echemi logo
Product
  • Product
  • Supplier
  • Inquiry
    Home > Active Ingredient News > Study of Nervous System > J Neuroinflammation Review—Wenfei Ni/Kailiang Zhou focused on the important role of STING pathway in neuroinflammation and cell death after CNS injury

    J Neuroinflammation Review—Wenfei Ni/Kailiang Zhou focused on the important role of STING pathway in neuroinflammation and cell death after CNS injury

    • Last Update: 2022-10-20
    • Source: Internet
    • Author: User
    Search more information of high quality chemicals, good prices and reliable suppliers, visit www.echemi.com


    Written by Hu Xinli - Wang Sizhen, Fang Yiyi

    Editor—Summer Leaf


    Central Nervous System (CNS) injury refers to the damage of the brain and spinal cord to external factors, mainly including traumatic brain injury (TBI), spinal cord injury ().
    SCI
    ), subarachnoid hemorrhage (SAH), and hypoxic-ischemic encephalopathy (HIE), among others [1–4].

    Due to the poor repair and regeneration ability of nerve cells, serious sequelae are often caused after injury, which brings a heavy burden to the patient's family and society [5].

    However, the current treatment of CNS injury has not achieved satisfactory results, so it is of great practical significance to find effective therapeutic targets and treatment options.

    The innate immune response plays an important role in CNS injury by repairing damaged tissues as well as the host to fight off infection and coordinating the healing of injured tissues [6].

    CNS injury usually occurs at the moment of injury, but the inflammatory microenvironment and uncontrolled cell death after secondary injury can further exacerbate the injury [7].

    Therefore, controlling neuroinflammation and inhibiting cell death are essential
    for the physiological function of the CNS as well as post-injury repair.


    Interferon gene stimulatory factor (Stimulator of Interferon Genes, STING As a central immune molecule in many signaling pathways, it is usually in a self-inhibited state in the form of dimers, and protein configuration changes occur after stimulation by upstream signals, and then are activated, which in turn causes the activation and production of a series of downstream immune cells and inflammatory factors, thereby promoting the occurrence of inflammatory reactions in the body and producing corresponding clinical symptoms [8].

    Studies have shown that
    the STING pathway is involved in the inflammatory response and nerve cell death after
    CNS injury.
    Therefore, the modulated cGAS-STING signaling pathway has become a frontier hot spot
    in the research of CNS therapy and post-injury regenerative repair.


    On October 4, 2022, the team of Ni Wenfei and Zhou Kailiang from the Second Affiliated Hospital of Wenzhou Medical University/Zhejiang Key Laboratory of Orthopedics held a meeting in the Journal of Neuroinflammation Published a review "Emerging role of STING signalling in CNS injury: infammation, autophagy, necroptosis, ferroptosis and pyroptosis", first author Dr.
    Xinli Hu and Haojie Zhang, M.
    S.
    , Corresponding Author
    Wenfei Ni ( Wenfei Ni) chief physician and associate researcher
    of Kailiang Zhou.
    This article systematically summarizes the frontier progress of STING abnormal activation to promote inflammation and cell death in CNS injury research, in addition STING signaling is involved in programmed cell death during CNS injury [Autophagy, cell pyrosis Pyroptosis), Ferroptosis, and Necroptosis].
    of the latest developments
    .
    On this basis, the potential drug regulation cGAS-STING signaling pathway in CNS damage is summarized It provides new ideas
    for the treatment of CNS injury.



    First, the cGAS-STING signal path

    cGAS (circular GMP-AMP synthetase) recognizes double-stranded DNA in the cytoplasm and catalyzes the synthesis of GTP and ATP into loops GMP-AMPcGAMP[9]
    cGAMP binds to and activates the endoplasmic reticulum protein STING, prompting STING translocation to the Golgi apparatus, which can subsequently activate the downstream TBK1, as well as the phosphorylated transcription factor IRF3, cytosolic IRF3 dimer enters the nucleus after phosphorylation, resulting in induction and I of IRF3 target gene transcription The release of type interferon, which promotes the occurrence of inflammation [10] (Figure 1).


    Figure 1 cGAS-STING signaling path

    (Source: Hu XL et al, J Neuroinflammation, 2022).



