1
|
Li G, Che X, Wang S, Liu D, Xie D, Jiang B, Zheng Z, Zheng X, Wu G. The role of cisplatin in modulating the tumor immune microenvironment and its combination therapy strategies: a new approach to enhance anti-tumor efficacy. Ann Med 2025; 57:2447403. [PMID: 39757995 PMCID: PMC11705547 DOI: 10.1080/07853890.2024.2447403] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Revised: 05/27/2024] [Accepted: 11/23/2024] [Indexed: 01/07/2025] Open
Abstract
Cisplatin is a platinum-based drug that is frequently used to treat multiple tumors. The anti-tumor effect of cisplatin is closely related to the tumor immune microenvironment (TIME), which includes several immune cell types, such as the tumor-associated macrophages (TAMs), cytotoxic T-lymphocytes (CTLs), dendritic cells (DCs), myeloid-derived suppressor cells (MDSCs), regulatory T cells (Tregs), and natural killer (NK) cells. The interaction between these immune cells can promote tumor survival and chemoresistance, and decrease the efficacy of cisplatin monotherapy. Therefore, various combination treatment strategies have been devised to enhance patient responsiveness to cisplatin therapy. Cisplatin can augment anti-tumor immune responses in combination with immune checkpoint blockers (such as PD-1/PD-L1 or CTLA4 inhibitors), lipid metabolism disruptors (like FASN inhibitors and SCD inhibitors) and nanoparticles (NPs), resulting in better outcomes. Exploring the interaction between cisplatin and the TIME will help identify potential therapeutic targets for improving the treatment outcomes in cancer patients.
Collapse
Affiliation(s)
- Guandu Li
- Department of Urology, the First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
| | - Xiangyu Che
- Department of Urology, the First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
| | - Shijin Wang
- Department of Urology, the First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
| | - Dequan Liu
- Department of Urology, the First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
| | - Deqian Xie
- Department of Urology, the First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
| | - Bowen Jiang
- Department of Urology, the First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
| | - Zunwen Zheng
- Department of Urology, the First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
| | - Xu Zheng
- Department of Cell Biology, College of Basic Medical Science, Dalian Medical University, Dalian, Liaoning, China
| | - Guangzhen Wu
- Department of Urology, the First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
| |
Collapse
|
2
|
Huang R, Wu Y, Shen F, Chen S, Yang X, Lin Y, Fang Y, Shen J. Manganese-coordinated nanoparticles loaded with CHK1 inhibitor dually activate cGAS-STING pathway and enhance efficacy of immune checkpoint therapy. Biomaterials 2025; 319:123199. [PMID: 40009899 DOI: 10.1016/j.biomaterials.2025.123199] [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: 11/03/2024] [Revised: 01/26/2025] [Accepted: 02/16/2025] [Indexed: 02/28/2025]
Abstract
Notable advancements have been made in utilizing immune checkpoint blockade (ICB) for the treatment of various cancers. However, the overall response rates and therapeutic effectiveness remain unsatisfactory. One cause is the inadequate immune environment characterized by poor T cell infiltration in tumors. To address these limitations, enhancing immune infiltration is crucial for optimizing the therapeutic efficacy of ICB. Activating the cyclic GMP-AMP synthase-stimulator of interferon genes (cGAS-STING) pathway is essential for initiating immune response and has become a potential target for developing combination therapies with ICB. In this study, we designed and fabricated manganese-containing nanoparticles loaded with the CHK1 inhibitor PF477736, which were subsequently encapsulated with macrophage membrane (PF/MMSN@MPM). This innovative design achieved excellent tumor targeting and demonstrated potent antitumor effects. The combination therapy dually amplified the cGAS-STING pathway, causing a cascade of enhanced therapeutic effects against tumors. Furthermore, single-cell mass cytometry (CyTOF) analysis revealed that PF/MMSN@MPM enhanced the activation and infiltration of immune cells. Moreover, the combination of PF/MMSN@MPM with anti-PD-1 (αPD-1) exhibited a stronger therapeutic effect compared to αPD-1 alone. PF/MMSN@MPM precisely and synergistically activated the cGAS-STING pathway, significantly improving therapeutic efficacy of ICB, and offering promising potential for tumor therapy.
