorale PU, Moon TJ, Zhang Y, Wiese M, Covarrubias G, Ravichandran S, Karathanasis E (2021) Immunostimulatory silica nanoparticle boosts innate immunity in brain tumors. Nanoscale Horiz 6(2):156–167 [DOI] [PMC free article] [PubMed] [Google Scholar]
  9. Bodda C, Reinert LS, Fruhwurth S, Richardo T, Sun C, Zhang BC, Kalamvoki M, Pohlmann A, Mogensen TH, Bergstrom P, Agholme L, O’Hare P, Sodeik B, Gyrd-Hansen M, Zetterberg H, Paludan SR (2020) HSV1 VP1–2 deubiquitinates STING to block type I interferon expression and promote brain infection. J Exp Med. 10.1084/jem.20191422 [DOI] [PMC free article] [PubMed] [Google Scholar]
  10. Boudreau CE, Najem H, Ott M, Horbinski C, Fang D, DeRay CM, Levine JM, Curran MA, Heimberger AB (2021) Intratumoral delivery of STING agonist results in clinical responses in canine glioblastoma. Clin Cancer Res 27(20):5528–5535 [DOI] [PMC free article] [PubMed] [Google Scholar]
  11. Cao Y, Shi M, Liu L, Zuo Y, Jia H, Min X, Liu X, Chen Z, Zhou Y, Li S, Yang G, Liu X, Deng Q, Chen F, Chen X, Zhang S, Zhang J (2023) Inhibition of neutrophil extracellular trap formation attenuates NLRP1-dependent neuronal pyroptosis via STING/IRE1alpha pathway after traumatic brain injury in mice. Front Immunol 14:1125759 [DOI] [PMC free article] [PubMed] [Google Scholar]
  12. Ceron S, North BJ, Taylor SA, Leib DA (2019) The STING agonist 5,6-dimethylxanthenone-4-acetic acid (DMXAA) stimulates an antiviral state and protects mice against herpes simplex virus-induced neurological disease. Virology 529:23–28 [DOI] [PMC free article] [PubMed] [Google Scholar]
  13. Chang H, Li Z, Zhang W, Lin C, Shen Y, Zhang G, Mao L, Ma C, Liu N, Lu H (2024) Transfer of cGAMP from neuron to microglia activates microglial type I interferon responses after subarachnoid hemorrhage. Cell Commun Signal 22(1):3 [DOI] [PMC free article] [PubMed] [Google Scholar]
  14. Chellen T, Bausart M, Maus P, Vanvarenberg K, Limaye N, Preat V, Malfanti A (2024) In situ administration of STING-activating hyaluronic acid conjugate primes anti-glioblastoma immune response. Mater Today Bio 26:101057 [DOI] [PMC free article] [PubMed] [Google Scholar]
  15. Chen Q, Boire A, Jin X, Valiente M, Er EE, Lopez-Soto A, Jacob L, Patwa R, Shah H, Xu K, Cross JR, Massagué J (2016) Carcinoma-astrocyte gap junctions promote brain metastasis by cGAMP transfer. Nature 533(7604):493–498 [DOI] [PMC free article] [PubMed] [Google Scholar]
  16. Chen Q, Boire A, Jin X, Valiente M, Emrah Er E, Lopez-Soto A, Jacob LS, Patwa R, Shah H, Xu K, Cross JR, Massagué J (2017) Corrigendum: carcinoma-astrocyte gap junctions promote brain metastasis by cGAMP transfer. Nature 544(7648):124 [DOI] [PubMed] [Google Scholar]
  17. Chen X, Yu M, Xu W, Kun P, Wan W, Yuhong X, Ye J, Liu Y, Luo J (2022) PCBP2 reduced oxidative stress-induced apoptosis in glioma through cGAS/STING pathway by METTL3-mediated m6A modification. Oxid Med Cell Longev 2022:9049571 [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
  18. Chen J, Zhu T, Yu D, Yan B, Zhang Y, Jin J, Yang Z, Zhang B, Hao X, Chen Z, Yan C, Yu J (2023) Moderate intensity of treadmill exercise rescues TBI-induced ferroptosis, neurodegeneration, and cognitive impairments via suppressing STING pathway. Mol Neurobiol. 10.1007/s12035-023-03379-8 [DOI] [PMC free article] [PubMed] [Google Scholar]
  19. Chen Y, Tian H, Zhang X, Nice EC, Huang C, Zhang H, Zheng S (2024) Copper-coordination driven brain-targeting nanoassembly for efficient glioblastoma multiforme immunotherapy by cuproptosis-mediated tumor immune microenvironment reprogramming. J Nanobiotechnol 22(1):801 [DOI] [PMC free article] [PubMed] [Google Scholar]
  20. Cherney EC, Zhang L, Lo J, Huynh T, Wei D, Ahuja V, Quesnelle C, Schieven GL, Futran A, Locke GA, Lin Z, Monereau L, Chaudhry C, Blum J, Li S, Fereshteh M, Li-Wang B, Gangwar S, Pan C, Chong C, Zhu X, Posy SL, Sack JS, Zhang P, Ruzanov M, Harner M, Akhtar F, Schroeder GM, Vite G, Fink B (2022) Discovery of non-nucleotide small-molecule STING agonists via chemotype hybridization. J Med Chem 65(4):3518–3538 [DOI] [PubMed] [Google Scholar]
  21. Chiu LY, Sun Q, Zenke FT, Blaukat A, Vassilev LT (2023) Selective ATM inhibition augments radiation-induced inflammatory signaling and cancer cell death. Aging (Albany NY) 15(2):492–512 [DOI] [PMC free article] [PubMed] [Google Scholar]
  22. Chu TT, Tu X, Yang K, Wu J, Repa JJ, Yan N (2021) Tonic prime-boost of STING signalling mediates Niemann-Pick disease type C. Nature 596(7873):570–575 [DOI] [PMC free article] [PubMed] [Google Scholar]
  23. Chung S, Jeong JH, Park JC, Han JW, Lee Y, Kim JI, Mook-Jung I (2024) Blockade of STING activation alleviates microglial dysfunction and a broad spectrum of Alzheimer’s disease pathologies. Exp Mol Med 56(9):1936–1951 [DOI] [PMC free article] [PubMed] [Google Scholar]
  24. Conlon J, Burdette DL, Sharma S, Bhat N, Thompson M, Jiang Z, Rathinam VA, Monks B, Jin T, Xiao TS, Vogel SN, Vance RE, Fitzgerald KA (2013) Mouse, but not human STING, binds and signals in response to the vascular disrupting agent 5,6-dimethylxanthenone-4-acetic acid. J Immunol 190(10):5216–5225 [DOI] [PMC free article] [PubMed]
  25. Costa Franco MM, Marim F, Guimarães ES, Assis NRG, Cerqueira DM, Alves-Silva J, Harms J, Splitter G, Smith J, Kanneganti TD, de Queiroz N, Gutman D, Barber GN, Oliveira SC (2018) Brucella abortus triggers a cGAS-independent STING pathway to induce host protection that involves guanylate-binding proteins and inflammasome activation. J Immunol 200(2):607–622 [DOI] [PMC free article] [PubMed] [Google Scholar]
  26. Ding R, Li H, Liu Y, Ou W, Zhang X, Chai H, Huang X, Yang W, Wang Q (2022) Activating cGAS-STING axis contributes to neuroinflammation in CVST mouse model and induces inflammasome activation and microglia pyroptosis. J Neuroinflamm 19(1):137 [DOI] [PMC free article] [PubMed] [Google Scholar]
  27. Dogan E, Yildirim Z, Akalin T, Ozgiray E, Akinturk N, Aktan C, Solmaz AE, Biceroglu H, Caliskan KE, Ertan Y, Yurtseven T, Kosova B, Bozok V (2024) Investigating the effects of PTEN mutations on cGAS-STING pathway in glioblastoma tumours. J Neurooncol 166(2):283–292 [DOI] [PMC free article] [PubMed] [Google Scholar]
  28. Dong X (2018) Current strategies for brain drug delivery. Theranostics 8(6):1481–1493 [DOI] [PMC free article] [PubMed] [Google Scholar]
  29. Dosta P, Cryer AM, Dion MZ, Shiraishi T, Langston SP, Lok D, Wang J, Harrison S, Hatten T, Ganno ML, Appleman VA, Taboada GM, Puigmal N, Ferber S, Kalash S, Prado M, Rodriguez AL, Kamoun WS, Abu-Yousif AO, Artzi N (2023) Investigation of the enhanced antitumour potency of STING agonist after conjugation to polymer nanoparticles. Nat Nanotechnol 18(11):1351–1363 [DOI] [PubMed] [Google Scholar]
