1
|
Nie AY, Xiao ZH, Deng JL, Li N, Hao LY, Li SH, Hu XY. Bidirectional regulation of the cyclic guanosine monophosphate-adenosine monophosphate synthase-stimulator of interferon gene pathway and its impact on hepatocellular carcinoma. World J Gastrointest Oncol 2025; 17:98556. [DOI: 10.4251/wjgo.v17.i2.98556] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/29/2024] [Revised: 10/30/2024] [Accepted: 11/18/2024] [Indexed: 01/18/2025] Open
Abstract
BACKGROUND Hepatocellular carcinoma (HCC) ranks as the fourth leading cause of cancer-related deaths in China, and the treatment options are limited. The cyclic guanosine monophosphate-adenosine monophosphate synthase (cGAS) activates the stimulator of interferon gene (STING) signaling pathway as a crucial immune response pathway in the cytoplasm, which detects cytoplasmic DNA to regulate innate and adaptive immune responses. As a potential therapeutic target, cGAS-STING pathway markedly inhibits tumor cell proliferation and metastasis, with its activation being particularly relevant in HCC. However, prolonged pathway activation may lead to an immunosuppressive tumor microenvironment, which fostering the invasion or metastasis of liver tumor cells.
AIM To investigate the dual-regulation mechanism of cGAS-STING in HCC.
METHODS This review was conducted according to the PRISMA guidelines. The study conducted a comprehensive search for articles related to HCC on PubMed and Web of Science databases. Through rigorous screening and meticulous analysis of the retrieved literature, the research aimed to summarize and elucidate the impact of the cGAS-STING pathway on HCC tumors.
RESULTS All authors collaboratively selected studies for inclusion, extracted data, and the initial search of online databases yielded 1445 studies. After removing duplicates, the remaining 964 records were screened. Ultimately, 55 articles met the inclusion criteria and were included in this review.
CONCLUSION Acute inflammation can have a few inhibitory effects on cancer, while chronic inflammation generally promotes its progression. Extended cGAS-STING pathway activation will result in a suppressive tumor microenvironment.
Collapse
Affiliation(s)
- Ai-Yu Nie
- College of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu 610075, Sichuan Province, China
| | - Zhong-Hui Xiao
- College of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu 610075, Sichuan Province, China
| | - Jia-Li Deng
- College of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu 610075, Sichuan Province, China
| | - Na Li
- College of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu 610075, Sichuan Province, China
| | - Li-Yuan Hao
- College of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu 610075, Sichuan Province, China
| | - Sheng-Hao Li
- College of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu 610075, Sichuan Province, China
| | - Xiao-Yu Hu
- Department of Infection, Affiliated Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu 610072, Sichuan Province, China
| |
Collapse
|
2
|
Jin Y, Dong N, Shimizu S, Li Y, Yao Y, Qiao H, Liu X, Liu S, Guo C, Wang L. Hesperidin enhanced anti-breast cancer effect and alleviated cisplatin induced nephrotoxicity through silk fibroin delivery system. Toxicol Appl Pharmacol 2025; 495:117234. [PMID: 39832567 DOI: 10.1016/j.taap.2025.117234] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2024] [Revised: 01/15/2025] [Accepted: 01/15/2025] [Indexed: 01/22/2025]
Abstract
The incidence rate and mortality rate of breast cancer remain high, and there is an urgent need for safe and effective drugs. The excellent biological activity of hesperidin (HE) is a potential drug for the treatment of breast cancer. In this study, silk fibroin peptides (SFP) were used as delivery carriers and HE loaded SFP nanofibers (SFP/HE NFs) was prepared. The in vitro results showed that SFP/HE NFs significantly inhibited the proliferation and migration of breast cancer cell MDA-MB-231 compared with free HE. The mechanism results demonstrated that SFP/HE NFs induced apoptosis and DNA double stranded damage (DSBs) and further activated the cyclic monophosphate guanosine adenosine monophosphate synthase- stimulator of interferon gene (cGAS-STING) pathway. The in vivo studies showed that SFP/HE NFs treatment significantly inhibited the growth of breast cancer, with an inhibition rate of 65.9 % (100 mg/kg). In vivo mechanism studies also demonstrated that the anti-tumor activity of SFP/HE NFs was related to the activation of the cGAS-STING pathway. Interestingly, we found that the combination of SFP/HE NFs and cisplatin not only enhanced the anti-tumor activity of cisplatin, but also alleviated cisplatin induced nephrotoxicity. In conclusion, our results demonstrate the benefits of activating the cGAS-STING pathway in the treatment of breast cancer, which is expected to provide potential candidates for combined treatment of breast cancer.
