Role of stimulator of interferon genes (STING) in the enteric nervous system in health and disease
Arun Balasubramaniam
Division of Digestive Diseases, Emory University School of Medicine, Atlanta, Georgia, USA
Atlanta Veterans Affairs Health Care System, Decatur, Georgia, USA
Search for more papers by this authorCorresponding Author
Shanthi Srinivasan
Division of Digestive Diseases, Emory University School of Medicine, Atlanta, Georgia, USA
Atlanta Veterans Affairs Health Care System, Decatur, Georgia, USA
Correspondence
Shanthi Srinivasan, Division of Digestive Diseases, Emory University School of Medicine, Atlanta, Georgia, USA.
Email: [email protected]
Search for more papers by this authorArun Balasubramaniam
Division of Digestive Diseases, Emory University School of Medicine, Atlanta, Georgia, USA
Atlanta Veterans Affairs Health Care System, Decatur, Georgia, USA
Search for more papers by this authorCorresponding Author
Shanthi Srinivasan
Division of Digestive Diseases, Emory University School of Medicine, Atlanta, Georgia, USA
Atlanta Veterans Affairs Health Care System, Decatur, Georgia, USA
Correspondence
Shanthi Srinivasan, Division of Digestive Diseases, Emory University School of Medicine, Atlanta, Georgia, USA.
Email: [email protected]
Search for more papers by this authorAbstract
Stimulator of Interferon Genes (STING) is a crucial protein that controls the immune system's reaction to bacterial and viral infections. As a pattern-recognition receptor, STING is found in immune cells as well as in neurons and glia in the enteric nervous system (ENS). Recent studies have linked STING to the pathogenesis of several neurological disorders like multiple sclerosis (MS), Alzheimer's disease (AD), and gastrointestinal disorders, including irritable bowel syndrome (IBS) and inflammatory bowel disease (IBD), which are characterized by chronic inflammation and dysregulation of the enteric nervous system (ENS) in the digestive tract. STING plays a crucial role in the pathway that induces the production of interferon in response to viral infection in the central nervous system (CNS). A new study by Dharshika et al. in the current issue of Neurogastroenterology and Motility has demonstrated distinct roles for STING in enteric neurons and glia, namely activation of STING leads to IFN-β production in enteric neurons but not in glia and reducing STING activation in enteric glia does not modulate the severity of Dextran sulfate sodium (DSS) colitis or subsequent loss of enteric neurons. Rather, the role of STING in enteric glia is related to enhancing autophagy. STING can influence gastrointestinal motility and barrier function and therefore be involved in the pathophysiology of IBS and IBD. This mini review highlights the current knowledge of STING in the pathophysiology of CNS and gastrointestinal diseases as well as these newly uncovered roles STING in enteric neurons and glia.
CONFLICT OF INTEREST STATEMENT
The authors have no conflicts of interest.
REFERENCES
- 1Barber GN. STING: infection, inflammation and cancer. Nat Rev Immunol. 2015; 15(12): 760-770.
- 2Kennedy RB, Haralambieva IH, Ovsyannikova IG, et al. Polymorphisms in STING affect human innate immune responses to poxviruses. Front Immunol. 2020; 11:567348.
- 3Teijaro JR. Type I interferons in viral control and immune regulation. Curr Opin Virol. 2016; 16: 31-40.
- 4Morere J, Hognon C, Miclot T, et al. How fragile we are: influence of stimulator of interferon genes (STING) variants on pathogen recognition and immune response efficiency. J Chem Inf Model. 2022; 62(12): 3096-3106.
- 5Banete A, Seaver K, Bakshi D, Gee K, Basta S. On taking the STING out of immune activation. J Leukoc Biol. 2018; 103(6): 1189-1195.
- 6Sun L, Wu J, Du F, Chen X, Chen ZJ. Cyclic GMP-AMP synthase is a cytosolic DNA sensor that activates the type I interferon pathway. Science. 2013; 339(6121): 786-791.