  30. Dunphy G, Flannery SM, Almine JF, Connolly DJ, Paulus C, Jonsson KL, Jakobsen MR, Nevels MM, Bowie AG, Unterholzner L (2018) Non-canonical activation of the DNA sensing adaptor STING by ATM and IFI16 mediates NF-kappaB signaling after nuclear DNA damage. Mol Cell 71(5):745-760 e745 [DOI] [PMC free article] [PubMed] [Google Scholar]
  31. Dwamena A, Asadi Y, Gilstrap E, Wang H (2024) Impaired proteasome induces mitochondrial DNA release to activate the cGAS-STING signaling pathway and cause necroptosis in mouse brain. bioRxiv
  32. Fang M, Zhang A, Du Y, Lu W, Wang J, Minze LJ, Cox TC, Li XC, Xing J, Zhang Z (2022) TRIM18 is a critical regulator of viral myocarditis and organ inflammation. J Biomed Sci 29(1):55 [DOI] [PMC free article] [PubMed] [Google Scholar]
  33. Fang R, Jiang Q, Jia X, Jiang Z (2023) ARMH3-mediated recruitment of PI4KB directs Golgi-to-endosome trafficking and activation of the antiviral effector STING. Immunity 56(3):500-515 e506 [DOI] [PubMed] [Google Scholar]
  34. Ferecskó AS, Smallwood MJ, Moore A, Liddle C, Newcombe J, Holley J, Whatmore J, Gutowski NJ, Eggleton P (2023) STING-triggered CNS inflammation in human neurodegenerative diseases. Biomedicines 11(5):1375 [DOI] [PMC free article] [PubMed] [Google Scholar]
  35. Fritsch LE, Ju J, Gudenschwager Basso EK, Soliman E, Paul S, Chen J, Kaloss AM, Kowalski EA, Tuhy TC, Somaiya RD, Wang X, Allen IC, Theus MH, Pickrell AM (2022) Type I interferon response is mediated by NLRX1-cGAS-STING signaling in brain injury. Front Mol Neurosci 15:852243 [DOI] [PMC free article] [PubMed] [Google Scholar]
  36. Fritsch LE, Kelly C, Leonard J, de Jager C, Wei X, Brindley S, Harris EA, Kaloss AM, DeFoor N, Paul S, O’Malley H, Ju J, Olsen ML, Theus MH, Pickrell AM (2024) STING-dependent signaling in microglia or peripheral immune cells orchestrates the early inflammatory response and influences brain injury outcome. J Neurosci 44(12):e0191232024 [DOI] [PMC free article] [PubMed] [Google Scholar]
  37. Fryer AL, Abdullah A, Taylor JM, Crack PJ (2021) The complexity of the cGAS-STING pathway in CNS pathologies. Front Neurosci 15:621501 [DOI] [PMC free article] [PubMed] [Google Scholar]
  38. Fryer AL, Abdullah A, Mobilio F, Jobling A, Moore Z, de Veer M, Zheng G, Wong BX, Taylor JM, Crack PJ (2024) Pharmacological inhibition of STING reduces neuroinflammation-mediated damage post-traumatic brain injury. Br J Pharmacol 181(17):3118–3135 [DOI] [PubMed] [Google Scholar]
  39. Galluzzi L, Vanpouille-Box C, Bakhoum SF, Demaria S (2018) SnapShot: CGAS-STING signaling. Cell 173(1):276-276 e271 [DOI] [PubMed] [Google Scholar]
  40. Gamdzyk M, Doycheva DM, Araujo C, Ocak U, Luo Y, Tang J, Zhang JH (2020) cGAS/STING pathway activation contributes to delayed neurodegeneration in neonatal hypoxia-ischemia rat model: possible involvement of LINE-1. Mol Neurobiol 57(6):2600–2619 [DOI] [PMC free article] [PubMed] [Google Scholar]
  41. Gao Z, Yang J (2024) GNB4 silencing promotes pyroptosis to inhibit the development of glioma by Activating cGAS-STING pathway. Mol Biotechnol. 10.1007/s12033-024-01194-7 [DOI] [PubMed] [Google Scholar]
  42. Gao D, Hao JP, Li BY, Zheng CC, Miao BB, Zhang L, Li YL, Li L, Li XJ, Zhang L (2023a) Tetrahydroxy stilbene glycoside ameliorates neuroinflammation for Alzheimer’s disease via cGAS-STING. Eur J Pharmacol 953:175809 [DOI] [PubMed] [Google Scholar]
  43. Gao Y, Li Y, Lin Z, Zeng Y, Huang Z, Han L, Zhong Y, Gong Y, Wu Q, Xie C (2023b) Ataxia telangiectasia mutated kinase inhibition promotes irradiation-induced PD-L1 expression in tumour-associated macrophages through IFN-I/JAK signalling pathway. Immunology 168(2):346–361 [DOI] [PubMed] [Google Scholar]
  44. Gonugunta VK, Sakai T, Pokatayev V, Yang K, Wu J, Dobbs N, Yan N (2017) Trafficking-mediated STING degradation requires sorting to acidified endolysosomes and can be targeted to enhance anti-tumor response. Cell Rep 21(11):3234–3242 [DOI] [PMC free article] [PubMed] [Google Scholar]
  45. Gui X, Yang H, Li T, Tan X, Shi P, Li M, Du F, Chen ZJ (2019) Autophagy induction via STING trafficking is a primordial function of the cGAS pathway. Nature 567(7747):262–266 [DOI] [PMC free article] [PubMed] [Google Scholar]
  46. Gul E, Sayar EH, Gungor B, Eroglu FK, Surucu N, Keles S, Guner SN, Findik S, Alpdundar E, Ayanoglu IC, Kayaoglu B, Geckin BN, Sanli HA, Kahraman T, Yakicier C, Muftuoglu M, Oguz B, Cagdas Ayvaz DN, Gursel I, Ozen S, Reisli I, Gursel M (2018) Type I IFN-related NETosis in ataxia telangiectasia and Artemis deficiency. J Allergy Clin Immunol 142(1):246–257 [DOI] [PubMed] [Google Scholar]
  47. Gulen MF, Koch U, Haag SM, Schuler F, Apetoh L, Villunger A, Radtke F, Ablasser A (2017) Signalling strength determines proapoptotic functions of STING. Nat Commun 8(1):427 [DOI] [PMC free article] [PubMed] [Google Scholar]
  48. Gulen MF, Samson N, Keller A, Schwabenland M, Liu C, Gluck S, Thacker VV, Favre L, Mangeat B, Kroese LJ, Krimpenfort P, Prinz M, Ablasser A (2023) cGAS-STING drives ageing-related inflammation and neurodegeneration. Nature 620(7973):374–380 [DOI] [PMC free article] [PubMed] [Google Scholar]
  49. Haase S, Banerjee K, Mujeeb AA, Hartlage CS, Nunez FM, Nunez FJ, Alghamri MS, Kadiyala P, Carney S, Barissi MN, Taher AW, Brumley EK, Thompson S, Dreyer JT, Alindogan CT, Garcia-Fabiani MB, Comba A, Venneti S, Ravikumar V, Koschmann C, Carcaboso AM, Vinci M, Rao A, Yu JS, Lowenstein PR, Castro MG (2022) H3.3–G34 mutations impair DNA repair and promote cGAS/STING-mediated immune responses in pediatric high-grade glioma models. J Clin Invest. 10.1172/JCI154229 [DOI] [PMC free article] [PubMed] [Google Scholar]
  50. Haj M, Levon A, Frey Y, Hourvitz N, Campisi J, Tzfati Y, Elkon R, Ziv Y, Shiloh Y (2023) Accelerated replicative senescence of ataxia-telangiectasia skin fibroblasts is retained at physiologic oxygen levels, with unique and common transcriptional patterns. Aging Cell 22(8):e13869 [DOI] [PMC free article] [PubMed] [Google Scholar]
  51. Haj M, Frey Y, Levon A, Maliah A, Ben-Yishay T, Slutsky R, Smoom R, Tzfati Y, Ben-David U, Levy C, Elkon R, Ziv Y, Shiloh Y (2025) The cGAS-STING, p38 MAPK, and p53 pathways link genome instability to accelerated cellular senescence in ATM-deficient murine lung fibroblasts. Proc Natl Acad Sci USA 122(2):e2419196122 [DOI] [PMC free article] [PubMed] [Google Scholar]
  52. Hancock-Cerutti W, Wu Z, Xu P, Yadavalli N, Leonzino M, Tharkeshwar AK, Ferguson SM, Shadel GS, De Camilli P (2022) ER-lysosome lipid transfer protein VPS13C/PARK23 prevents aberrant mtDNA-dependent STING signaling. J Cell Biol 221(7):e202106046 [DOI] [PMC free article] [PubMed] [Google Scholar]
  53. Hartlova A, Erttmann SF, Raffi FA, Schmalz AM, Resch U, Anugula S, Lienenklaus S, Nilsson LM, Kroger A, Nilsson JA, Ek T, Weiss S, Gekara NO (2015) DNA damage primes the type I interferon system via the cytosolic DNA sensor STING to promote anti-microbial innate immunity. Immunity 42(2):332–343 [DOI] [PubMed] [Google Scholar]