Collapse
Affiliation(s)
- Yonglong Jin
- Department of Radiotherapy, The Affiliated Hospital of Qingdao University, Qingdao 266000, China; School of Public Health, Qingdao University, Qingdao 266071, China
| | - Nina Dong
- Department of Radiotherapy, The Affiliated Hospital of Qingdao University, Qingdao 266000, China
| | - Shosei Shimizu
- Department of Radiotherapy, Yizhou Tumor Hospital, Zhuozhou 072750, China; Department of Radiotherapy, University of Tsukuba Hospital, Tsukuba, Japan
| | - Yinuo Li
- Department of Radiotherapy, University of Tsukuba Hospital, Tsukuba, Japan
| | - Yuan Yao
- Graduate School of Environmental Science, Hokkaido University, Sapporo, Hokkaido, Japan
| | - Hong Qiao
- Hauolilly-MEDICAL company, Tokyo, Japan
| | - Xiguang Liu
- Department of Radiotherapy, The Affiliated Hospital of Qingdao University, Qingdao 266000, China
| | - Shuai Liu
- Qingdao University of Science and Technology, Qingdao 266041, China
| | - Chuanlong Guo
- Qingdao University of Science and Technology, Qingdao 266041, China
| | - Lijie Wang
- Department of Radiotherapy, The Affiliated Hospital of Qingdao University, Qingdao 266000, China.
| |
Collapse
|
3
|
Gong Y, Cheng Y, Zeng F, Liu X, Yang Y, Zhang F, Wen C, Yang F, Li H, He Y, Ni B, Xu Y, Xiao L, Zhang Q, Zhou L, Zheng J, Chen W. A self-gelling hemostatic powder boosting radiotherapy-elicited NK cell immunity to combat postoperative hepatocellular carcinoma relapse. Biomaterials 2024; 317:123068. [PMID: 39813968 DOI: 10.1016/j.biomaterials.2024.123068] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2024] [Revised: 12/06/2024] [Accepted: 12/27/2024] [Indexed: 01/18/2025]
Abstract
Liver resection represents a main curative treatment for patients with early-stage hepatocellular carcinoma (HCC), but there is a rather high incidence of postoperative HCC relapse, which severely shortens long-term survival time. Currently, no standard adjuvant strategies are available for preventing HCC relapse in clinical practice. Impaired natural killer (NK) cell anti-tumor immunity has been disclosed as a crucial root of HCC relapse, indicating that reinstating NK cell anti-tumor immunity may show promise to curb HCC relapse. Coincidently, mounting evidence shows that radiotherapy (RT) can trigger NK cell anti-tumor immunity, though its mechanisms have never been completely elucidated. Herein, we uncover that RT can induce immunogenic cell death and activate cGAS-STING pathway in HCC cells to elicit NK cell anti-tumor immunity. However, RT is also revealed to enhance autophagy and CD73 expression in HCC cells, as well as neutrophil extracellular traps (NETs) formation, which largely limits RT-induced activation of NK cell anti-tumor immunity. Therefore, a cocktail of autophagy inhibitor 3-methyladenine, CD73 inhibitor ARL 67156 trisodium and NETs lyase DNase I may sensitize RT to reinvigorate NK cell anti-tumor immunity and thus prevent HCC relapse postresection. To minimize therapy-related side effects, a nanocomposite powder encapsulating such a triple-drug cocktail is developed. This powder can rapidly form adhesive hydrogel in situ after applied to surgical margin, consequently fulfilling liver-localized sustained drug delivery. Importantly, it can sensitize RT to reinstate NK cell anti-tumor immunity to combat postoperative HCC relapse in Heap1-6-HCC murine model. Besides, this powder can also generate rapid hemostasis in rat and porcine models. Altogether, this work provides an innovative strategy to thwart postoperative HCC relapse and bleeding.
Collapse
Affiliation(s)
- Yihang Gong
- Department of Hepatic Surgery and Liver Transplantation Center, The Third Affiliated Hospital of Sun Yat-sen University, Organ Transplantation Institute, Sun Yat-sen University, Organ Transplantation Research Center of Guangdong Province, Guangdong Province Engineering Laboratory for Transplantation Medicine, Guangzhou, 510630, China; Guangdong Key Laboratory of Liver Disease Research, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510630, China
| | - Yusheng Cheng
- Department of General Surgery, The Second Clinical Medical College, Lanzhou University, Lanzhou, 730030, China; The Second Clinical Medical College, Lanzhou University, Lanzhou, 730030, China
| | - Fanxin Zeng
- Department of Hepatic Surgery and Liver Transplantation Center, The Third Affiliated Hospital of Sun Yat-sen University, Organ Transplantation Institute, Sun Yat-sen University, Organ Transplantation Research Center of Guangdong Province, Guangdong Province Engineering Laboratory for Transplantation Medicine, Guangzhou, 510630, China; Guangdong Key Laboratory of Liver Disease Research, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510630, China
| | - Xiaoquan Liu
- Department of Infectious Diseases, Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, 510630, China
| | - Yang Yang
- Department of Hepatic Surgery and Liver Transplantation Center, The Third Affiliated Hospital of Sun Yat-sen University, Organ Transplantation Institute, Sun Yat-sen University, Organ Transplantation Research Center of Guangdong Province, Guangdong Province Engineering Laboratory for Transplantation Medicine, Guangzhou, 510630, China; Guangdong Key Laboratory of Liver Disease Research, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510630, China
| | - Feng Zhang
