sting pathway
Recently Published Documents


TOTAL DOCUMENTS

382
(FIVE YEARS 316)

H-INDEX

29
(FIVE YEARS 14)

2022 ◽  
Vol 12 ◽  
Author(s):  
Chen Liu ◽  
Yuhan Huang ◽  
Yaoyuan Cui ◽  
Jun Zhou ◽  
Xu Qin ◽  
...  

BackgroundOvarian cancer (OC) is one of the most lethal gynecologic cancers. Growing evidence has proven that CDK4/6 plays a key role in tumor immunity and the prognosis of many cancers. However, the expression and function of CDK4/6 in OC remain unclear. Therefore, we aimed to explore the influence of CDK4/6 in OC, especially on immunity.MethodsWe analyzed CDK4/6 expression and prognosis using The Cancer Genome Atlas (TCGA), Gene Expression Omnibus (GEO) and Genotype Tissue Expression (GTEx) data. Subsequently, we used the cytoHubba plug-in of Cytoscape software and starBase to identify the noncoding RNAs (ncRNAs) regulating CDK4/6. Finally, we verified the effect of CDK4/6 on immunity in OC cell lines and animal models.ResultsCDK4/6 expression was higher in OC tissues than in normal ovarian tissues, and the high expression levels of CDK4/6 contributed to the immunosuppressive state of OC and were thus related to the poor prognosis of OC patients. This was also in general agreement with the results of OC cell line and animal experiments. Mechanistically, the CDK4/6 inhibitor palbociclib increased the secretion of interferon (IFN)-γ and the interferon-stimulated gene (ISG) response, thereby upregulating the expression of antigen-presenting molecules; this effect was partly dependent on the STING pathway and thus activated immunity in OC. Additionally, according to public data, the LRRC75A-AS1-hsa-miR-330-5p axis could inhibit the immune response of OC patients by upregulating CDK4/6, leading to a poor prognosis.ConclusionCDK4/6 affects the immune microenvironment of OC and correlates with the prognosis of OC patients.


2022 ◽  
Vol 12 ◽  
Author(s):  
Nanxin Liu ◽  
Xiaoxiao Pang ◽  
Hua Zhang ◽  
Ping Ji

Cyclic guanosine monophosphate (GMP)-adenosine monophosphate (AMP) (cGAMP) synthase (cGAS), along with the adaptor stimulator of interferon genes (STING), are crucial components of the innate immune system, and their study has become a research hotspot in recent years. Many biochemical and structural studies that have collectively elucidated the mechanism of activation of the cGAS-STING pathway with atomic resolution have provided insights into the roles of the cGAS-STING pathway in innate immunity and clues to the origin and evolution of the modern cGAS-STING signaling pathway. The cGAS-STING pathway has been identified to protect the host against viral infection. After detecting viral dsDNA, cGAS synthesizes a second messenger to activate STING, eliciting antiviral immune responses by promoting the expression of interferons (IFNs) and hundreds of IFN-stimulated genes (ISGs). Recently, the cGAS-STING pathway has also been found to be involved in response to bacterial infections, including bacterial pneumonia, melioidosis, tuberculosis, and sepsis. However, compared with its functions in viral infection, the cGAS-STING signaling pathway in bacterial infection is more complex and diverse since the protective and detrimental effects of type I IFN (IFN-I) on the host depend on the bacterial species and infection mode. Besides, STING activation can also affect infection prognosis through other mechanisms in different bacterial infections, independent of the IFN-I response. Interestingly, the core protein components of the mammalian cGAS-STING signaling pathway have been found in the bacterial defense system, suggesting that this widespread signaling pathway may have originated in bacteria. Here, we review recent findings related to the structures of major molecules involved in the cGAS-STING pathway and the effects of the cGAS-STING pathway in various bacterial infections and bacterial immunity, which may pave the way for the development of new antibacterial drugs that specifically kill bacteria without harmful effects on the host.


Author(s):  
Sumaiah S. Al-Asmari ◽  
Aleksandra Rajapakse ◽  
Tomalika R. Ullah ◽  
Geneviève Pépin ◽  
Laura V. Croft ◽  
...  

Activation of the STING pathway upon genotoxic treatment of cancer cells has been shown to lead to anti-tumoral effects, mediated through the acute production of interferon (IFN)-β. Conversely, the pathway also correlates with the expression of NF-κB-driven pro-tumorigenic genes, but these associations are only poorly defined in the context of genotoxic treatment, and are thought to correlate with a chronic engagement of the pathway. We demonstrate here that half of the STING-expressing cancer cells from the NCI60 panel rapidly increased expression of pro-tumorigenic IL-6 upon genotoxic DNA damage, often independent of type-I IFN responses. While preferentially dependent on canonical STING, we demonstrate that genotoxic DNA damage induced by camptothecin (CPT) also drove IL-6 production through non-canonical STING signaling in selected cancer cells. Consequently, pharmacological inhibition of canonical STING failed to broadly inhibit IL-6 production induced by CPT, although this could be achieved through downstream ERK1/2 inhibition. Finally, prolonged inhibition of canonical STING signaling was associated with increased colony formation of MG-63 cells, highlighting the duality of STING signaling in also restraining the growth of selected cancer cells. Collectively, our findings demonstrate that genotoxic-induced DNA damage frequently leads to the rapid production of pro-tumorigenic IL-6 in cancer cells, independent of an IFN signature, through canonical and non-canonical STING activation; this underlines the complexity of STING engagement in human cancer cells, with frequent acute pro-tumorigenic activities induced by DNA damage. We propose that inhibition of ERK1/2 may help curb such pro-tumorigenic responses to DNA-damage, while preserving the anti-proliferative effects of the STING-interferon axis.