  54. He XC, Wang J, Du HZ, Liu CM, Teng ZQ (2022) Intranasal administration of Agomir-let-7i improves cognitive function in mice with traumatic brain injury. Cells 11(8):1348 [DOI] [PMC free article] [PubMed] [Google Scholar]
  55. He S, Li X, Mittra N, Bhattacharjee A, Wang H, Zhao S, Liu F, Han X (2023) Microglial cGAS deletion protects against amyloid-beta induced Alzheimer's disease pathogenesis. bioRxiv [DOI] [PMC free article] [PubMed]
  56. He Y, Yang Y, Huang W, Yang S, Xue X, Zhu K, Tan H, Sun T, Yang W (2023b) Manganese facilitated cGAS-STING-IFNI pathway activation induced by ionizing radiation in glioma cells. Int J Radiat Biol 99(12):1890–1907 [DOI] [PubMed] [Google Scholar]
  57. He W, Lv W, Liu L, Gong Y, Song K, Xu J, Zhao W, Li S, Min Z, Chen Q, Yin J, Chen Y, Fang H, Xin H, Fang X (2024) Enhanced antiglioma effect by a Vitamin D3-inserted lipid hybrid neutrophil membrane biomimetic multimodal nanoplatform. ACS Nano 18:35559–35574 [DOI] [PubMed] [Google Scholar]
  58. He S, Li X, Mittra N, Bhattacharjee A, Wang H, Song S, Zhao S, Liu F, Han X (2025) Microglial cGAS deletion preserves intercellular communication and alleviates amyloid-beta-induced pathogenesis of Alzheimer’s disease. Adv Sci (Weinh) 12:e2410910 [DOI] [PMC free article] [PubMed] [Google Scholar]
  59. Helbi S, Ravanbakhsh B, Karimi M, Kooti W, Jivad N (2020) Aligned expression of IFI16 and STING genes in RRMS patients’ blood. Endocr Metab Immune Disord Drug Targets 20(6):878–886 [DOI] [PubMed] [Google Scholar]
  60. Hinkle JT, Patel J, Panicker N, Karuppagounder SS, Biswas D, Belingon B, Chen R, Brahmachari S, Pletnikova O, Troncoso JC, Dawson VL, Dawson TM (2022) STING mediates neurodegeneration and neuroinflammation in nigrostriatal alpha-synucleinopathy. Proc Natl Acad Sci USA 119(15):e2118819119 [DOI] [PMC free article] [PubMed] [Google Scholar]
  61. Ho WS, Mondal I, Xu B, Das O, Sun R, Chiou P, Cai X, Tahmasebinia F, Wu CY, Wu Z, Matsui W, Lim M, Meng Z, Lu RO (2023) PP2Ac/STRN4 negatively regulates STING-type I IFN signaling in tumor-associated macrophages. J Clin Invest. 10.1172/JCI162139 [DOI] [PMC free article] [PubMed] [Google Scholar]
  62. Holm CK, Rahbek SH, Gad HH, Bak RO, Jakobsen MR, Jiang Z, Hansen AL, Jensen SK, Sun C, Thomsen MK, Laustsen A, Nielsen CG, Severinsen K, Xiong Y, Burdette DL, Hornung V, Lebbink RJ, Duch M, Fitzgerald KA, Bahrami S, Mikkelsen JG, Hartmann R, Paludan SR (2016) Influenza A virus targets a cGAS-independent STING pathway that controls enveloped RNA viruses. Nat Commun 7:10680 [DOI] [PMC free article] [PubMed] [Google Scholar]
  63. Hou Y, Liang H, Rao E, Zheng W, Huang X, Deng L, Zhang Y, Yu X, Xu M, Mauceri H, Arina A, Weichselbaum RR, Fu YX (2018) Non-canonical NF-kappaB antagonizes STING sensor-mediated DNA sensing in radiotherapy. Immunity 49(3):490-503 e494 [DOI] [PMC free article] [PubMed] [Google Scholar]
  64. Hou Y, Wei Y, Lautrup S, Yang B, Wang Y, Cordonnier S, Mattson MP, Croteau DL, Bohr VA (2021) NAD(+) supplementation reduces neuroinflammation and cell senescence in a transgenic mouse model of Alzheimer’s disease via cGAS-STING. Proc Natl Acad Sci USA. 10.1073/pnas.2011226118 [DOI] [PMC free article] [PubMed] [Google Scholar]
  65. Hu M, Zhou M, Bao X, Pan D, Jiao M, Liu X, Li F, Li CY (2021) ATM inhibition enhances cancer immunotherapy by promoting mtDNA leakage and cGAS/STING activation. J Clin Invest. 10.1172/JCI139333 [DOI] [PMC free article] [PubMed] [Google Scholar]
  66. Huang Z, Li S, Zhong L, Su Y, Li M, Wang X, Wang Z, Wang Z, Ye C, Ren Z, Wang X, Zeng Q, Zheng K, Wang Y (2024) Effect of resveratrol on herpesvirus encephalitis: evidences for its mechanisms of action. Phytomedicine 127:155476 [DOI] [PubMed] [Google Scholar]
  67. Huang FQ, Wang HF, Yang T, Yang D, Liu P, Alolga RN, Ma G, Liu B, Pan A, Liu SJ, Qi LW (2025) Ceramides increase mitochondrial permeabilization to trigger mtDNA-dependent inflammation in astrocytes during brain ischemia. Metabolism 156:156161 [DOI] [PubMed] [Google Scholar]
  68. Inoue Y, Ayaki T, Ishimoto T, Yamakado H, Maki T, Matsuzawa S, Sawamoto N, Takahashi R (2021) The stimulator of interferon genes (STING) pathway is upregulated in striatal astrocytes of patients with multiple system atrophy. Neurosci Lett 757:135972 [DOI] [PubMed] [Google Scholar]
  69. Jauhari A, Baranov SV, Suofu Y, Kim J, Singh T, Yablonska S, Li F, Wang X, Oberly P, Minnigh MB, Poloyac SM, Carlisle DL, Friedlander RM (2020) Melatonin inhibits cytosolic mitochondrial DNA-induced neuroinflammatory signaling in accelerated aging and neurodegeneration. J Clin Invest 130(6):3124–3136 [DOI] [PMC free article] [PubMed] [Google Scholar]
  70. Jiang GL, Yang XL, Zhou HJ, Long J, Liu B, Zhang LM, Lu D (2021) cGAS knockdown promotes microglial M2 polarization to alleviate neuroinflammation by inhibiting cGAS-STING signaling pathway in cerebral ischemic stroke. Brain Res Bull 171:183–195 [DOI] [PubMed] [Google Scholar]
  71. Jiang SY, Tian T, Yao H, Xia XM, Wang C, Cao L, Hu G, Du RH, Lu M (2023) The cGAS-STING-YY1 axis accelerates progression of neurodegeneration in a mouse model of Parkinson’s disease via LCN2-dependent astrocyte senescence. Cell Death Differ 30:2280–2292 [DOI] [PMC free article] [PubMed] [Google Scholar]
  72. Jiang GY, Yang HR, Li C, Liu N, Ma SJ, Jin BX, Yan C, Gong HD, Li JY, Yan HC, Ye GX, Wang WY, Gao C (2025) Ginsenoside Rd alleviates early brain injury by inhibiting ferroptosis through cGAS/STING/DHODH pathway after subarachnoid hemorrhage. Free Radic Biol Med 228:299–318 [DOI] [PubMed] [Google Scholar]
  73. Jin M, Shiwaku H, Tanaka H, Obita T, Ohuchi S, Yoshioka Y, Jin X, Kondo K, Fujita K, Homma H, Nakajima K, Mizuguchi M, Okazawa H (2021) Tau activates microglia via the PQBP1-cGAS-STING pathway to promote brain inflammation. Nat Commun 12(1):6565 [DOI] [PMC free article] [PubMed] [Google Scholar]
  74. Kang L, Yu H, Yang X, Zhu Y, Bai X, Wang R, Cao Y, Xu H, Luo H, Lu L, Shi MJ, Tian Y, Fan W, Zhao BQ (2020) Neutrophil extracellular traps released by neutrophils impair revascularization and vascular remodeling after stroke. Nat Commun 11(1):2488 [DOI] [PMC free article] [PubMed] [Google Scholar]
  75. Kong L, Li W, Chang E, Wang W, Shen N, Xu X, Wang X, Zhang Y, Sun W, Hu W, Xu P, Liu X (2022) mtDNA-STING Axis mediates microglial polarization via IRF3/NF-kappaB signaling after ischemic stroke. Front Immunol 13:860977 [DOI] [PMC free article] [PubMed] [Google Scholar]
  76. Kranzusch PJ (2019) cGAS and CD-NTase enzymes: structure, mechanism, and evolution. Curr Opin Struct Biol 59:178–187 [DOI] [PMC free article] [PubMed] [Google Scholar]
  77. Kundu N, Kumar A, Corona C, Chen Y, Seth S, Karuppagounder SS, Ratan RR (2022) A STING agonist preconditions against ischaemic stroke via an adaptive antiviral Type 1 interferon response. Brain Commun 4(3):fcac133 [DOI] [PMC free article] [PubMed] [Google Scholar]