- Department of Hepatic Surgery and Liver Transplantation Center, The Third Affiliated Hospital of Sun Yat-sen University, Organ Transplantation Institute, Sun Yat-sen University, Organ Transplantation Research Center of Guangdong Province, Guangdong Province Engineering Laboratory for Transplantation Medicine, Guangzhou, 510630, China; Biotherapy Centre & Cell-gene Therapy Translational Medicine Research Centre, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510630, China
| | - Chaoyao Wen
- School of Chemical Engineering and Light Industry, Guangdong University of Technology, Guangzhou, 510006, China
| | - Fan Yang
- Biotherapy Centre & Cell-gene Therapy Translational Medicine Research Centre, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510630, China
| | - Hua Li
- Department of Hepatic Surgery and Liver Transplantation Center, The Third Affiliated Hospital of Sun Yat-sen University, Organ Transplantation Institute, Sun Yat-sen University, Organ Transplantation Research Center of Guangdong Province, Guangdong Province Engineering Laboratory for Transplantation Medicine, Guangzhou, 510630, China; Guangdong Key Laboratory of Liver Disease Research, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510630, China
| | - Yizhan He
- Biotherapy Centre & Cell-gene Therapy Translational Medicine Research Centre, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510630, China
| | - Beibei Ni
- Biotherapy Centre & Cell-gene Therapy Translational Medicine Research Centre, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510630, China
| | - Yan Xu
- Department of Hepatic Surgery and Liver Transplantation Center, The Third Affiliated Hospital of Sun Yat-sen University, Organ Transplantation Institute, Sun Yat-sen University, Organ Transplantation Research Center of Guangdong Province, Guangdong Province Engineering Laboratory for Transplantation Medicine, Guangzhou, 510630, China; Biotherapy Centre & Cell-gene Therapy Translational Medicine Research Centre, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510630, China
| | - Lan Xiao
- Department of Gynecology Oncology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230026, Anhui, China
| | - Qi Zhang
- Department of Hepatic Surgery and Liver Transplantation Center, The Third Affiliated Hospital of Sun Yat-sen University, Organ Transplantation Institute, Sun Yat-sen University, Organ Transplantation Research Center of Guangdong Province, Guangdong Province Engineering Laboratory for Transplantation Medicine, Guangzhou, 510630, China; Biotherapy Centre & Cell-gene Therapy Translational Medicine Research Centre, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510630, China.
| | - Lei Zhou
- Guangzhou Key Laboratory of Spine Disease Prevention and Treatment, Department of Orthopaedic Surgery, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510150, China.
| | - Jun Zheng
- Department of Hepatic Surgery and Liver Transplantation Center, The Third Affiliated Hospital of Sun Yat-sen University, Organ Transplantation Institute, Sun Yat-sen University, Organ Transplantation Research Center of Guangdong Province, Guangdong Province Engineering Laboratory for Transplantation Medicine, Guangzhou, 510630, China; Guangdong Key Laboratory of Liver Disease Research, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510630, China.
| | - Wenjie Chen
- Department of Hepatic Surgery and Liver Transplantation Center, The Third Affiliated Hospital of Sun Yat-sen University, Organ Transplantation Institute, Sun Yat-sen University, Organ Transplantation Research Center of Guangdong Province, Guangdong Province Engineering Laboratory for Transplantation Medicine, Guangzhou, 510630, China; Biotherapy Centre & Cell-gene Therapy Translational Medicine Research Centre, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510630, China.
| |
Collapse
|
4
|
Yin Y, Ma Z, Yuan S, Xu K, Wang X. OPA3 inhibits the cGAS-STING pathway mediated by mtDNA stress to promote colorectal cancer progression. In Vitro Cell Dev Biol Anim 2024:10.1007/s11626-024-01000-3. [PMID: 39725842 DOI: 10.1007/s11626-024-01000-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2024] [Accepted: 11/23/2024] [Indexed: 12/28/2024]
Abstract
Colorectal cancer (CRC) is an extremely harmful malignant tumor. Optic atrophy 3 (OPA3) is highly expressed in multiple tumors, but its action in CRC is still unknown. This research aims to explore the role of OPA3 and its related molecular mechanisms for CRC. Firstly, we overexpressed and knocked down OPA3 to examine its effect on CRC cell (HT29 cell) growth. CRC cell viability, migration, invasion, and levels of proliferation markers and cell cycle-associated proteins were measured. Then, we treated cells with carbonyl cyanide m-chlorophenyl hydrazone (CCCP) to explore mitochondrial dysfunction and mtDNA stress in HT29 cells. Next, we overexpressed cGAS and STING to examine their correlation with OPA3. The results showed that OPA3 overexpression enhanced CRC cell viability, migration, invasion, and the levels of PCNA, Cyclin A2, and Cyclin B1. Knockdown of OPA3 had the opposite effects. Moreover, OPA3 knockdown facilitated mitochondrial dysfunction and mtDNA stress in CRC cells. OPA3 overexpression also inhibited CCCP-induced mitochondrial stress disorder. Additionally, OPA3 knockdown elevated the protein levels of p-STING and cGAS and the mRNA level of STING target genes. Furthermore, overexpression of either cGAS or STING partially alleviated the enhancement of HT29 cell proliferation, migration, and invasion mediated by OPA3 overexpression. In conclusion, OPA3 promotes CRC progression via inhibiting the cGAS-STING pathway, which is mediated by mtDNA stress. OPA3 may be a new potential target for CRC.