- 7Smith JA. STING, the endoplasmic reticulum, and mitochondria: is three a crowd or a conversation? Front Immunol. 2020; 11:611347.
- 8Li Z, Cai S, Sun Y, Li L, Ding S, Wang X. When STING Meets Viruses: Sensing, Trafficking and Response. Front Immunol. 2020; 11: 2064.
- 9Mao Y, Luo W, Zhang L, et al. STING–IRF3 triggers endothelial inflammation in response to free fatty acid-induced mitochondrial damage in diet-induced obesity. Arterioscler Thromb Vasc Biol. 2017; 37(5): 920-929.
- 10Li T, Chen ZJ. The cGAS–cGAMP–STING pathway connects DNA damage to inflammation, senescence, and cancer. J Exp Med. 2018; 215(5): 1287-1299.
- 11Zhu Y, An X, Zhang X, Qiao Y, Zheng T, Li X. STING: a master regulator in the cancer-immunity cycle. Mol Cancer. 2019; 18(1): 152.
- 12Wu Y-t, Fang Y, Wei Q, et al. Tumor-targeted delivery of a STING agonist improves cancer immunotherapy. Proc Natl Acad Sci USA. 2022; 119(49):e2214278119.
- 13Zhu Q, Hu H, Liu H, Shen H, Yan Z, Gao L. A synthetic STING agonist inhibits the replication of human parainfluenza virus 3 and rhinovirus 16 through distinct mechanisms. Antiviral Res. 2020; 183:104933.
- 14Dharshika C, Gonzales J, Chow A, Morales-Soto W, Gulbransen BD. Stimulator of interferon genes (STING) expression in the enteric nervous system and contributions of glial STING in disease. Neurogastroenterol Motil. 2023:e14553.
- 15Henn RE, Noureldein MH, Elzinga SE, Kim B, Savelieff MG, Feldman EL. Glial-neuron crosstalk in health and disease: a focus on metabolism, obesity, and cognitive impairment. Neurobiol Dis. 2022; 170:105766.
- 16Wanrooy BJ, Kumar KP, Wen SW, Qin CX, Ritchie RH, Wong CH. Distinct contributions of hyperglycemia and high-fat feeding in metabolic syndrome-induced neuroinflammation. J Neuroinflammation. 2018; 15: 1-13.
- 17Söderbom G, Zeng B-Y. The NLRP3 inflammasome as a bridge between neuro-inflammation in metabolic and neurodegenerative diseases. Int Rev Neurobiol. 2020; 154: 345-391.
- 18Elzinga SE, Henn R, Murdock BJ, et al. cGAS/STING and innate brain inflammation following acute high-fat feeding. Front Immunol. 2022; 13: 1012594.
- 19Bai J, Cervantes C, Liu J, et al. DsbA-L prevents obesity-induced inflammation and insulin resistance by suppressing the mtDNA release-activated cGAS-cGAMP-STING pathway. Proc Natl Acad Sci U S A. 2017; 114(46): 12196-12201.
- 20Bai J, Liu F. The cGAS-cGAMP-STING pathway: a molecular link between immunity and metabolism. Diabetes. 2019; 68(6): 1099-1108.
- 21Hinkle JT, Patel J, Panicker N, et al. STING mediates neurodegeneration and neuroinflammation in nigrostriatal α-synucleinopathy. Proc Natl Acad Sci. 2022; 119(15):e2118819119.
- 22Szego EM, Malz L, Bernhardt N, Rösen-Wolff A, Falkenburger BH, Luksch H. Constitutively active STING causes neuroinflammation and degeneration of dopaminergic neurons in mice. Elife. 2022; 11:e81943.
- 23Ke X, Hu T, Jiang M. cGAS-STING signaling pathway in gastrointestinal inflammatory disease and cancers. FASEB J. 2022; 36(1):e22029.