  78. Kwon J, Bakhoum SF (2020) The cytosolic DNA-sensing cGAS-STING pathway in cancer. Cancer Discov 10(1):26–39 [DOI] [PMC free article] [PubMed] [Google Scholar]
  79. Kwon OC, Song JJ, Yang Y, Kim SH, Kim JY, Seok MJ, Hwang I, Yu JW, Karmacharya J, Maeng HJ, Kim J, Jho EH, Ko SY, Son H, Chang MY, Lee SH (2021) SGK1 inhibition in glia ameliorates pathologies and symptoms in Parkinson disease animal models. EMBO Mol Med 13(4):e13076 [DOI] [PMC free article] [PubMed] [Google Scholar]
  80. Layer JP, Kronmüller MT, Quast T, van den Boorn-Konijnenberg D, Effern M, Hinze D, Althoff K, Schramm A, Westermann F, Peifer M, Hartmann G, Tüting T, Kolanus W, Fischer M, Schulte J, Hölzel M (2017) Amplification of N-Myc is associated with a T-cell-poor microenvironment in metastatic neuroblastoma restraining interferon pathway activity and chemokine expression. Oncoimmunology 6(6):e1320626 [DOI] [PMC free article] [PubMed] [Google Scholar]
  81. Lee JJ, Andreazza S, Whitworth AJ (2020) The STING pathway does not contribute to behavioural or mitochondrial phenotypes in Drosophila Pink1/parkin or mtDNA mutator models. Sci Rep 10(1):2693 [DOI] [PMC free article] [PubMed] [Google Scholar]
  82. Li X, Vemireddy V, Cai Q, Xiong H, Kang P, Li X, Giannotta M, Hayenga HN, Pan E, Sirsi SR, Mateo C, Kleinfeld D, Greene C, Campbell M, Dejana E, Bachoo R, Qin Z (2021) Reversibly modulating the blood-brain barrier by laser stimulation of molecular-targeted nanoparticles. Nano Lett 21(22):9805–9815 [DOI] [PMC free article] [PubMed] [Google Scholar]
  83. Li S, Wernersbach I, Harms GS, Schafer MKE (2022) Microglia subtypes show substrate- and time-dependent phagocytosis preferences and phenotype plasticity. Front Immunol 13:945485 [DOI] [PMC free article] [PubMed] [Google Scholar]
  84. Li B, Wang W, Li Y, Wang S, Liu H, Xia Z, Gao W, Zhao B (2023) cGAS-STING pathway aggravates early cerebral ischemia-reperfusion injury in mice by activating NCOA4-mediated ferritinophagy. Exp Neurol 359:114269 [DOI] [PubMed] [Google Scholar]
  85. Li W, Shen N, Kong L, Huang H, Wang X, Zhang Y, Wang G, Xu P, Hu W (2023) STING mediates microglial pyroptosis via interaction with NLRP3 in cerebral ischaemic stroke. Stroke Vasc Neurol. 10.1136/svn-2023-002320 [DOI] [PMC free article] [PubMed] [Google Scholar]
  86. Li Q, Yang L, Wang K, Chen Z, Liu H, Yang X, Xu Y, Chen Y, Gong Z, Jia Y (2024a) Oxidized mitochondrial DNA activates the cGAS-STING pathway in the neuronal intrinsic immune system after brain ischemia-reperfusion injury. Neurotherapeutics 21(4):e00368 [DOI] [PMC free article] [PubMed] [Google Scholar]
  87. Li X, Ding H, Jing J, Qian S, Ma Y, Lv M, Gao Y, Zhang Y, Li T (2024b) Sulfasalazine improves neuronal function in mice with ischemic stroke by inhibiting the STING/NF-kappaB pathway. Naunyn Schmiedebergs Arch Pharmacol. 10.1007/s00210-024-03656-w [DOI] [PubMed] [Google Scholar]
  88. Li LJ, Liang SY, Sun XY, Zhu J, Niu XY, Du XY, Huang YR, Liu RT (2025) Microglial double stranded DNA accumulation induced by DNase II deficiency drives neuroinflammation and neurodegeneration. J Neuroinflamm 22(1):11 [DOI] [PMC free article] [PubMed] [Google Scholar]
  89. Liao Y, Cheng J, Kong X, Li S, Li X, Zhang M, Zhang H, Yang T, Dong Y, Li J, Xu Y, Yuan Z (2020) HDAC3 inhibition ameliorates ischemia/reperfusion-induced brain injury by regulating the microglial cGAS-STING pathway. Theranostics 10(21):9644–9662 [DOI] [PMC free article] [PubMed] [Google Scholar]
  90. Lin LC, Huang CY, Yao BY, Lin JC, Agrawal A, Algaissi A, Peng BH, Liu YH, Huang PH, Juang RH, Chang YC, Tseng CT, Chen HW, Hu CJ (2019) Viromimetic STING agonist-loaded hollow polymeric nanoparticles for safe and effective vaccination against middle east respiratory syndrome coronavirus. Adv Funct Mater 29(28):1807616 [DOI] [PMC free article] [PubMed] [Google Scholar]
  91. Lin F, Yao X, Kong C, Liu X, Zhao Z, Rao S, Wang L, Li S, Wang J, Dai Q (2021) 25-Hydroxycholesterol protecting from cerebral ischemia-reperfusion injury through the inhibition of STING activity. Aging (Albany NY) 13(16):20149–20163 [DOI] [PMC free article] [PubMed] [Google Scholar]
  92. Lin C, He C, Li L, Liu Y, Tang L, Ni Z, Zhang N, Lai T, Chen X, Wang X (2024) Alpha-lipoic acid (ALA) ameliorates early brain injury after subarachnoid hemorrhage in Sprague-Dawley (SD) rats via inhibiting STING-NLRP3 inflammatory signaling. NeuroReport 35(4):250–257 [DOI] [PMC free article] [PubMed] [Google Scholar]
  93. Liu S, Cai X, Wu J, Cong Q, Chen X, Li T, Du F, Ren J, Wu YT, Grishin NV, Chen ZJ (2015) Phosphorylation of innate immune adaptor proteins MAVS, STING, and TRIF induces IRF3 activation. Science 347(6227):aaa2630 [DOI] [PubMed] [Google Scholar]
  94. Liu Y, Gordesky-Gold B, Leney-Greene M, Weinbren NL, Tudor M, Cherry S (2018) Inflammation-induced, STING-dependent autophagy restricts zika virus infection in the drosophila brain. Cell Host Microbe 24(1):57–6853 [DOI] [PMC free article] [PubMed] [Google Scholar]
  95. Liu X, Chen W, Wang C, Liu W, Hayashi T, Mizuno K, Hattori S, Fujisaki H, Ikejima T (2021) Silibinin ameliorates depression/anxiety-like behaviors of Parkinson’s disease mouse model and is associated with attenuated STING-IRF3-IFN-beta pathway activation and neuroinflammation. Physiol Behav 241:113593 [DOI] [PubMed] [Google Scholar]
  96. Liu M, Li Y, Han S, Wang H, Li J (2023a) Activin A alleviates neuronal injury through inhibiting cGAS-STING-mediated autophagy in mice with ischemic stroke. J Cereb Blood Flow Metab 43(5):736–748 [DOI] [PMC free article] [PubMed] [Google Scholar]
  97. Liu P, Chen W, Kang Y, Wang C, Wang X, Liu W, Hayashi T, Qiu Z, Mizuno K, Hattori S, Fujisaki H, Ikejima T (2023b) Silibinin ameliorates STING-mediated neuroinflammation via downregulation of ferroptotic damage in a sporadic Alzheimer’s disease model. Arch Biochem Biophys 744:109691 [DOI] [PubMed] [Google Scholar]
  98. Liu M, Pan J, Li X, Zhang X, Tian F, Li M, Wu X, Zhang L, Qin C (2024a) Interleukin-6 deficiency reduces neuroinflammation by inhibiting the STAT3-cGAS-STING pathway in Alzheimer’s disease mice. J Neuroinflamm 21(1):282 [DOI] [PMC free article] [PubMed] [Google Scholar]
  99. Liu Z, Chen Y, Chen Y, Zheng J, Wu W, Wang L, Wang H, Yu Y (2024b) Effect of regulation of chemerin/chemokine-like receptor 1/stimulator of interferon genes pathway on astrocyte recruitment to abeta plaques. Int J Mol Sci 25(8):4324 [DOI] [PMC free article] [PubMed] [Google Scholar]
  100. Liu Z, Qin Q, Wang S, Kang X, Liu Y, Wei L, Lu Z, Cai W, Hu M (2024c) STING activation in macrophages and microglia drives poststroke inflammation: implications for neuroinflammatory mechanisms and therapeutic interventions. CNS Neurosci Ther 30(12):e70106 [DOI] [PMC free article] [PubMed] [Google Scholar]