Collapse
Affiliation(s)
- Rui Huang
- Department of Ophthalmology, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China; Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, 200025, China; Institute of Translational Medicine, National Facility for Translational Medicine, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Yijia Wu
- Department of Ophthalmology, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China; Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, 200025, China; Institute of Translational Medicine, National Facility for Translational Medicine, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Feiyang Shen
- Department of Ophthalmology, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China; Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, 200025, China; Institute of Translational Medicine, National Facility for Translational Medicine, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Shuai Chen
- Institute of Translational Medicine, National Facility for Translational Medicine, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Xiaoyu Yang
- Department of Ophthalmology, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China; Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, 200025, China; Institute of Translational Medicine, National Facility for Translational Medicine, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Yao Lin
- Institute of Translational Medicine, National Facility for Translational Medicine, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Yan Fang
- Department of Ophthalmology, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China; Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, 200025, China; Institute of Translational Medicine, National Facility for Translational Medicine, Shanghai Jiao Tong University, Shanghai, 200240, China.
| | - Jianfeng Shen
- Department of Ophthalmology, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China; Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, 200025, China; Institute of Translational Medicine, National Facility for Translational Medicine, Shanghai Jiao Tong University, Shanghai, 200240, China.
| |
Collapse
|
3
|
Zhang W, Wang J, Ji J, Wang P, Yuan G, Fang S, Liu F, Jin G, Zhang J. Glioblastoma cells secrete ICAM1 via FASN signaling to promote glioma-associated macrophage infiltration. Cell Signal 2025; 132:111823. [PMID: 40252818 DOI: 10.1016/j.cellsig.2025.111823] [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: 01/30/2025] [Revised: 04/14/2025] [Accepted: 04/16/2025] [Indexed: 04/21/2025]
Abstract
Glioma-associated macrophages (GAMs) constitute the most abundant subset of immune cells in the glioblastoma (GBM) microenvironment, but the underlying mechanism of intense infiltration needs to be elucidated. In this study, we found that GBM cells secrete ICAM1 via FASN signaling to promote GAM infiltration. FASN expression is correlated with GAM density in GBM patients. In vitro experiments revealed that FASN regulates the type-I interferon pathway, particularly STAT1 expression. Moreover, disrupting FASN-STAT1 signaling through the overexpression or inhibition of FASN or STAT1 in GBM cells strongly influences microglial recruitment. Additionally, ICAM1 acts as a direct transcriptional candidate of FASN-STAT1 and a paracrine soluble factor, recruiting microglia to GBM tumors. This study revealed crosstalk between GBM cells and GAMs through FASN-STAT1-ICAM1 signaling to promote microglial infiltration, suggesting potential strategies for treating GBM patients.
Collapse
Affiliation(s)
- Wenxin Zhang
- Brain Tumor Research Center, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China
| | - Jialin Wang
- Brain Tumor Research Center, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China
| | - Jiayu Ji
- Brain Tumor Research Center, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China; China Rehabilitation Science Institute, China Rehabilitation Research Center, School of Rehabilitation, Capital Medical University, Beijing, PR China
| | - Peiwen Wang
- Brain Tumor Research Center, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China
| | - Guiqiang Yuan
- Brain Tumor Research Center, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China
| | - Sheng Fang
- Brain Tumor Research Center, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China
| | - Fusheng Liu
- Brain Tumor Research Center, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China; Department of Neurosurgery, Beijing Tiantan Hosipital, Capital Medical University, Beijing, China
| | - Guishan Jin
- Brain Tumor Research Center, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China.
| | - Junwen Zhang
- Brain Tumor Research Center, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China.