Collapse
Affiliation(s)
- Yuqiang Yin
- Department of General Surgery, The First People's Hospital of Pingjiang County, Yueyang, 410400, China
| | - Zhenxin Ma
- Department of General Surgery, The First People's Hospital of Pingjiang County, Yueyang, 410400, China
| | - Siwen Yuan
- Department of General Surgery, The First People's Hospital of Pingjiang County, Yueyang, 410400, China
| | - Kangfeng Xu
- Department of General Surgery, The First People's Hospital of Pingjiang County, Yueyang, 410400, China
| | - Xiaofeng Wang
- The First People's Hospital of Pingjiang County, Yueyang, 410400, China.
| |
Collapse
|
5
|
Yang C, Xu X, Wu M, Zhao Z, Feng Y, Liang W, Xu C, Jiang T, Zhang G. Huang-Jin-Shuang-Shen Decoction promotes CD8+ T-cell-mediated anti-tumor immunity by regulating chemokine CXCL10 in gastric cancer. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 135:156065. [PMID: 39341128 DOI: 10.1016/j.phymed.2024.156065] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Revised: 08/20/2024] [Accepted: 09/15/2024] [Indexed: 09/30/2024]
Abstract
BACKGROUND Research on immunotherapy for gastric cancer is currently receiving significant attention, with particular emphasis on the role of CD8+ T cells in anti-tumor immune responses. In recent years, the importance of the chemokine CXCL10 in promoting anti-tumor immunity has been increasingly recognized because it plays a crucial role in recruiting CD8+ T cells to the tumor microenvironment. The Huang-Jin-Shuang-Shen (HJSS) Decoction, a Chinese medicine, has been used as an adjuvant drug for gastric cancer chemotherapy. Its mechanism of action may be related to the activation of anti-tumor immunity. PURPOSE To assess the role of the HJSS Decoction in regulating the immune microenvironment of gastric cancer and elucidate its mechanism. STUDY DESIGN/METHODS Ultra-high performance liquid chromatography Q Exactive-mass spectrometry was used to analyze the main components of the HJSS Decoction and evaluate the therapeutic effect of the HJSS Decoction synergized with 5-fluorouracil (5-FU) on gastric cancer. The proportions of CD8+ T cells and killing markers were determined using flow cytometry. Mechanisms of action and targets were screened using network pharmacology. The level of CXCL10 was detected using enzyme-linked immunosorbent assay and western blot, and nuclear factor kappa-light-chain-enhancer of activated B cells (NFκB) related signaling pathway was detected in vitro. The target function was validated by siRNA transfection. RESULTS The combination of HJSS Decoction and 5-FU demonstrated a synergistic effect in impeding the progression of subcutaneous gastric cancer. This was achieved through the facilitation of apoptosis and suppression of proliferation. Furthermore, HJSS Decoction exhibited the ability to enhance the population of CD8+ T cells and augment their cytotoxic capabilities, both in laboratory settings and in living organisms. Notably, HJSS Decoction upregulated the expression of CXCL10, and mechanistically, it activated the NFκB-related signaling pathway to initiate subsequent transcription of chemokines. CONCLUSION The present study aimed to investigate the pharmacological mechanism of the HJSS Decoction and its potential clinical application in inhibiting gastric cancer in mice. HJSS Decoction can cooperate with 5-FU to inhibit gastric cancer, and the optimal dose is medium. HJSS Decoction exerts anti-tumor immunity by activating the NFκB-related signaling pathway and promoting the expression of CXCL10, which in turn recruits CD8+ T cells into the tumor immune microenvironment. Overall, these findings provide valuable evidence for the potential clinical application of HJSS Decoction.
Collapse
Affiliation(s)
- Chuqi Yang
- Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China; Zhejiang Key Laboratory of Blood-Stasis-Toxin Syndrome, Zhejiang Chinese Medical University, Hangzhou 310053, China; Suzhou Ninth Hospital affiliated to Soochow University (Suzhou Ninth People's Hospital), Suzhou 215200, China
| | - Xuefei Xu
- Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China; Zhejiang Key Laboratory of Blood-Stasis-Toxin Syndrome, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Mengting Wu
- Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China; Zhejiang Key Laboratory of Blood-Stasis-Toxin Syndrome, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Zhengqi Zhao
- Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China; Zhejiang Key Laboratory of Blood-Stasis-Toxin Syndrome, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Yewen Feng
- Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China; Zhejiang Key Laboratory of Blood-Stasis-Toxin Syndrome, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Weiyu Liang
- Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China; Zhejiang Key Laboratory of Blood-Stasis-Toxin Syndrome, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Chuyun Xu
- The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou 310053, China
| | - Tao Jiang
- Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China; Zhejiang Key Laboratory of Blood-Stasis-Toxin Syndrome, Zhejiang Chinese Medical University, Hangzhou 310053, China.
| | - Guangji Zhang
- Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China; Zhejiang Key Laboratory of Blood-Stasis-Toxin Syndrome, Zhejiang Chinese Medical University, Hangzhou 310053, China; Traditional Chinese Medicine "Preventing Disease" Wisdom Health Project Research Center of Zhejiang, Hangzhou 310053, China.