  101. Lu Z, Liu Z, Wang C, Jiang R, Wang Z, Liao W, Wang W, Chen J, Zhu X, Zhao J, Liu Q, Yang Y, Gong P (2024) CD300LF(+) microglia impede the neuroinflammation following traumatic brain injury by inhibiting STING pathway. CNS Neurosci Ther 30(7):e14824 [DOI] [PMC free article] [PubMed] [Google Scholar]
  102. Lu X, Xia H, Gao W, Pan X, Yan Y, Zhang R, He Y, Wang Y, Chen B, Mei D (2025) A pH-responsive and guanidinium-rich nanoadjuvant efficiently delivers STING agonist for cancer immunotherapy. ACS Nano. 10.1021/acsnano.4c10202 [DOI] [PubMed] [Google Scholar]
  103. Ma C, Liu Y, Li S, Ma C, Huang J, Wen S, Yang S, Wang B (2023) Microglial cGAS drives neuroinflammation in the MPTP mouse models of Parkinson’s disease. CNS Neurosci Ther 29:2018–2035 [DOI] [PMC free article] [PubMed] [Google Scholar]
  104. Ma Y, Liu Z, Deng L, Du J, Fan Z, Ma T, Xiong J, Xiuyun X, Gu N, Di Z, Zhang Y (2024) FGF21 attenuates neuroinflammation following subarachnoid hemorrhage through promoting mitophagy and inhibiting the cGAS-STING pathway. J Transl Med 22(1):436 [DOI] [PMC free article] [PubMed] [Google Scholar]
  105. Maimaiti M, Li C, Cheng M, Zhong Z, Hu J, Yang L, Zhang L, Hong Z, Song J, Pan M, Ma X, Cui S, Zhang P, Hao H, Wang C, Hu H (2024) Blocking cGAS-STING pathway promotes post-stroke functional recovery in an extended treatment window via facilitating remyelination. Med 5(6):622–644628 [DOI] [PubMed] [Google Scholar]
  106. Masanneck L, Eichler S, Vogelsang A, Korsen M, Wiendl H, Budde T, Meuth SG (2020) The STING-IFN-beta-dependent axis is markedly low in patients with relapsing-remitting multiple sclerosis. Int J Mol Sci 21(23):9249 [DOI] [PMC free article] [PubMed] [Google Scholar]
  107. McCauley ME, O’Rourke JG, Yanez A, Markman JL, Ho R, Wang X, Chen S, Lall D, Jin M, Muhammad A, Bell S, Landeros J, Valencia V, Harms M, Arditi M, Jefferies C, Baloh RH (2020) C9orf72 in myeloid cells suppresses STING-induced inflammation. Nature 585(7823):96–101 [DOI] [PMC free article] [PubMed] [Google Scholar]
  108. McGuckin Wuertz K, Treuting PM, Hemann EA, Esser-Nobis K, Snyder AG, Graham JB, Daniels BP, Wilkins C, Snyder JM, Voss KM, Oberst A, Lund J, Gale M Jr (2019) STING is required for host defense against neuropathological West Nile virus infection. PLoS Pathog 15(8):e1007899 [DOI] [PMC free article] [PubMed] [Google Scholar]
  109. Meibers HE, Warrick KA, VonHandorf A, Vallez CN, Kawarizadeh K, Saha I, Donmez O, Jain VG, Kottyan LC, Weirauch MT, Pasare C (2023) Effector memory T cells induce innate inflammation by triggering DNA damage and a non-canonical STING pathway in dendritic cells. Cell Rep 42(10):113180 [DOI] [PMC free article] [PubMed] [Google Scholar]
  110. Moehlman AT, Kanfer G, Youle RJ (2023) Loss of STING in parkin mutant flies suppresses muscle defects and mitochondria damage. PLoS Genet 19(7):e1010828 [DOI] [PMC free article] [PubMed] [Google Scholar]
  111. Mondal I, Das O, Sun R, Gao J, Yu B, Diaz A, Behnan J, Dubey A, Meng Z, Eskandar E, Xu B, Lu RO, Ho WS (2023) PP2Ac deficiency enhances tumor immunogenicity by activating STING-type I interferon signaling in glioblastoma. Cancer Res 83:2527–2542 [DOI] [PMC free article] [PubMed] [Google Scholar]
  112. Motani K, Ito S, Nagata S (2015) DNA-mediated Cyclic GMP-AMP synthase-dependent and -independent regulation of innate immune responses. J Immunol 194(10):4914–4923 [DOI] [PubMed] [Google Scholar]
  113. Movert E, Lienard J, Valfridsson C, Nordström T, Johansson-Lindbom B, Carlsson F (2018) Streptococcal M protein promotes IL-10 production by cGAS-independent activation of the STING signaling pathway. PLoS Pathog 14(3):e1006969 [DOI] [PMC free article] [PubMed] [Google Scholar]
  114. Najem H, Lea ST, Tripathi S, Hurley L, Chen CH, William I, Sooreshjani M, Bowie M, Hartley G, Dussold C, Pacheco S, Dmello C, Lee-Chang C, McCortney K, Steffens A, Walshon J, Ott M, Wei J, Marisetty A, Balyasnikova I, Stupp R, Lukas RV, Hu J, James CD, Horbinski CM, Lesniak MS, Ashley DM, Priebe W, Platanias LC, Curran MA, Heimberger AB (2024) STING agonist 8803 reprograms the immune microenvironment and increases survival in preclinical models of glioblastoma. J Clin Invest 134(12):e175033 [DOI] [PMC free article] [PubMed] [Google Scholar]
  115. Nazmi A, Mukhopadhyay R, Dutta K, Basu A (2012) STING mediates neuronal innate immune response following Japanese encephalitis virus infection. Sci Rep 2:347 [DOI] [PMC free article] [PubMed] [Google Scholar]
  116. Nazmi A, Field RH, Griffin EW, Haugh O, Hennessy E, Cox D, Reis R, Tortorelli L, Murray CL, Lopez-Rodriguez AB, Jin L, Lavelle EC, Dunne A, Cunningham C (2019) Chronic neurodegeneration induces type I interferon synthesis via STING, shaping microglial phenotype and accelerating disease progression. Glia 67(7):1254–1276 [DOI] [PMC free article] [PubMed] [Google Scholar]
  117. Nelson TJ, Xu Y (2023) Sting and p53 DNA repair pathways are compromised in Alzheimer’s disease. Sci Rep 13(1):8304 [DOI] [PMC free article] [PubMed] [Google Scholar]
  118. Ohkuri T, Ghosh A, Kosaka A, Zhu J, Ikeura M, David M, Watkins SC, Sarkar SN, Okada H (2014) STING contributes to antiglioma immunity via triggering type I IFN signals in the tumor microenvironment. Cancer Immunol Res 2(12):1199–1208 [DOI] [PMC free article] [PubMed] [Google Scholar]
  119. Packer JM, Bray CE, Beckman NB, Wangler LM, Davis AC, Goodman EJ, Klingele NE, Godbout JP (2024) Impaired cortical neuronal homeostasis and cognition after diffuse traumatic brain injury are dependent on microglia and type I interferon responses. Glia 72(2):300–321 [DOI] [PMC free article] [PubMed] [Google Scholar]
  120. Pang W, Hu F (2023) C9ORF72 suppresses JAK-STAT mediated inflammation. iScience 26(5):106579 [DOI] [PMC free article] [PubMed] [Google Scholar]
  121. Parker ZM, Murphy AA, Leib DA (2015) Role of the DNA sensor STING in protection from lethal infection following corneal and intracerebral challenge with herpes simplex virus 1. J Virol 89(21):11080–11091 [DOI] [PMC free article] [PubMed] [Google Scholar]
  122. Peng Y, Zhuang J, Ying G, Zeng H, Zhou H, Cao Y, Chen H, Xu C, Fu X, Xu H, Li J, Cao S, Chen J, Gu C, Yan F, Chen G (2020) Stimulator of IFN genes mediates neuroinflammatory injury by suppressing AMPK signal in experimental subarachnoid hemorrhage. J Neuroinflamm 17(1):165 [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
  123. Petrovic M, Majchrzak OB, Marecar R, Laingoniaina AC, Walker PR, Borchard G, Jordan O, Tankov S (2024) Combining antimiR-25 and cGAMP nanocomplexes enhances immune responses via M2 macrophage reprogramming. Int J Mol Sci 25(23):12787 [DOI] [PMC free article] [PubMed] [Google Scholar]
  124. Pokatayev V, Yang K, Tu X, Dobbs N, Wu J, Kalb RG, Yan N (2020) Homeostatic regulation of STING protein at the resting state by stabilizer TOLLIP. Nat Immunol 21(2):158–167 [DOI] [PMC free article] [PubMed] [Google Scholar]