| |
Collapse
|
4
|
Zhou S, Li T, Zhang W, Wu J, Hong H, Quan W, Qiao X, Cui C, Qiao C, Zhao W, Shen Y. The cGAS-STING-interferon regulatory factor 7 pathway regulates neuroinflammation in Parkinson's disease. Neural Regen Res 2025; 20:2361-2372. [PMID: 39359093 PMCID: PMC11759022 DOI: 10.4103/nrr.nrr-d-23-01684] [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: 10/10/2023] [Revised: 01/05/2024] [Accepted: 02/06/2024] [Indexed: 10/04/2024] Open
Abstract
JOURNAL/nrgr/04.03/01300535-202508000-00026/figure1/v/2024-09-30T120553Z/r/image-tiff Interferon regulatory factor 7 plays a crucial role in the innate immune response. However, whether interferon regulatory factor 7-mediated signaling contributes to Parkinson's disease remains unknown. Here we report that interferon regulatory factor 7 is markedly up-regulated in a 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine-induced mouse model of Parkinson's disease and co-localizes with microglial cells. Both the selective cyclic guanosine monophosphate adenosine monophosphate synthase inhibitor RU.521 and the stimulator of interferon genes inhibitor H151 effectively suppressed interferon regulatory factor 7 activation in BV2 microglia exposed to 1-methyl-4-phenylpyridinium and inhibited transformation of mouse BV2 microglia into the neurotoxic M1 phenotype. In addition, siRNA-mediated knockdown of interferon regulatory factor 7 expression in BV2 microglia reduced the expression of inducible nitric oxide synthase, tumor necrosis factor α, CD16, CD32, and CD86 and increased the expression of the anti-inflammatory markers ARG1 and YM1. Taken together, our findings indicate that the cyclic guanosine monophosphate adenosine monophosphate synthase-stimulator of interferon genes-interferon regulatory factor 7 pathway plays a crucial role in the pathogenesis of Parkinson's disease.
Collapse
Affiliation(s)
- Shengyang Zhou
- Laboratory of Neurodegenerative and Neuroinjury Diseases, Wuxi Medicine School, Jiangnan University, Wuxi, Jiangsu Province, China
| | - Ting Li
- Laboratory of Neurodegenerative and Neuroinjury Diseases, Wuxi Medicine School, Jiangnan University, Wuxi, Jiangsu Province, China
| | - Wei Zhang
- Laboratory of Neurodegenerative and Neuroinjury Diseases, Wuxi Medicine School, Jiangnan University, Wuxi, Jiangsu Province, China
| | - Jian Wu
- Laboratory of Neurodegenerative and Neuroinjury Diseases, Wuxi Medicine School, Jiangnan University, Wuxi, Jiangsu Province, China
| | - Hui Hong
- Laboratory of Neurodegenerative and Neuroinjury Diseases, Wuxi Medicine School, Jiangnan University, Wuxi, Jiangsu Province, China
| | - Wei Quan
- Laboratory of Neurodegenerative and Neuroinjury Diseases, Wuxi Medicine School, Jiangnan University, Wuxi, Jiangsu Province, China
| | - Xinyu Qiao
- Laboratory of Neurodegenerative and Neuroinjury Diseases, Wuxi Medicine School, Jiangnan University, Wuxi, Jiangsu Province, China
| | - Chun Cui
- Laboratory of Neurodegenerative and Neuroinjury Diseases, Wuxi Medicine School, Jiangnan University, Wuxi, Jiangsu Province, China
| | - Chenmeng Qiao
- Laboratory of Neurodegenerative and Neuroinjury Diseases, Wuxi Medicine School, Jiangnan University, Wuxi, Jiangsu Province, China
| | - Weijiang Zhao
- Laboratory of Neurodegenerative and Neuroinjury Diseases, Wuxi Medicine School, Jiangnan University, Wuxi, Jiangsu Province, China
| | - Yanqin Shen
- Laboratory of Neurodegenerative and Neuroinjury Diseases, Wuxi Medicine School, Jiangnan University, Wuxi, Jiangsu Province, China
| |
Collapse
|
5
|
Ren KX, Feng L, Wu P, Liu Y, Ren HM, Jin XW, Zhong CB, Zhou XQ, Jiang WD. Mitigation of the toxic effects of nitrite: Role and mechanism of isoleucine in mitigating mitochondrial DNA leakage-induced inflammation in grass carp (Ctenopharyngodon idella) under nitrite exposure. JOURNAL OF HAZARDOUS MATERIALS 2025; 491:138016. [PMID: 40147124 DOI: 10.1016/j.jhazmat.2025.138016] [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: 11/26/2024] [Revised: 03/12/2025] [Accepted: 03/19/2025] [Indexed: 03/29/2025]