    Second, cGAS-STING signaling pathway and CNS damage

    In CNS including traumatic brain injury (TBI), spinal cord injury (SCI), subarachnoid hemorrhage (SAH), and hypoxic-ischemic encephalopathy (HIE).
    In the injury disease, excessive activation of the cGAS-STING signaling pathway has been found to mediate the overexpression of inflammation and the programmed death
    of nerve cells.
    Overactivated inflammation and cell death that cannot be effectively suppressed are considered to be
    one of the core problems in CNS injury, and how to inhibit excessive inflammatory activation and programmed cell death is the core step of
    treatment.
    This review summarizes the activation of STING in different types of CNS injuries and focuses on the latest research progress
    .




    cGAS-STING signaling pathway and programmed cell death

    (I) cGAS-STING The autophagy process with autophagy consists of five stages: induction phase - nucleation - extension - autophagosome formation - lysis [11]
    cGAS-STING activation can initiate autophagy, after the start of autophagy, cGAS-STING ubiquitinated and bound to p62, and then packaged into an autophagosome, and combined with lysosomes to form autophagy lysosomes, digest autophagy substrates, The final process of autophagy is completed (Figure 2).

    Figure 2 cGAS-STING and autophagy (Source: Hu XL et al, J Neuroinflammation, 2022



    (ii) cGAS-STING and cell necropolis apoptosis

    Activation of the cGAS-STING signaling pathway can mediate cell necrotizing apoptosis [12].

    Intracytoplasmic
    DNA activates the cGAS-STING signaling pathway leading to the production
    of IFN and TNF.
    The binding of TNF to TNFR1 results in the activation of RIPK1/RIPK3 when Caspase-8 is inhibited
    .
    RIPK1/RIPK3 can phosphorylate MLKL leading to cell necrotic apoptosis
    .
    In addition, the overexpression of IFN can upregulate RIPK3 and MLKL (Figure 3).

    Figure 3 cGAS-STING and cell necroposis (Source: Hu XL et al, J Neuroinflammation , 2022


    (iii) cGAS-STING and cell iron death

    System Xc− is a reversing carrier of isodimeric cystine/glutamate that introduces cystine to maintain redox homeostasis, which is then reduced to cysteine and used to synthesize the main antioxidant, GSH [13].

    The core regulatory molecule of cell iron death is
    GPX4, a family of enzymes that reduces peroxides
    at the expense of the oxidation of two GSH molecules.
    Inactivation of GPX4 can lead to iron death in cells [14].

    In addition
    , GPX4 can promote STING carbonylation and inhibit its transfer from the endoplasmic reticulum to the Golgi complex, which plays a role in inhibiting the expression
    of the cGAS-STING signaling pathway.
    Therefore, GPX4 is the junction of cell siderogenesis and STING (Figure 4).


    Figure 4 cGAS-STING and cell iron death

    (Source: Hu XL et al, J Neuroinflammation, 2022).


    (iv) cGAS-STING and cytopyrotic melanoma deficiency factor 2 (Absent in Melanoma 2, AIM2 Inflammasomes are a class of inflammasomes primarily expressed in neurons, and are the only known inflammasomes that recognize ectopic double-stranded DNA.

    Activated
    AIM2 inflammasomes can induce pyrosis in neurons [15].

    At the same time
    , cGAS-STING-mediated increase in IFN can promote AIM2 activity, thereby increasing the expression of Caspase-1 and promoting cell pyrosis
    In addition, Caspase-1 overexpression and potassium efflux due to pyroptosis can inhibit cGAS function (Figure 5).


    Figure 5 cGAS-STING and cell pyrosis

    (Source: Hu XL et al, J Neuroinflammation, 2022).



    Potential strategies for inhibiting STING signaling pathway in the treatment of CNS injury

    Aseptic inflammation and cell death caused by the cGAS-STING signaling pathway play an important role
    in CNS injury.
    Inhibitors of the cGAS-STING pathway may be potential targets for CNS injury therapy
    .
    cGAS, which catalyzes the production
    of cGAMP by recognizing DNA in the cytoplasm after cell death.
    The cGAMP then transmits a signal to the downstream STING, which translocates to the Golgi apparatus to activate downstream TBK1 and phosphorylated transcription factors IRF3, the cytosolic IRF3 dimer, enters the nucleus after phosphorylation, resulting in induction of transcription of the IRF3 target gene and release of type I interferon
    .
    This process activates an innate immune response
    [16].

    Therefore
    , DNA in the cytoplasm is an important factor in the activation of cGAS-STING, so it can reduce DNA in the cytoplasm and inhibit cGAS and STING to suppress the cGAS-STING signaling
    pathway.
    A number of drugs have been shown to inhibit the cGAS–STING signaling pathway and provide viable treatments for related diseases
    (Table 1) [17-20].