- 24Hu X, Zhang H, Zhang Q, Yao X, Ni W, Zhou K. Emerging role of STING signalling in CNS injury: inflammation, autophagy, necroptosis, ferroptosis and pyroptosis. J Neuroinflammation. 2022; 19(1): 242.
- 25Deng Y, Yi X, Gong Y, et al. Palmitic acid induces nDNA release to cytosol and promotes microglial M1 polarization via cGAS-STING signaling pathway. Biochimica et Biophysica Acta (BBA)–molecular. Cell Res. 2023; 1870(1):119385.
- 26Yuan T, Dong L, Pearsall EA, Zhou K, Cheng R, Ma JX. The protective role of microglial PPARα in diabetic retinal neurodegeneration and neurovascular dysfunction. Cell. 2022; 11(23):3869.
- 27Leonoudakis D, Rane A, Angeli S, Lithgow GJ, Andersen JK, Chinta SJ. Anti-inflammatory and neuroprotective role of natural product securinine in activated glial cells: implications for Parkinson's disease. Mediators Inflamm. 2017; 2017: 1-11.
- 28Iwata-Ichikawa E, Kondo Y, Miyazaki I, Asanuma M, Ogawa N. Glial cells protect neurons against oxidative stress via transcriptional up-regulation of the glutathione synthesis. J Neurochem. 1999; 72(6): 2334-2344.
- 29Cheng H, Wu JP, Tzeng SF. Neuroprotection of glial cell line-derived neurotrophic factor in damaged spinal cords following contusive injury. J Neurosci Res. 2002; 69(3): 397-405.
- 30Abdo H, Derkinderen P, Gomes P, et al. Enteric glial cells protect neurons from oxidative stress in part via reduced glutathione. FASEB J. 2010; 24(4): 1082-1094.
- 31Ramesh G, MacLean AG, Philipp MT. Cytokines and chemokines at the crossroads of Neuroinflammation, neurodegeneration, and neuropathic pain. Mediators Inflamm. 2013; 2013:480739.
- 32Hu Q, Ren H, Li G, et al. STING-mediated intestinal barrier dysfunction contributes to lethal sepsis. EBioMedicine. 2019; 41: 497-508.
- 33Canesso M, Lemos L, Neves T, et al. The cytosolic sensor STING is required for intestinal homeostasis and control of inflammation. Mucosal Immunol. 2018; 11(3): 820-834.
- 34Wan D, Jiang W, Hao J. Research advances in how the cGAS-STING pathway controls the cellular inflammatory response. Front Immunol. 2020; 11: 615.
- 35Howell KJ, Kraiczy J, Nayak KM, et al. DNA methylation and transcription patterns in intestinal epithelial cells from pediatric patients with inflammatory bowel diseases differentiate disease subtypes and associate with outcome. Gastroenterology. 2018; 154(3): 585-598.
- 36Chow AK, Gulbransen BD. Potential roles of enteric glia in bridging neuroimmune communication in the gut. Am J Physiol Gastrointest Liver Physiol. 2017; 312(2): G145-G152.
- 37Hu Q, Zhou Q, Xia X, et al. Cytosolic sensor STING in mucosal immunity: a master regulator of gut inflammation and carcinogenesis. J Exp Clin Cancer Res. 2021; 40(1): 39.
- 38Chen C, Zhang Y, Tao M, et al. Atrial natriuretic peptide attenuates colitis via inhibition of the cGAS-STING pathway in colonic epithelial cells. Int J Biol Sci. 2022; 18(4): 1737-1754.
- 39Fischer JC, Bscheider M, Eisenkolb G, et al. RIG-I/MAVS and STING signaling promote gut integrity during irradiation-and immune-mediated tissue injury. Sci Transl Med. 2017; 9:eaag2513.
- 40Mayer L. Mucosal immunity. Immunol Rev. 2005; 206: 5.
- 41Rakoff-Nahoum S, Paglino J, Eslami-Varzaneh F, Edberg S, Medzhitov R. Recognition of commensal microflora by toll-like receptors is required for intestinal homeostasis. Cell. 2004; 118(2): 229-241.