Abstract
The physiological and growth processes of fish are closely associated with their surrounding environment. This study investigated the role and underlying mechanisms of isoleucine (Ile) in alleviating mitochondrial DNA (mtDNA) leakage-induced inflammation in grass carp under nitrite exposure. Grass carp were fed six experimental diets containing different Ile levels (0.00, 3.00, 6.00, 9.00, 12.00 and 15.00 g/kg) for 9 weeks, followed by a 96-hour nitrite exposure trial. Ile supplementation mitigated the deterioration of blood parameters including glutamic oxaloacetic transaminase (GOT), glutamic alanine transaminase (GPT), glucose, cortisol and lactate dehydrogenase (LDH) induced by nitrite exposure. Additionally, Ile enhanced its transport to the liver and mitochondria, as well as increased metabolism of Ile in mitochondria. Histological analyses revealed that Ile mitigated nitrite exposure-induced liver damage and mitochondrial cristae disruption. Furthermore, Ile preserved the mitochondrial cristae homeostasis by upregulating key proteins involved in mitochondrial structure maintenance, while inhibiting mtDNA leakage. Mechanistically, Ile attenuated mtDNA leakage-induced inflammation under nitrite exposure associated with the cyclic guanosine monophosphate-adenosine monophosphate synthase (cGAS)-Stimulator of Interferon Genes (STING) and NOD-like receptor thermal protein domain associated protein 3 (NLRP3) pathways. These findings highlight the protective role of Ile in reducing inflammation triggered by environmental nitrite exposure, offering new insights into aquatic toxicology, and determined that Ile concentration of 11.13 g/kg diet could be optimal for mitigating nitrite-induced stress in grass carp, providing a theoretical basis for formulating anti- nitrite stress diets in aquaculture.
Collapse
Affiliation(s)
- Kai-Xuan Ren
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, China
| | - Lin Feng
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, China; Fish Nutrition and Safety Production University Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China; Key Laboratory of Animal Disease-Resistance Nutrition, Ministry of Education, Ministry of Agriculture and Rural Affairs, Key Laboratory of Sichuan Province, Sichuan 611130, China
| | - Pei Wu
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, China; Fish Nutrition and Safety Production University Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China; Key Laboratory of Animal Disease-Resistance Nutrition, Ministry of Education, Ministry of Agriculture and Rural Affairs, Key Laboratory of Sichuan Province, Sichuan 611130, China
| | - Yang Liu
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, China; Fish Nutrition and Safety Production University Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China; Key Laboratory of Animal Disease-Resistance Nutrition, Ministry of Education, Ministry of Agriculture and Rural Affairs, Key Laboratory of Sichuan Province, Sichuan 611130, China
| | - Hong-Mei Ren
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, China
| | - Xiao-Wan Jin
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, China
| | - Cheng-Bo Zhong
- Sichuan Animal Science Academy, Sichuan Animtech Feed Co.Ltd, Chengdu 610066, China
| | - Xiao-Qiu Zhou
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, China; Fish Nutrition and Safety Production University Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China; Key Laboratory of Animal Disease-Resistance Nutrition, Ministry of Education, Ministry of Agriculture and Rural Affairs, Key Laboratory of Sichuan Province, Sichuan 611130, China.