    However, more research
    is needed on the therapeutic effect of inhibiting the cGAS-STING pathway in the field of CNS injury.


    Table 1 Inhibitors of the cGAS-STING signaling pathway

    (Source: Hu XL et al, J Neuroinflammation, 2022).


    V.
    Summary and prospects


    Excessive inflammatory response and uncontrolled nerve cell death play an important role
    in CNS injury.
    As a newly discovered pathway in recent years, STING signaling pathway plays a very important role
    in regulating neuroinflammation and cell death.
    At present
    , there is an increasing number of research literature on cGAS-STING signaling pathway on CNS injury, and it is urgent to comprehensively summarize the role of cGAS-STING signaling pathway on CNS injury.
    Lay a solid foundation
    for follow-up research.
    Therefore,
    the purpose of this review is to summarize the research progress of cGAS-STING signaling pathway in CNS injury.
    The
    link between the cGAS-STING signaling pathway and programmed cell death was explored.
    Finally
    , the possible methods and strategies for alleviating neuroinflammation and cell death using cGAS-STING signaling pathway as a target are summarized, aiming to provide new ideas
    for CNS injury.





    Original link: style="margin-bottom: 0px;white-space: normal;margin-top: 0px;">

    National Natural Science Foundation of China (8207219), Zhejiang Public Welfare Technology Project (LGF20H150003), Natural Science Foundation of Zhejiang Province (LY17H060009, Y21H060050), Wenzhou Science and Technology Bureau Fund Support Project (Y20210438

    First authors: Dr.
    Hu Xinli (first from left), Master Zhang Haojie (second from left); Corresponding authors: Associate Researcher Zhou Kailiang (second from right), Chief Physician Ni Wenfei (first from right).

    (Photo courtesy of: Zhou Kailiang/Ni Wenfei team)




    A selection of past articles

    [1] Sci Adv-Sheng Neng-Yin/Mao Bingyu/Ding Yuqiang teamwork discovered a new mechanism of AMPA receptor ubiquitination in the regulation of excitatory synaptic function

    [2] Brain—Liu Gang/Hu Qingmao's team revealed the driving role of auxiliary motor areas in the change of whole brain structural network in patients with blepharospasm

    [3] FASEB J—Liu Yang's team found that antipsychotic drugs on experimental animals caused vascular abnormalities in hematopoietic organs

    [4] Science-Du Yang team and others collaborated to develop non-opioid non-addictive analgesics that target adrenergic receptors

    [5] NPJ Parkinsons Dis—Wang Xijim's team found that high PSQI is an independent risk factor for dyskinesia in Parkinson's disease

    [6] Ann Neurol-Chen Wanjin/Fan Dongsheng's research group revealed that heterozygous mutations in the SerRS gene cause peroneal muscular atrophy

    [7] Sci ADV—astrocyte subset plasticity regulates heroin relapse, providing a basis for the treatment of drug addiction relapse

    [8] J Neurosci-Hu Bo's research group revealed the role of dorsal hippocampal small albumin-positive interneurons in joint motor learning and their network activity mechanism

    [9] CNSNT—Gu Xiaoping's team revealed that enhancing astrocyte networks can improve brain network abnormalities and cognitive dysfunction caused by long-term isoflurane anesthesia

    [10] Nat Aging—Glial cells in mouse with Alzheimer's disease are involved in synaptic clearance through the complement pathway

    Recommended high-quality scientific research training courses[1] Symposium on Single Cell Sequencing and Spatial Transcriptomics Data Analysis (October 29-30, Tencent Online Conference)[2] Symposium on Patch-clamping and Optogenetics and Calcium Imaging Technology (October 15-16, 2022 Tencent Conference) Conference/Forum Preview & Review

    [1] Preview | Neuromodulation and Brain-Computer Interface Conference (October 13-14, Beijing time) (U.
    S.
    Pacific Time: October 12-13)

    [2] Conference report - The human brain and the machine are gradually getting closer, and the brain-computer interface "black technology" is shining into reality

    Welcome to "Logical Neuroscience"[1]" "Logical Neuroscience" Recruitment for Editor/Operation Positions ( Online Office)[2] Talent Recruitment - " Logical Neuroscience " Recruitment Article Interpretation/Writing Position ( Network Part-time, Online Office)
    References (Swipe up and down to read).