| |
Collapse
|
6
|
Wang R, Liu Q, You W, Wang H, Chen Y. A transformer-based deep learning survival prediction model and an explainable XGBoost anti-PD-1/PD-L1 outcome prediction model based on the cGAS-STING-centered pathways in hepatocellular carcinoma. Brief Bioinform 2024; 26:bbae686. [PMID: 39749665 PMCID: PMC11695900 DOI: 10.1093/bib/bbae686] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2024] [Revised: 11/13/2024] [Accepted: 12/27/2024] [Indexed: 01/04/2025] Open
Abstract
Recent studies suggest cGAS-STING pathway may play a crucial role in the genesis and development of hepatocellular carcinoma (HCC), closely associated with classical pathways and tumor immunity. We aimed to develop models predicting survival and anti-PD-1/PD-L1 outcomes centered on the cGAS-STING pathway in HCC. We identified classical pathways highly correlated with cGAS-STING pathway and constructed transformer survival model preserving raw structure of pathways. We also developed explainable XGBoost model for predicting anti-PD-1/PD-L1 outcomes using SHAP algorithm. We trained and validated transformer survival model on pan-cancer cohort and tested it on three independent HCC cohorts. Using 0.5 as threshold across cohorts, we divided each HCC cohort into two groups and calculated P values with log-rank test. TCGA-LIHC: C-index = 0.750, P = 1.52e-11; ICGC-LIRI-JP: C-index = 0.741, P = .00138; GSE144269: C-index = 0.647, P = .0233. We trained and validated [area under the receiver operating characteristic curve (AUC) = 0.777] XGBoost model on immunotherapy datasets and tested it on GSE78220 (AUC = 0.789); we also tested XGBoost model on HCC anti-PD-L1 cohort (AUC = 0.719). Our deep learning model and XGBoost model demonstrate potential in predicting survival risks and anti-PD-1/PD-L1 outcomes in HCC. We deployed these two prediction models to the GitHub repository and provided detailed instructions for their usage: deep learning survival model, https://github.com/mlwalker123/CSP_survival_model; XGBoost immunotherapy model, https://github.com/mlwalker123/CSP_immunotherapy_model.
Collapse
Affiliation(s)
- Ren Wang
- The Affiliated Wuxi People’s Hospital of Nanjing Medical University, Wuxi People’s Hospital, Wuxi Medical Center, Department of Immunology, School of Basic Medical Sciences, Nanjing Medical University, 101 Longmian Avenue, Nanjing 211166, Jiangsu Province, China
- The Affiliated Huai’an No. 1 People’s Hospital, Nanjing Medical University, West Road of the Yellow River, Huai’an 223300, Jiangsu Province, China
- Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, 101 Longmian Avenue, Nanjing 211166, Jiangsu Province, China
| | - Qiumei Liu
- The Affiliated Wuxi People’s Hospital of Nanjing Medical University, Wuxi People’s Hospital, Wuxi Medical Center, Department of Immunology, School of Basic Medical Sciences, Nanjing Medical University, 101 Longmian Avenue, Nanjing 211166, Jiangsu Province, China
- The Affiliated Huai’an No. 1 People’s Hospital, Nanjing Medical University, West Road of the Yellow River, Huai’an 223300, Jiangsu Province, China
- Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, 101 Longmian Avenue, Nanjing 211166, Jiangsu Province, China
| | - Wenhua You
- The Affiliated Wuxi People’s Hospital of Nanjing Medical University, Wuxi People’s Hospital, Wuxi Medical Center, Department of Immunology, School of Basic Medical Sciences, Nanjing Medical University, 101 Longmian Avenue, Nanjing 211166, Jiangsu Province, China
- The Affiliated Huai’an No. 1 People’s Hospital, Nanjing Medical University, West Road of the Yellow River, Huai’an 223300, Jiangsu Province, China
- Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, 101 Longmian Avenue, Nanjing 211166, Jiangsu Province, China
| | - Huiyu Wang
- The Affiliated Wuxi People’s Hospital of Nanjing Medical University, Wuxi People’s Hospital, Wuxi Medical Center, Department of Immunology, School of Basic Medical Sciences, Nanjing Medical University, 101 Longmian Avenue, Nanjing 211166, Jiangsu Province, China
| | - Yun Chen
- The Affiliated Wuxi People’s Hospital of Nanjing Medical University, Wuxi People’s Hospital, Wuxi Medical Center, Department of Immunology, School of Basic Medical Sciences, Nanjing Medical University, 101 Longmian Avenue, Nanjing 211166, Jiangsu Province, China
- The Affiliated Huai’an No. 1 People’s Hospital, Nanjing Medical University, West Road of the Yellow River, Huai’an 223300, Jiangsu Province, China
- Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, 101 Longmian Avenue, Nanjing 211166, Jiangsu Province, China
| |
Collapse
|
7
|
Zhong Y, Cao H, Li W, Deng J, Li D, Deng J. Investigating the impact of STING pathway activation on breast cancer treatment outcomes: development and validation of a prognostic model. Front Immunol 2024; 15:1438364. [PMID: 39185402 PMCID: PMC11341366 DOI: 10.3389/fimmu.2024.1438364] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2024] [Accepted: 07/09/2024] [Indexed: 08/27/2024] Open
Abstract