- 42Sauer J-D, Sotelo-Troha K, Von Moltke J, et al. The N-ethyl-N-nitrosourea-induced Goldenticket mouse mutant reveals an essential function of Sting in the in vivo interferon response to listeria monocytogenes and cyclic dinucleotides. Infect Immun. 2011; 79(2): 688-694.
- 43Ahn J, Son S, Oliveira SC, Barber GN. STING-dependent signaling underlies IL-10 controlled inflammatory colitis. Cell Rep. 2017; 21(13): 3873-3884.
- 44Yang W, Yu T, Zhou G, et al. Intrinsic STING switches off Pathogenetic programs of Th1 cells to inhibit colitis. Cell Mol Gastroenterol Hepatol. 2023; 15(5): 1161-1117.
- 45Song S, Peng P, Tang Z, et al. Decreased expression of STING predicts poor prognosis in patients with gastric cancer. Sci Rep. 2017; 7(1):39858.
- 46Xia T, Konno H, Ahn J, Barber GN. Deregulation of STING signaling in colorectal carcinoma constrains DNA damage responses and correlates with tumorigenesis. Cell Rep. 2016; 14(2): 282-297.
- 47Zhu Q, Man SM, Gurung P, et al. Cutting edge: STING mediates protection against colorectal tumorigenesis by governing the magnitude of intestinal inflammation. J Immunol. 2014; 193(10): 4779-4782.
- 48Chen S-Y, Chen S, Feng W, Li Z, Luo Y, Zhu X. A STING-related prognostic score predicts high-risk patients of colorectal cancer and provides insights into immunotherapy. Ann Transl Med. 2020; 9(1): 14.
- 49Luo X, Li H, Ma L, et al. Expression of STING is increased in liver tissues from patients with NAFLD and promotes macrophage-mediated hepatic inflammation and fibrosis in mice. Gastroenterology. 2018; 155(6): 1971-1984.e4.
- 50Yu Y, Liu Y, An W, Song J, Zhang Y, Zhao X. STING-mediated inflammation in Kupffer cells contributes to progression of nonalcoholic steatohepatitis. J Clin Invest. 2019; 129(2): 546-555.
- 51Wang X, Rao H, Zhao J, et al. STING expression in monocyte-derived macrophages is associated with the progression of liver inflammation and fibrosis in patients with nonalcoholic fatty liver disease. Lab Invest. 2020; 100(4): 542-552.
- 52Lv J, Xing C, Chen Y, et al. The STING in non-alcoholic fatty liver diseases: potential therapeutic targets in inflammation-carcinogenesis pathway. Pharmaceuticals (Basel). 2022; 15(10): 1241.
- 53Furness JB. The enteric nervous system and neurogastroenterology. Nat Rev Gastroenterol Hepatol. 2012; 9(5): 286-294.
- 54Gershon MD. Nerves, reflexes, and the enteric nervous system: pathogenesis of the irritable bowel syndrome. J Clin Gastroenterol. 2005; 39(5): S184-S193.
- 55Furness JB. The Enteric Nervous System. John Wiley & Sons; 2008.
- 56Kabouridis PS, Lasrado R, McCallum S, et al. Microbiota controls the homeostasis of glial cells in the gut lamina propria. Neuron. 2015; 85(2): 289-295.
- 57Kabouridis PS, Lasrado R, McCallum S, et al. The gut microbiota keeps enteric glial cells on the move; prospective roles of the gut epithelium and immune system. Gut Microbes. 2015; 6(6): 398-403.
- 58Seguella L, Gulbransen BD. Enteric glial biology, intercellular signalling and roles in gastrointestinal disease. Nat Rev Gastroenterol Hepatol. 2021; 18(8): 571-587.
- 59Carabotti M, Scirocco A, Maselli MA, Severi C. The gut-brain axis: interactions between enteric microbiota, central and enteric nervous systems. Ann Gastroenterol. 2015; 28(2): 203-209.