  125. Quek H, Luff J, Cheung K, Kozlov S, Gatei M, Lee CS, Bellingham MC, Noakes PG, Lim YC, Barnett NL, Dingwall S, Wolvetang E, Mashimo T, Roberts TL, Lavin MF (2017) A rat model of ataxia-telangiectasia: evidence for a neurodegenerative phenotype. Hum Mol Genet 26(1):109–123 [DOI] [PubMed] [Google Scholar]
  126. Reinert LS, Lopusna K, Winther H, Sun C, Thomsen MK, Nandakumar R, Mogensen TH, Meyer M, Vaegter C, Nyengaard JR, Fitzgerald KA, Paludan SR (2016) Sensing of HSV-1 by the cGAS-STING pathway in microglia orchestrates antiviral defence in the CNS. Nat Commun 7:13348 [DOI] [PMC free article] [PubMed] [Google Scholar]
  127. Reinert LS, Rashidi AS, Tran DN, Katzilieris-Petras G, Hvidt AK, Gohr M, Fruhwurth S, Bodda C, Thomsen MK, Vendelbo MH, Khan AR, Hansen B, Bergstrom P, Agholme L, Mogensen TH, Christensen MH, Nyengaard JR, Sen GC, Zetterberg H, Verjans GM, Paludan SR (2021) Brain immune cells undergo cGAS/STING-dependent apoptosis during herpes simplex virus type 1 infection to limit type I IFN production. J Clin Invest. 10.1172/JCI136824 [DOI] [PMC free article] [PubMed] [Google Scholar]
  128. Saraiva C, Praca C, Ferreira R, Santos T, Ferreira L, Bernardino L (2016) Nanoparticle-mediated brain drug delivery: overcoming blood-brain barrier to treat neurodegenerative diseases. J Control Release 235:34–47 [DOI] [PubMed] [Google Scholar]
  129. Schwab N, Taskina D, Leung E, Innes BT, Bader GD, Hazrati LN (2022) Neurons and glial cells acquire a senescent signature after repeated mild traumatic brain injury in a sex-dependent manner. Front Neurosci 16:1027116 [DOI] [PMC free article] [PubMed] [Google Scholar]
  130. Sen T, Saha P, Gupta R, Foley LM, Jiang T, Abakumova OS, Hitchens TK, Sen N (2020) Aberrant ER stress induced neuronal-IFNbeta elicits white matter injury due to microglial activation and T-cell infiltration after TBI. J Neurosci 40(2):424–446 [DOI] [PMC free article] [PubMed] [Google Scholar]
  131. Shao L, Hu F, Xu R, Nie H, Zhang H, Zhang P (2024) METTL14 regulates the m6A modification of TRAF6 to suppress mitochondrial dysfunction and ferroptosis in dopaminergic neurons via the cGAS-STING pathway. Curr Mol Med 24(12):1518–1528 [DOI] [PubMed] [Google Scholar]
  132. Sharma M, Rajendrarao S, Shahani N, Ramirez-Jarquin UN, Subramaniam S (2020) Cyclic GMP-AMP synthase promotes the inflammatory and autophagy responses in Huntington disease. Proc Natl Acad Sci USA 117(27):15989–15999 [DOI] [PMC free article] [PubMed] [Google Scholar]
  133. Sharma KB, Chhabra S, Aggarwal S, Tripathi A, Banerjee A, Yadav AK, Vrati S, Kalia M (2021) Proteomic landscape of Japanese encephalitis virus-infected fibroblasts. J Gen Virol 102(9):001657 [DOI] [PubMed] [Google Scholar]
  134. Shen A, Chen M, Chen Q, Liu Z, Zhang A (2022) Recent advances in the development of STING inhibitors: an updated patent review. Expert Opin Ther Pat 32(11):1131–1143 [DOI] [PubMed] [Google Scholar]
  135. Shi J, Yang Y, Yin N, Liu C, Zhao Y, Cheng H, Zhou T, Zhang Z, Zhang K (2022) Engineering CXCL12 biomimetic decoy-integrated versatile immunosuppressive nanoparticle for ischemic stroke therapy with management of overactivated brain immune microenvironment. Small Methods 6(1):e2101158 [DOI] [PubMed] [Google Scholar]
  136. Shi G, Liu L, Cao Y, Ma G, Zhu Y, Xu J, Zhang X, Li T, Mi L, Jia H, Zhang Y, Liu X, Zhou Y, Li S, Yang G, Liu X, Chen F, Wang B, Deng Q, Zhang S, Zhang J (2023) Inhibition of neutrophil extracellular trap formation ameliorates neuroinflammation and neuronal apoptosis via STING-dependent IRE1alpha/ASK1/JNK signaling pathway in mice with traumatic brain injury. J Neuroinflamm 20(1):222 [DOI] [PMC free article] [PubMed] [Google Scholar]
  137. Shih AY, Damm-Ganamet KL, Mirzadegan T (2018) Dynamic structural differences between human and mouse STING lead to differing sensitivity to DMXAA. Biophys J 114(1):32–39 [DOI] [PMC free article] [PubMed] [Google Scholar]
  138. Sliter DA, Martinez J, Hao L, Chen X, Sun N, Fischer TD, Burman JL, Li Y, Zhang Z, Narendra DP, Cai H, Borsche M, Klein C, Youle RJ (2018) Parkin and PINK1 mitigate STING-induced inflammation. Nature 561(7722):258–262 [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
  139. Song X, Ma F, Herrup K (2019) Accumulation of cytoplasmic DNA Due to ATM deficiency activates the microglial viral response system with neurotoxic consequences. J Neurosci 39(32):6378–6394 [DOI] [PMC free article] [PubMed] [Google Scholar]
  140. Song N, Song R, Ma P (2022) MiR-340-5p alleviates neuroinflammation and neuronal injury via suppressing STING in subarachnoid hemorrhage. Brain Behav 12(9):e2687 [DOI] [PMC free article] [PubMed] [Google Scholar]
  141. Song M, Qiang Y, Wang S, Shan S, Zhang L, Liu C, Song F, Zhao X (2024) High-fat diet exacerbates 1-Bromopropane-induced loss of dopaminergic neurons in the substantia nigra of mice through mitochondrial damage associated necroptotic pathway. Ecotoxicol Environ Saf 276:116280 [DOI] [PubMed] [Google Scholar]
  142. Sui H, Sun Z, Liu C, Xi H (2025) Ferritinophagy promotes microglia ferroptosis to aggravate neuroinflammation induced by cerebral ischemia-reperfusion injury via activation of the cGAS-STING signaling pathway. Neurochem Int 183:105920 [DOI] [PubMed] [Google Scholar]
  143. Sun Z, Hornung V (2022) cGAS-STING signaling. Curr Biol 32(13):R730–R734 [DOI] [PubMed] [Google Scholar]
  144. Sun L, Wu J, Du F, Chen X, Chen ZJ (2013) Cyclic GMP-AMP synthase is a cytosolic DNA sensor that activates the type I interferon pathway. Science 339(6121):786–791 [DOI] [PMC free article] [PubMed] [Google Scholar]
  145. Sun S, Lv W, Li S, Zhang Q, He W, Min Z, Teng C, Chen Y, Liu L, Yin J, Zhu B, Xu M, Dai D, Xin H (2023) Smart liposomal nanocarrier enhanced the treatment of ischemic stroke through neutrophil extracellular traps and cyclic guanosine monophosphate-adenosine monophosphate synthase-stimulator of interferon genes (cGAS-STING) pathway inhibition of Ischemic penumbra. ACS Nano 17(18):17845–17857 [DOI] [PubMed] [Google Scholar]
  146. Suschak JJ, Wang S, Fitzgerald KA, Lu S (2016) A cGAS-independent STING/IRF7 pathway mediates the immunogenicity of DNA vaccines. J Immunol 196(1):310–316 [DOI] [PMC free article] [PubMed] [Google Scholar]
  147. Szego EM, Malz L, Bernhardt N, Rosen-Wolff A, Falkenburger BH, Luksch H (2022) Constitutively active STING causes neuroinflammation and degeneration of dopaminergic neurons in mice. Elife 11:e81943 [DOI] [PMC free article] [PubMed] [Google Scholar]
  148. Taffoni C, Marines J, Chamma H, Guha S, Saccas M, Bouzid A, Valadao AC, Maghe C, Jardine J, Park MK, Polak K, De Martino M, Vanpouille-Box C, Del Rio M, Gongora C, Gavard J, Bidere N, Song MS, Pineau D, Hugnot JP, Kissa K, Fontenille L, Blanchet FP, Vila IK, Laguette N (2023) DNA damage repair kinase DNA-PK and cGAS synergize to induce cancer-related inflammation in glioblastoma. EMBO J 42(7):e111961 [DOI] [PMC free article] [PubMed] [Google Scholar]