    1.
    Langlois JA, Rutland-Brown W, Wald MM.
    The epidemiology and impact of traumatic brain injury: a brief overview.
    J Head Trauma Rehabil.
    2006; 21:375–8.
    https://doi.
    org/10.
    1097/00001199-200609000-00001.

    2.
    Cao HQ, Dong ED.
    An update on spinal cord injury research.
    Neurosci Bull.
    2013; 29:94–102.
    https://doi.
    org/10.
    1007/s12264-012-1277-8.

    3.
    O’Donnell MJ, Xavier D, Liu L, Zhang H, Chin SL, Rao-Melacini P, Rangarajan S, Islam S, Pais P, McQueen MJ, et al.
    Risk factors for ischaemic and intracerebral haemorrhagic stroke in 22 countries (the INTERSTROKE study): a case-control study.
    Lancet.
    2010; 376:112–23.
    https://doi.
    org/10.
    1016/s0140-6736(10)60834-3.

    4.
    Sekhon LH, Fehlings MG.
    Epidemiology, demographics, and pathophysiology of acute spinal cord injury.
    Spine.
    2001; 26:S2-12.
    https://doi.
    org/10.
    1097/00007632-200112151-00002.

    5.
    Janca A, Aarli JA, Prilipko L, Dua T, Saxena S, Saraceno B.
    WHO/WFN Survey of neurological services: a worldwide perspective.
    J Neurol Sci.
    2006; 247:29–34.
    https://doi.
    org/10.
    1016/j.
    jns.
    2006.
    03.
    003.

    6.
    Werner C, Engelhard K.
    Pathophysiology of traumatic brain injury.
    Br J Anaesth.
    2007; 99:4–9.
    https://doi.
    org/10.
    1093/bja/aem131.

    7.
    Beattie MS, Hermann GE, Rogers RC, Bresnahan JC.
    Cell death in models of spinal cord injury.
    Prog Brain Res.
    2002; 137:37–47.
    https://doi.
    org/10.
    1016/s0079-6123(02)37006-7

    8.
    Balka KR, De Nardo D.
    Molecular and spatial mechanisms governing STING signalling.
    Febs j.
    2020.
    https://doi.
    org/10.
    1111/febs.
    15640.

    9.
    Gao P, Ascano M, Wu Y, Barchet W, Gafney BL, Zillinger T, Serganov AA, Liu Y, Jones RA, Hartmann G, et al.
    Cyclic [G(2’,5’)pA(3’,5’)p] is the metazoan second messenger produced by DNA-activated cyclic GMP-AMP synthase.
    Cell.
    2013; 153:1094–107.
    https://doi.
    org/10.
    1016/j.
    cell.
    2013.
    04.
    046.

    10.
    Chen Q, Sun L, Chen ZJ.
    Regulation and function of the cGAS-STING pathway of cytosolic DNA sensing.
    Nat Immunol.
    2016; 17:1142–9.
    https://doi.
    org/10.
    1038/ni.
    3558.

    11.
    Levine B, Mizushima N, Virgin HW.
    Autophagy in immunity and infammation.
    Nature.
    2011; 469:323–35.
    https://doi.
    org/10.
    1038/nature09782.

    12.
    Sarhan J, Liu BC, Muendlein HI, Weindel CG, Smirnova I, Tang AY, Ilyukha V, Sorokin M, Buzdin A, Fitzgerald KA, Poltorak A.
    Constitutive interferon signaling maintains critical threshold of MLKL expression to license necroptosis.
    Cell Death Difer.
    2019; 26:332–47.
    https://doi.
    org/10.
    1038/s41418-018-0122-7.

    13.
    Dixon SJ, Lemberg KM, Lamprecht MR, Skouta R, Zaitsev EM, Gleason CE, Patel DN, Bauer AJ, Cantley AM, Yang WS, et al.
    Ferroptosis: an irondependent form of nonapoptotic cell death.
    Cell.
    2012; 149:1060–72.
    https://doi.
    org/10.
    1016/j.
    cell.
    2012.
    03.
    042.

    14.
    Jia M, Qin D, Zhao C, Chai L, Yu Z, Wang W, Tong L, Lv L, Wang Y, Rehwinkel J, et al.
    Redox homeostasis maintained by GPX4 facilitates STING activation.
    Nat Immunol.
    2020; 21:727–35.
    https://doi.
    org/10.
    1038/s41590-020-0699-0

    15.
    Li XQ, Yu Q, Fang B, Zhang ZL, Ma H.
    Knockdown of the AIM2 molecule attenuates ischemia-reperfusion-induced spinal neuronal pyroptosis by inhibiting AIM2 infammasome activation and subsequent release of cleaved caspase-1 and IL-1β.
    Neuropharmacology.
    2019; 160: 107661.
    https://doi.
    org/10.
    1016/j.
    neuropharm.
    2019.
    05.
    038.