Introduction Breast cancer (BRCA) is a significant cause of cancer-associated mortality across the globe. Current therapeutic approaches face challenges such as drug resistance and metastasis. Immune signaling is triggered by chromosomal instability (CIN) generates misplaced DNA structures that activate the cyclic GMP-AMP synthase-stimulator of interferon genes (cGAS-STING) pathway, triggering. Studies have linked STING activation to BRCA treatment. Methods The bulk RNA-seq data for patients with BRCA were collected from the TCGA-BRCA cohort, GSE20685, and GSE96058 cohorts. STING pathway-related genes (SRGs) were obtained from the Reactome database. Differentially expressed genes were analyzed using the limma package. Immune cell infiltration was analyzed using the IOBR package. Gene Ontology biological processes, Kyoto Encyclopedia of Genes and Genomes pathways, and cancer hallmark pathways were analyzed using the MSigDB database. Prognostic models were prepared using the least absolute shrinkage and selection operator and multiple-factor Cox regression analysis. Single-cell analysis was performed using the Seurat and SCP pipeline. Results The expression patterns and clinical relevance of SRGs were analyzed in patients with BRCA. Transcriptional differences in the SRGs were observed between normal and tumorous tissues, with global down-regulated STING1 and up-regulated TBK1 in BRCA tissue. Tumor tissues were classified through consensus clustering analysis into two distinct groups, with differences in clinical characteristics and immune infiltration. A prognostic model related to the differences in STING pathway activity-high prognostic stratification potency-was developed and validated. Correlation analysis revealed suppressed overall immune activation in patients with BRCA having higher risk scores. Gemcitabine had a more favorable outcome in the low-risk group. The activity of the prognostic model at the single-cell level was confirmed through single-cell analysis, particularly in CD8 T cells and intratumor natural killer cells. Conclusion A STING pathway-related prognostic model developed and validated and the model could accurately predict BRCA patient outcomes. These findings have important implications for the personalized treatment and management of patients with BRCA.
Collapse
Affiliation(s)
- YangYan Zhong
- The Second Affiliated Hospital, Department of Breast and Thyroid Surgery, Hengyang Medical School, University of South China, Hengyang, Hunan, China
- Clinical Research Center for Breast and Thyroid Disease Prevention and Control in Hunan Province, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Hong Cao
- The Second Affiliated Hospital, Department of Breast and Thyroid Surgery, Hengyang Medical School, University of South China, Hengyang, Hunan, China
- Clinical Research Center for Breast and Thyroid Disease Prevention and Control in Hunan Province, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Wei Li
- The Second Affiliated Hospital, Department of Breast and Thyroid Surgery, Hengyang Medical School, University of South China, Hengyang, Hunan, China
- Clinical Research Center for Breast and Thyroid Disease Prevention and Control in Hunan Province, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Jian Deng
- The Second Affiliated Hospital, Department of Breast and Thyroid Surgery, Hengyang Medical School, University of South China, Hengyang, Hunan, China
- Clinical Research Center for Breast and Thyroid Disease Prevention and Control in Hunan Province, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Dan Li
- The Second Affiliated Hospital, Department of Breast and Thyroid Surgery, Hengyang Medical School, University of South China, Hengyang, Hunan, China
- Clinical Research Center for Breast and Thyroid Disease Prevention and Control in Hunan Province, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - JunJie Deng
- The Second Affiliated Hospital, Department of Breast and Thyroid Surgery, Hengyang Medical School, University of South China, Hengyang, Hunan, China
- Clinical Research Center for Breast and Thyroid Disease Prevention and Control in Hunan Province, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| |
Collapse
|
8
|
Hao Q, Dong R, Bai W, Chang D, Yao X, Zhang Y, Xu H, Li H, Kui X, Wang F, Wang Y, Wang C, Lei Y, Chen Y, Shen J, Sang L, Bai Y, Sun J. Screening for metastasis-related genes in mouse melanoma cells through sequential tail vein injection. BIOPHYSICS REPORTS 2024; 10:15-21. [PMID: 38737474 PMCID: PMC11079599 DOI: 10.52601/bpr.2023.230043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Accepted: 12/29/2023] [Indexed: 05/14/2024] Open
Abstract
Tumor metastasis, responsible for approximately 90% of cancer-associated mortality, remains poorly understood. Here in this study, we employed a melanoma lung metastasis model to screen for metastasis-related genes. By sequential tail vein injection of mouse melanoma B16F10 cells and the subsequently derived cells from lung metastasis into BALB/c mice, we successfully obtained highly metastatic B16F15 cells after five rounds of in vivo screening. RNA-sequencing analysis of B16F15 and B16F10 cells revealed a number of differentially expressed genes, some of these genes have previously been associated with tumor metastasis while others are novel discoveries. The identification of these metastasis-related genes not only improves our understanding of the metastasis mechanisms, but also provides potential diagnostic biomarkers and therapeutic targets for metastatic melanoma.