  149. Tan HY, Yong YK, Xue YC, Liu H, Furihata T, Shankar EM, Ng CS (2022) cGAS and DDX41-STING mediated intrinsic immunity spreads intercellularly to promote neuroinflammation in SOD1 ALS model. iScience 25(6):104404 [DOI] [PMC free article] [PubMed] [Google Scholar]
  150. Tankov S, Petrovic M, Lecoultre M, Espinoza F, El-Harane N, Bes V, Chliate S, Bedoya DM, Jordan O, Borchard G, Migliorini D, Dutoit V, Walker PR (2024) Hypoxic glioblastoma-cell-derived extracellular vesicles impair cGAS-STING activity in macrophages. Cell Commun Signal 22(1):144 [DOI] [PMC free article] [PubMed] [Google Scholar]
  151. Tu Y, Yin XJ, Liu Q, Zhang S, Wang J, Ji BZ, Zhang J, Sun MS, Yang Y, Wang CH, Yin L, Liu Y (2022) MITA oligomerization upon viral infection is dependent on its N-glycosylation mediated by DDOST. PLoS Pathog 18(11):e1010989 [DOI] [PMC free article] [PubMed] [Google Scholar]
  152. von Roemeling CA, Wang Y, Qie Y, Yuan H, Zhao H, Liu X, Yang Z, Yang M, Deng W, Bruno KA, Chan CK, Lee AS, Rosenfeld SS, Yun K, Johnson AJ, Mitchell DA, Jiang W, Kim BYS (2020) Therapeutic modulation of phagocytosis in glioblastoma can activate both innate and adaptive antitumour immunity. Nat Commun 11(1):1508 [DOI] [PMC free article] [PubMed] [Google Scholar]
  153. Wang X, Majumdar T, Kessler P, Ozhegov E, Zhang Y, Chattopadhyay S, Barik S, Sen GC (2016) STING requires the adaptor TRIF to trigger innate immune responses to microbial infection. Cell Host Microbe 20(3):329–341 [DOI] [PMC free article] [PubMed] [Google Scholar]
  154. Wang S, Wang L, Qin X, Turdi S, Sun D, Culver B, Reiter RJ, Wang X, Zhou H, Ren J (2020) 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 5(1):119 [DOI] [PMC free article] [PubMed] [Google Scholar]
  155. Wang R, Zhu Y, Liu Z, Chang L, Bai X, Kang L, Cao Y, Yang X, Yu H, Shi MJ, Hu Y, Fan W, Zhao BQ (2021) Neutrophil extracellular traps promote tPA-induced brain hemorrhage via cGAS in mice with stroke. Blood 138(1):91–103 [DOI] [PMC free article] [PubMed] [Google Scholar]
  156. Wang B, Tang M, Yuan Z, Li Z, Hu B, Bai X, Chu J, Xu X, Zhang XQ (2022a) Targeted delivery of a STING agonist to brain tumors using bioengineered protein nanoparticles for enhanced immunotherapy. Bioact Mater 16:232–248 [DOI] [PMC free article] [PubMed] [Google Scholar]
  157. Wang Y, Luo W, Wang X, Ma Y, Huang L, Wang Y (2022b) MAMDC2, a gene highly expressed in microglia in experimental models of Alzheimers disease, positively regulates the innate antiviral response during neurotropic virus infection. J Infect 84(2):187–204 [DOI] [PubMed] [Google Scholar]
  158. Wang YS, Liu D, Wang X, Luo QL, Ding L, Fan DY, Cai QL, Tang CY, Yang W, Guan YG, Li TF, Wang PG, Luan GM, An J (2022c) Rasmussen’s encephalitis is characterized by relatively lower production of IFN-beta and activated cytotoxic T cell upon herpes viruses infection. J Neuroinflamm 19(1):70 [DOI] [PMC free article] [PubMed] [Google Scholar]
  159. Wang B, Wang Y, Qiu J, Gao S, Yu S, Sun D, Lou H (2023) The STING inhibitor C-176 attenuates MPTP-induced neuroinflammation and neurodegeneration in mouse parkinsonian models. Int Immunopharmacol 124(Pt A):110827 [DOI] [PubMed] [Google Scholar]
  160. Wang B, Yu W, Jiang H, Meng X, Tang D, Liu D (2024a) Clinical applications of STING agonists in cancer immunotherapy: current progress and future prospects. Front Immunol 15:1485546 [DOI] [PMC free article] [PubMed] [Google Scholar]
  161. Wang M, Tian T, Zhou H, Jiang SY, Jiao YY, Zhu Z, Xia J, Ma JH, Du RH (2024b) Metformin normalizes mitochondrial function to delay astrocyte senescence in a mouse model of Parkinson’s disease through Mfn2-cGAS signaling. J Neuroinflamm 21(1):81 [DOI] [PMC free article] [PubMed] [Google Scholar]
  162. Wang S, Qi Y, Zhao R, Pan Z, Li B, Qiu W, Zhao S, Guo X, Ni S, Li G, Xue H (2024c) Copy number gain of FAM131B-AS2 promotes the progression of glioblastoma by mitigating replication stress. Neuro Oncol 26(6):1027–1041 [DOI] [PMC free article] [PubMed] [Google Scholar]
  163. Wang-Bishop L, Wehbe M, Shae D, James J, Hacker BC, Garland K, Chistov PP, Rafat M, Balko JM, Wilson JT (2020) Potent STING activation stimulates immunogenic cell death to enhance antitumor immunity in neuroblastoma. J Immunother Cancer 8(1):e000282 [DOI] [PMC free article] [PubMed] [Google Scholar]
  164. Wangler LM, Bray CE, Packer JM, Tapp ZM, Davis AC, O’Neil SM, Baetz K, Ouvina M, Witzel M, Godbout JP (2022) Amplified gliosis and interferon-associated inflammation in the aging brain following diffuse traumatic brain injury. J Neurosci 42(48):9082–9096 [DOI] [PMC free article] [PubMed] [Google Scholar]
  165. Wolpaw AJ, Grossmann LD, Dessau JL, Dong MM, Aaron BJ, Brafford PA, Volgina D, Pascual-Pasto G, Rodriguez-Garcia A, Uzun Y, Arsenian-Henriksson M, Powell Jr DJ, Bosse KR, Kossenkov A, Tan K, Hogarty MD, Maris JM, Dang CV (2022) Epigenetic state determines inflammatory sensing in neuroblastoma. Proc Natl Acad Sci USA 119(6) [DOI] [PMC free article] [PubMed]
  166. Wu JJ, Zhao L, Hu HG, Li WH, Li YM (2020) Agonists and inhibitors of the STING pathway: potential agents for immunotherapy. Med Res Rev 40(3):1117–1141 [DOI] [PubMed] [Google Scholar]
  167. Wu C, Zhang S, Sun H, Li A, Hou F, Qi L, Liao H (2024) STING inhibition suppresses microglia-mediated synapses engulfment and alleviates motor functional deficits after stroke. J Neuroinflamm 21(1):86 [DOI] [PMC free article] [PubMed] [Google Scholar]
  168. Xia X, He X, Zhao T, Yang J, Bi Z, Fu Q, Liu J, Ao D, Wei Y, Wei X (2023) Inhibiting mtDNA-STING-NLRP3/IL-1beta axis-mediated neutrophil infiltration protects neurons in Alzheimer’s disease. Cell Prolif 57:e13529 [DOI] [PMC free article] [PubMed] [Google Scholar]
  169. Xie X, Ma G, Li X, Zhao J, Zhao Z, Zeng J (2023) Activation of innate immune cGAS-STING pathway contributes to Alzheimer’s pathogenesis in 5xFAD mice. Nat Aging 3(2):202–212 [DOI] [PubMed] [Google Scholar]
  170. Xu Q, Xu W, Cheng H, Yuan H, Tan X (2019) Efficacy and mechanism of cGAMP to suppress Alzheimer’s disease by elevating TREM2. Brain Behav Immun 81:495–508 [DOI] [PubMed] [Google Scholar]
  171. Xu H, Jin J, Chen Y, Wu G, Zhu H, Wang Q, Wang J, Li S, Grigore FN, Ma J, Chen CC, Lan Q, Li M (2022) GBP3 promotes glioblastoma resistance to temozolomide by enhancing DNA damage repair. Oncogene 41(31):3876–3885 [DOI] [PubMed] [Google Scholar]
  172. Yang K, Han W, Jiang X, Piffko A, Bugno J, Han C, Li S, Liang H, Xu Z, Zheng W, Wang L, Wang J, Huang X, Ting JPY, Fu YX, Lin W, Weichselbaum RR (2022) Zinc cyclic di-AMP nanoparticles target and suppress tumours via endothelial STING activation and tumour-associated macrophage reinvigoration. Nat Nanotechnol 17(12):1322–1331 [DOI] [PubMed] [Google Scholar]