    16.
    Lousberg EL, Fraser CK, Tovey MG, Diener KR, Hayball JD.
    Type I interferons mediate the innate cytokine response to recombinant fowlpox virus but not the induction of plasmacytoid dendritic cell-dependent adaptive immunity.
    J Virol.
    2010; 84:6549–63.
    https://doi.
    org/10.
    1128/jvi.
    02618-09.

    17.
    Vincent J, Adura C, Gao P, Luz A, Lama L, Asano Y, Okamoto R, Imaeda T, Aida J, Rothamel K, et al.
    Small molecule inhibition of cGAS reduces interferon expression in primary macrophages from autoimmune mice.

    Nat Commun.
    2017; 8:750.
    https://doi.
    org/10.
    1038/s41467-017-00833-9.

    18.
    An J, Woodward JJ, Sasaki T, Minie M, Elkon KB.
    Cutting edge: antimalarial drugs inhibit IFN-β production through blockade of cyclic GMPAMP synthase-DNA interaction.
    J Immunol.
    2015; 194:4089–93.
    https://doi.
    org/10.
    4049/jimmunol.
    1402793.

    19.
    Dai J, Huang YJ, He X, Zhao M, Wang X, Liu ZS, Xue W, Cai H, Zhan XY, Huang SY, et al.
    Acetylation blocks cGAS activity and inhibits self-DNAinduced autoimmunity.
    Cell.
    2019; 176:1447-1460.
    e1414.
    https://doi.
    org/10.
    1016/j.
    cell.
    2019.
    01.
    016.

    20.
    Liu ZS, Cai H, Xue W, Wang M, Xia T, Li WJ, Xing JQ, Zhao M, Huang YJ, Chen S, et al.
    G3BP1 promotes DNA binding and activation of cGAS.
    Nat Immunol.
    2019; 20:18–28.
    https://doi.
    org/10.
    1038/s41590-018-0262-4.

    21.
    Gamdzyk M, Doycheva DM, Araujo C, Ocak U, Luo Y, Tang J, Zhang JH.
    cGAS/STING pathway activation contributes to delayed neurodegeneration in neonatal hypoxia-ischemia rat model: possible involvement of LINE-1.
    Mol Neurobiol.
    2020; 57:2600–19.
    https://doi.
    org/10.
    1007/s12035-020-01904-7.

    22.
    Abdullah A, Zhang M, Frugier T, Bedoui S, Taylor JM, Crack PJ.
    STINGmediated type-I interferons contribute to the neuroinfammatory process and detrimental efects following traumatic brain injury.
    J Neuroinfamm.
    2018; 15:323.
    https://doi.
    org/10.
    1186/s12974-018-1354-7.

    23.
    Peng Y, Zhuang J, Ying G, Zeng H, Zhou H, Cao Y, Chen H, Xu C, Fu X, Xu H, et al.
    Stimulator of IFN genes mediates neuroinfammatory injury by suppressing AMPK signal in experimental subarachnoid hemorrhage.
    J Neuroinfamm.
    2020; 17:165.
    https://doi.
    org/10.
    1186/s12974-020-01830-4

    24.
    Wang YY, Shen D, Zhao LJ, Zeng N, Hu TH.
    Sting is a critical regulator of spinal cord injury by regulating microglial infammation via interacting with TBK1 in mice.
    Biochem Biophys Res Commun.
    2019; 517:741–8.
    https://doi.
    org/10.
    1016/j.
    bbrc.
    2019.
    07.
    125.



    End of this article


    This article is an English version of an article which is originally in the Chinese language on echemi.com and is provided for information purposes only. This website makes no representation or warranty of any kind, either expressed or implied, as to the accuracy, completeness ownership or reliability of the article or any translations thereof. If you have any concerns or complaints relating to the article, please send an email, providing a detailed description of the concern or complaint, to service@echemi.com. A staff member will contact you within 5 working days. Once verified, infringing content will be removed immediately.

    Contact Us

    The source of this page with content of products and services is from Internet, which doesn't represent ECHEMI's opinion. If you have any queries, please write to service@echemi.com. It will be replied within 5 days.

    Moreover, if you find any instances of plagiarism from the page, please send email to service@echemi.com with relevant evidence.