Collapse
Affiliation(s)
- Qinggang Hao
- Center for Life Sciences, Yunnan Key Laboratory of Cell Metabolism and Diseases, State Key Laboratory for Conservation and Utilization of Bio-Resources in Yunnan, School of Life Sciences, Yunnan University, Kunming 650500, China
- Key Laboratory of Tumor Immunological Prevention and Treatment of Yunnan Province, Yan'an Hospital Affiliated to Kunming Medical University, Kunming 650051, China
| | - Rui Dong
- Center for Life Sciences, Yunnan Key Laboratory of Cell Metabolism and Diseases, State Key Laboratory for Conservation and Utilization of Bio-Resources in Yunnan, School of Life Sciences, Yunnan University, Kunming 650500, China
| | - Weiyu Bai
- Center for Life Sciences, Yunnan Key Laboratory of Cell Metabolism and Diseases, State Key Laboratory for Conservation and Utilization of Bio-Resources in Yunnan, School of Life Sciences, Yunnan University, Kunming 650500, China
| | - Dong Chang
- Center for Life Sciences, Yunnan Key Laboratory of Cell Metabolism and Diseases, State Key Laboratory for Conservation and Utilization of Bio-Resources in Yunnan, School of Life Sciences, Yunnan University, Kunming 650500, China
| | - Xinyi Yao
- Center for Life Sciences, Yunnan Key Laboratory of Cell Metabolism and Diseases, State Key Laboratory for Conservation and Utilization of Bio-Resources in Yunnan, School of Life Sciences, Yunnan University, Kunming 650500, China
| | - Yingru Zhang
- Center for Life Sciences, Yunnan Key Laboratory of Cell Metabolism and Diseases, State Key Laboratory for Conservation and Utilization of Bio-Resources in Yunnan, School of Life Sciences, Yunnan University, Kunming 650500, China
| | - Huangying Xu
- Center for Life Sciences, Yunnan Key Laboratory of Cell Metabolism and Diseases, State Key Laboratory for Conservation and Utilization of Bio-Resources in Yunnan, School of Life Sciences, Yunnan University, Kunming 650500, China
| | - Huiyan Li
- Shandong Provincial Key Laboratory of Animal Resistance Biology, Collaborative Innovation Center of Cell Biology in Universities of Shandong, Center for Cell Structure and Function, Institute of Biomedical Science, College of Life Sciences, Shandong Normal University, Jinan 250014, China
| | - Xiang Kui
- Department of Pathology, The Second Affiliated Hospital of Kunming Medical University, Kunming 650101, China
| | - Feng Wang
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Institute of Genomic Medicine, College of Pharmacy, Jinan University, Guangzhou 510632, China
| | - Yan Wang
- Department of Pathology, The Second Affiliated Hospital of Kunming Medical University, Kunming 650101, China
| | - Chengqin Wang
- Department of Pathology, School of Basic Medicine, Qingdao University, Qingdao 266000, China
| | - Yujie Lei
- Department of Thoracic Surgery, Yunnan Cancer Hospital & The Third Affiliated Hospital of Kunming Medical University & Yunnan Cancer Center, Kunming 650106, China
| | - Yan Chen
- Shandong Provincial Key Laboratory of Animal Resistance Biology, Collaborative Innovation Center of Cell Biology in Universities of Shandong, Center for Cell Structure and Function, Institute of Biomedical Science, College of Life Sciences, Shandong Normal University, Jinan 250014, China
| | - Junling Shen
- Center for Life Sciences, Yunnan Key Laboratory of Cell Metabolism and Diseases, State Key Laboratory for Conservation and Utilization of Bio-Resources in Yunnan, School of Life Sciences, Yunnan University, Kunming 650500, China
| | - Lei Sang
- Center for Life Sciences, Yunnan Key Laboratory of Cell Metabolism and Diseases, State Key Laboratory for Conservation and Utilization of Bio-Resources in Yunnan, School of Life Sciences, Yunnan University, Kunming 650500, China
| | - Yan Bai
- Center for Life Sciences, Yunnan Key Laboratory of Cell Metabolism and Diseases, State Key Laboratory for Conservation and Utilization of Bio-Resources in Yunnan, School of Life Sciences, Yunnan University, Kunming 650500, China
| | - Jianwei Sun
- Center for Life Sciences, Yunnan Key Laboratory of Cell Metabolism and Diseases, State Key Laboratory for Conservation and Utilization of Bio-Resources in Yunnan, School of Life Sciences, Yunnan University, Kunming 650500, China
| |
Collapse
|
9
|
Khorasani M, Alaei M. cGAS-STING and PD1/PDL-1 pathway in breast cancer: a window to new therapies. J Recept Signal Transduct Res 2024; 44:1-7. [PMID: 38470108 DOI: 10.1080/10799893.2024.2325353] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Accepted: 02/23/2024] [Indexed: 03/13/2024]
Abstract
Breast cancer is a complex malignancy with diverse molecular and cellular subtypes and clinical outcomes. Despite advances in treatment, breast cancer remains a significant health challenge. However, recent advances in cancer immunotherapy have shown promising results in the treatment of breast cancer, particularly the use of inhibitors that target the immune checkpoint PD1/PDL1. Also, the cGAS-STING pathway, an important part of the innate immune response, has been considered as a major potential therapeutic target for breast cancer. In this narrative review, we provide an overview of the cGAS-STING and PD1/PDL-1 pathway in breast cancer, including their role in tumor development, progression, and response to treatment. We also discuss potential future directions for research.