  173. Yang G, Kantapan J, Mazhar M, Hu Q, Bai X, Zou Y, Wang H, Yang S, Wang L, Dechsupa N (2024a) Pretreated MSCs with IronQ transplantation attenuate microglia neuroinflammation via the cGAS-STING signaling pathway. J Inflamm Res 17:1643–1658 [DOI] [PMC free article] [PubMed] [Google Scholar]
  174. Yang Y, Luo X, Wang Y, Xu A, Peng L, Zhang X, Wang Z, Ying Y, Li K (2024b) beta-Mangostin targets and suppresses glioma via STING activation and tumor-associated microglia polarization. Biomed Pharmacother 177:117074 [DOI] [PubMed] [Google Scholar]
  175. Yildirim Z, Dogan E, Guler Kara H, Kosova B, Bozok V (2024) STING activation increases the efficiency of temozolomide in PTEN harbouring glioblastoma cells. Turk J Med Sci 54(3):607–614 [DOI] [PMC free article] [PubMed] [Google Scholar]
  176. You F, Wang P, Yang L, Yang G, Zhao YO, Qian F, Walker W, Sutton R, Montgomery R, Lin R, Iwasaki A, Fikrig E (2013) ELF4 is critical for induction of type I interferon and the host antiviral response. Nat Immunol 14(12):1237–1246 [DOI] [PMC free article] [PubMed] [Google Scholar]
  177. Yu CH, Davidson S, Harapas CR, Hilton JB, Mlodzianoski MJ, Laohamonthonkul P, Louis C, Low RRJ, Moecking J, De Nardo D, Balka KR, Calleja DJ, Moghaddas F, Ni E, McLean CA, Samson AL, Tyebji S, Tonkin CJ, Bye CR, Turner BJ, Pepin G, Gantier MP, Rogers KL, McArthur K, Crouch PJ, Masters SL (2020) TDP-43 triggers mitochondrial DNA release via mPTP to activate cGAS/STING in ALS. Cell 183(3):636–649618 [DOI] [PMC free article] [PubMed] [Google Scholar]
  178. Yu Z, Zheng L, Geng Y, Zhang Y, Wang Y, You G, Cai M, Li M, Cheng X, Zan J (2023) FTO alleviates cerebral ischemia/reperfusion-induced neuroinflammation by decreasing cGAS mRNA stability in an m6A-dependent manner. Cell Signal 109:110751 [DOI] [PubMed] [Google Scholar]
  179. Yu H, Liu W, Ding K, Wu J, Wang C, Wang S, Wu L, Tang Q, Yin X, Jiang K, Yan D, Wang X, Chen S, Yan S (2025) Sequential release HydroLipo system for STING gene epigenetic reprogramming and immune activation in glioblastoma. Adv Sci (Weinh) 12(5):e2408323 [DOI] [PMC free article] [PubMed] [Google Scholar]
  180. Yuan H, Yang J, Qin G, Sun Y, Zhao C, Wang C, Ren J, Qu X (2025) Regulation of STING G-quadruplex for rescuing cellular senescence and Abeta phagocytic capacity of microglia. Chem Sci 16(2):693–699 [DOI] [PMC free article] [PubMed] [Google Scholar]
  181. Zamiri K, Kesari S, Paul K, Hwang SH, Hammock B, Kaczor-Urbanowicz KE, Urbanowicz A, Gao L, Whitelegge J, Fiala M (2023) Therapy of autoimmune inflammation in sporadic amyotrophic lateral sclerosis: dimethyl fumarate and H-151 downregulate inflammatory cytokines in the cGAS-STING pathway. FASEB J 37(8):e23068 [DOI] [PMC free article] [PubMed] [Google Scholar]
  182. Zhan Y, Huang J, Tang X, Du B, Yang B (2025) Semen strychni pulveratum and vomicine alleviate neuroinflammation in amyotrophic lateral sclerosis through cGAS-STING-TBK1 pathway. J Ethnopharmacol 336:118741 [DOI] [PubMed] [Google Scholar]
  183. Zhang C, Shang G, Gui X, Zhang X, Bai XC, Chen ZJ (2019) Structural basis of STING binding with and phosphorylation by TBK1. Nature 567(7748):394–398 [DOI] [PMC free article] [PubMed] [Google Scholar]
  184. Zhang CX, Huang DJ, Baloche V, Zhang L, Xu JX, Li BW, Zhao XR, He J, Mai HQ, Chen QY, Zhang XS, Busson P, Cui J, Li J (2020) Galectin-9 promotes a suppressive microenvironment in human cancer by enhancing STING degradation. Oncogenesis 9(7):65 [DOI] [PMC free article] [PubMed] [Google Scholar]
  185. Zhang LM, Xin Y, Wu ZY, Song RX, Miao HT, Zheng WC, Li Y, Zhang DX, Zhao XC (2022) STING mediates neuroinflammatory response by activating NLRP3-related pyroptosis in severe traumatic brain injury. J Neurochem 162(5):444–462 [DOI] [PubMed] [Google Scholar]
  186. Zhang P, Rashidi A, Zhao J, Silvers C, Wang H, Castro B, Ellingwood A, Han Y, Lopez-Rosas A, Zannikou M, Dmello C, Levine R, Xiao T, Cordero A, Sonabend AM, Balyasnikova IV, Lee-Chang C, Miska J, Lesniak MS (2023a) STING agonist-loaded, CD47/PD-L1-targeting nanoparticles potentiate antitumor immunity and radiotherapy for glioblastoma. Nat Commun 14(1):1610 [DOI] [PMC free article] [PubMed] [Google Scholar]
  187. Zhang W, Xu M, Chen F, Su Y, Yu M, Xing L, Chang Y, Yan T (2023b) Targeting the JAK2-STAT3 pathway to inhibit cGAS-STING activation improves neuronal senescence after ischemic stroke. Exp Neurol 368:114474 [DOI] [PubMed] [Google Scholar]
  188. Zhang Y, Yan J, Hou X, Wang C, Kang DD, Xue Y, Du S, Deng B, McComb DW, Liu SL, Zhong Y, Dong Y (2023c) STING agonist-derived LNP-mRNA vaccine enhances protective immunity against SARS-CoV-2. Nano Lett 23(7):2593–2600 [DOI] [PMC free article] [PubMed] [Google Scholar]
  189. Zhang W, Lu L, Zhu Z, Deng F, Zhang W, Wang F, Zeng P, Shi H, Wang T, Chen Y, Song Y, Liu Y, Kang T, Li K, Mao J, Liu Z, Zhang L (2024) A manganese-based nanodriver coordinates tumor prevention and suppression through STING activation in glioblastoma. Adv Healthc Mater 13(19):e2400421 [DOI] [PubMed] [Google Scholar]
  190. Zhao M, Wang B, Zhang C, Su Z, Guo B, Zhao Y, Zheng R (2021) The DJ1-Nrf2-STING axis mediates the neuroprotective effects of Withaferin A in Parkinson’s disease. Cell Death Differ 28(8):2517–2535 [DOI] [PMC free article] [PubMed] [Google Scholar]
  191. Zhao L, Li Y, Wang W, Qi X, Wang S, Song W, Li T, Gao W (2024) Regulating NCOA4-Mediated ferritinophagy for therapeutic intervention in cerebral ischemia-reperfusion injury. Neurochem Res 49(7):1806–1822 [DOI] [PubMed] [Google Scholar]
  192. Zhong M, Long M, Han C, Ji S, Yang Q (2024) STING is significantly increased in high-grade glioma with high risk of recurrence. Oncoimmunology 13(1):2327682 [DOI] [PMC free article] [PubMed] [Google Scholar]
  193. Zhou Y, Guo Y, Chen L, Zhang X, Wu W, Yang Z, Li X, Wang Y, Hu Z, Wang Z (2022) Co-delivery of phagocytosis checkpoint and STING agonist by a Trojan horse nanocapsule for orthotopic glioma immunotherapy. Theranostics 12(12):5488–5503 [DOI] [PMC free article] [PubMed] [Google Scholar]
  194. Zhou S, Li T, Zhang W, Wu J, Hong H, Quan W, Qiao X, Cui C, Qiao C, Zhao W, Shen Y (2025) The cGAS-STING-interferon regulatory factor 7 pathway regulates neuroinflammation in Parkinson’s disease. Neural Regen Res 20(8):2361–2372 [DOI] [PMC free article] [PubMed] [Google Scholar]
  195. Zhu W, Rao J, Zhang LH, Xue KM, Li L, Li JJ, Chen QZ, Fu R (2024) OMA1 competitively binds to HSPA9 to promote mitophagy and activate the cGAS-STING pathway to mediate GBM immune escape. J Immunother Cancer 12(4):e008718 [DOI] [PMC free article] [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Data Availability Statement

No datasets were generated or analysed during the current study.


Articles from Cellular and Molecular Neurobiology are provided here courtesy of Springer

RESOURCES