2022 ◽  
Vol 20 (1) ◽  
Author(s):  
Meixiao Zhan ◽  
Xiangrong Yu ◽  
Wei Zhao ◽  
Yongjun Peng ◽  
Shaojun Peng ◽  
...  

AbstractRegulation of stimulator of interferon genes (STING) pathway using agonists can boost antitumor immunity for cancer treatment, while the rapid plasma clearance, limited membrane permeability, and inefficient cytosolic transport of STING agonists greatly compromise their therapeutic efficacy. In this study, we describe an extracellular matrix (ECM)-degrading nanoagonist (dNAc) with second near-infrared (NIR-II) light controlled activation of intracellular STING pathway for mild photothermal-augmented chemodynamic-immunotherapy of breast cancer. The dNAc consists of a thermal-responsive liposome inside loading with ferrous sulfide (FeS2) nanoparticles as both NIR-II photothermal converters and Fenton catalysts, 2′3′-cyclic guanosine monophosphate-adenosine monophosphate (cGAMP) as the STING agonist, and an ECM-degrading enzyme (bromelain) on the liposome surface. Mild heat generated by dNAc upon NIR-II photoirradiation improves Fenton reaction efficacy to kill tumor cells and cause immunogenic cell death (ICD). Meanwhile, the generated heat triggers a controlled release of cGAMP from thermal-responsive liposomes to active STING pathway. The mild photothermal activation of STING pathway combined with ICD promotes anti-tumor immune responses, which leads to improved infiltration of effector T cells into tumor tissues after bromelain-mediated ECM degradation. As a result, after treatment with dNAc upon NIR-II photoactivation, both primary and distant tumors in a murine mouse model are inhibited and the liver and lung metastasis are effectively suppressed. This work presents a photoactivatable system for STING pathway and combinational immunotherapy with improved therapeutic outcome. Graphical Abstract


2022 ◽  
Author(s):  
Xiaodan Sun ◽  
Peiyan Zhao ◽  
Hui Li ◽  
Yan liu ◽  
Ying Cheng

Abstract Background: KRAS/KEAP1 (KK) co-mutant lung adenocarcinoma (LUAD) exhibited poor response to immune checkpoint inhibitors (ICI) via shaping a suppressive tumor immune microenvironment, the mechanism remains to be elucidated. Methods: The mRNA and protein expression of target molecules were analyzed by qRT-PCR and Western blot, respectively. The subcellular location of NRF2 was observed by immunofluorescence staining, and nuclear and cytoplasm isolation. After exogenous over-expression and knockdown of NRF2 and the addition of a STING pathway inhibitor in tumor cells, the effects on the CD8+ T cell recruitment was detected using chemotaxis assay, and the secretion of chemokines CCL5 and CXCL10 was analyzed by ELISA. The potential NRF2 target BRCA1 was identified using bioinformatic approaches and verified by a dual luciferase reporter assay. Results: NRF2, the target of KEAP1, was overexpressed and activated in KK type cells. NRF2 effected as a negative regulator of CD8+ T cells recruitment by decreasing CCL5 and CXCL10 chemokines in KK type LUAD. Mechanistically, NRF2 promoted the transcription and expression of BRCA1 to repair DNA damage, resulting in STING pathway inactivation. Conclusion: The combination of NRF2 inhibitor or STING agonist with ICI may be a promising therapeutic approach for patients with KK type LUAD.


2022 ◽  
pp. 353-373
Author(s):  
Ting Su ◽  
Nadia Tasnim Ahmed ◽  
Shurong Zhou ◽  
Xiang Liu ◽  
Guizhi Zhu

2022 ◽  
Vol 63 (1) ◽  
pp. 42
Author(s):  
Chun-Bong Synn ◽  
Dong Kwon Kim ◽  
Jae Hwan Kim ◽  
Youngseon Byeon ◽  
Young Seob Kim ◽  
...  

2021 ◽  
pp. 2105783
Author(s):  
Jie Yan ◽  
Guohao Wang ◽  
Lisi Xie ◽  
Hao Tian ◽  
Jie Li ◽  
...  

Sign in / Sign up

Export Citation Format

Share Document