Collapse
Affiliation(s)
- Milad Khorasani
- Healthy Ageing Research Centre, Neyshabur University of Medical Sciences, Neyshabur, Iran
- Department of Clinical Biochemistry, Neyshabur University of Medical Sciences, Neyshabur, Iran
| | - Maryam Alaei
- Department of Clinical Biochemistry, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
10
|
Zhang J, Yu S, Peng Q, Wang P, Fang L. Emerging mechanisms and implications of cGAS-STING signaling in cancer immunotherapy strategies. Cancer Biol Med 2024; 21:j.issn.2095-3941.2023.0440. [PMID: 38172538 PMCID: PMC10875285 DOI: 10.20892/j.issn.2095-3941.2023.0440] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2023] [Accepted: 12/14/2023] [Indexed: 01/05/2024] Open
Abstract
The intricate interplay between the human immune system and cancer development underscores the central role of immunotherapy in cancer treatment. Within this landscape, the innate immune system, a critical sentinel protecting against tumor incursion, is a key player. The cyclic GMP-AMP synthase (cGAS) and stimulator of interferon genes (STING) pathway has been found to be a linchpin of innate immunity: activation of this signaling pathway orchestrates the production of type I interferon (IFN-α/β), thus fostering the maturation, differentiation, and mobilization of immune effectors in the tumor microenvironment. Furthermore, STING activation facilitates the release and presentation of tumor antigens, and therefore is an attractive target for cancer immunotherapy. Current strategies to activate the STING pathway, including use of pharmacological agonists, have made substantial advancements, particularly when combined with immune checkpoint inhibitors. These approaches have shown promise in preclinical and clinical settings, by enhancing patient survival rates. This review describes the evolving understanding of the cGAS-STING pathway's involvement in tumor biology and therapy. Moreover, this review explores classical and non-classical STING agonists, providing insights into their mechanisms of action and potential for optimizing immunotherapy strategies. Despite challenges and complexities, the cGAS-STING pathway, a promising avenue for enhancing cancer treatment efficacy, has the potential to revolutionize patient outcomes.
Collapse
Affiliation(s)
- Jiawen Zhang
- Tongji University Cancer Center, Shanghai Tenth People’s Hospital, School of Medicine, Tongji University, Shanghai 200072, China
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 201620, China
| | - Sihui Yu
- Department of Obstetrics and Gynecology, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Qiao Peng
- Tongji University Cancer Center, Shanghai Tenth People’s Hospital, School of Medicine, Tongji University, Shanghai 200072, China
| | - Ping Wang
- Tongji University Cancer Center, Shanghai Tenth People’s Hospital, School of Medicine, Tongji University, Shanghai 200072, China
| | - Lan Fang
- Tongji University Cancer Center, Shanghai Tenth People’s Hospital, School of Medicine, Tongji University, Shanghai 200072, China
| |
Collapse
|
11
|
Choi MC, Kim SK, Choi YJ, Choi YJ, Kim S, Jegal KH, Lim SC, Kang KW. Role of monocarboxylate transporter I/lactate dehydrogenase B-mediated lactate recycling in tamoxifen-resistant breast cancer cells. Arch Pharm Res 2023; 46:907-923. [PMID: 38048029 DOI: 10.1007/s12272-023-01474-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Accepted: 11/25/2023] [Indexed: 12/05/2023]
Abstract
Although tamoxifen (TAM) is widely used in patients with estrogen receptor-positive breast cancer, the development of tamoxifen resistance is common. The previous finding suggests that the development of tamoxifen resistance is driven by epiregulin or hypoxia-inducible factor-1α-dependent glycolysis activation. Nonetheless, the mechanisms responsible for cancer cell survival and growth in a lactic acid-rich environment remain elusive. We found that the growth and survival of tamoxifen-resistant MCF-7 cells (TAMR-MCF-7) depend on glycolysis rather than oxidative phosphorylation. The levels of the glycolytic enzymes were higher in TAMR-MCF-7 cells than in parental MCF-7 cells, whereas the mitochondrial number and complex I level were decreased. Importantly, TAMR-MCF-7 cells were more resistant to low glucose and high lactate growth conditions. Isotope tracing analysis using 13C-lactate confirmed that lactate conversion to pyruvate was enhanced in TAMR-MCF-7 cells. We identified monocarboxylate transporter1 (MCT1) and lactate dehydrogenase B (LDHB) as important mediators of lactate influx and its conversion to pyruvate, respectively. Consistently, AR-C155858 (MCT1 inhibitor) inhibited the proliferation, migration, spheroid formation, and in vivo tumor growth of TAMR-MCF-7 cells. Our findings suggest that TAMR-MCF-7 cells depend on glycolysis and glutaminolysis for energy and support that targeting MCT1- and LDHB-dependent lactate recycling may be a promising strategy to treat patients with TAM-resistant breast cancer.
Collapse
Affiliation(s)
- Min Chang Choi
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, 08826, Republic of Korea
| | - Sang Kyum Kim
- College of Pharmacy, Chungnam University, Daejeon, 34134, Republic of Korea
| | - Young Jae Choi
- College of Pharmacy, Chungnam University, Daejeon, 34134, Republic of Korea
| | - Yong June Choi
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, 08826, Republic of Korea
| | - Suntae Kim
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, 08826, Republic of Korea
| | - Kyung Hwan Jegal
- College of Oriental Medicine, Daegu Haany University, Kyongsan, 38610, Republic of Korea
| | - Sung Chul Lim
- Department of Pathology, College of Medicine, Chosun University, Gwangju, 61452, Republic of Korea
| | - Keon Wook Kang
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, 08826, Republic of Korea.
| |
Collapse
|