| | - Wei-Dan Jiang
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, China; Fish Nutrition and Safety Production University Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China; Key Laboratory of Animal Disease-Resistance Nutrition, Ministry of Education, Ministry of Agriculture and Rural Affairs, Key Laboratory of Sichuan Province, Sichuan 611130, China.
| |
Collapse
|
6
|
Yang H, Xia Y, Ma Y, Gao M, Hou S, Xu S, Wang Y. Inhibition of the cGAS-STING pathway: contributing to the treatment of cerebral ischemia-reperfusion injury. Neural Regen Res 2025; 20:1900-1918. [PMID: 38993125 PMCID: PMC11691458 DOI: 10.4103/nrr.nrr-d-24-00015] [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: 01/05/2024] [Revised: 03/05/2024] [Accepted: 05/02/2024] [Indexed: 07/13/2024] Open
Abstract
The cGAS-STING pathway plays an important role in ischemia-reperfusion injury in the heart, liver, brain, and kidney, but its role and mechanisms in cerebral ischemia-reperfusion injury have not been systematically reviewed. Here, we outline the components of the cGAS-STING pathway and then analyze its role in autophagy, ferroptosis, cellular pyroptosis, disequilibrium of calcium homeostasis, inflammatory responses, disruption of the blood-brain barrier, microglia transformation, and complement system activation following cerebral ischemia-reperfusion injury. We further analyze the value of cGAS-STING pathway inhibitors in the treatment of cerebral ischemia-reperfusion injury and conclude that the pathway can regulate cerebral ischemia-reperfusion injury through multiple mechanisms. Inhibition of the cGAS-STING pathway may be helpful in the treatment of cerebral ischemia-reperfusion injury.
Collapse
Affiliation(s)
- Hang Yang
- School of Clinical Medicine, Shandong Second Medical University, Weifang, Shandong Province, China
| | - Yulei Xia
- School of Clinical Medicine, Shandong Second Medical University, Weifang, Shandong Province, China
| | - Yue Ma
- School of Clinical Medicine, Shandong Second Medical University, Weifang, Shandong Province, China
| | - Mingtong Gao
- Department of Emergency, The Affiliated Hospital of Weifang Medical University, Weifang, Shandong Province, China
| | - Shuai Hou
- School of Clinical Medicine, Shandong Second Medical University, Weifang, Shandong Province, China
| | - Shanshan Xu
- Department of Emergency, The Affiliated Hospital of Weifang Medical University, Weifang, Shandong Province, China
| | - Yanqiang Wang
- Department of Neurology II, The Affiliated Hospital of Weifang Medical University, Weifang, Shandong Province, China
| |
Collapse
|
7
|
Shen Y, Wei W, Lu Y, Song M, Yang S, Liu H, Xu X, Zhou H. Berberine alleviates tendinopathy by suppressing the cGAS-STING pathway and Relieving ferroptosis. Biochem Biophys Res Commun 2025; 767:151923. [PMID: 40319817 DOI: 10.1016/j.bbrc.2025.151923] [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: 02/28/2025] [Revised: 04/29/2025] [Accepted: 04/29/2025] [Indexed: 05/07/2025]
Abstract
Berberine, a key bioactive component of Coptis rhizome, has been extensively studied for its therapeutic effects on various diseases. This research aimed to investigate the potential benefits of berberine in treating tendinopathy and to elucidate the underlying mechanisms through animal and laboratory studies. Our findings indicated that berberine effectively treated type I collagenase-induced tendinopathy in rats, confirmed by cellular-level validation. At the molecular level, berberine reduced the activation of the cGAS-STING signaling pathway and decreased the accumulation of malondialdehyde (MDA) and reactive oxygen species (ROS) in both animal models and cell cultures. Additionally, berberine upregulated the expression of glutathione (GSH) and glutathione peroxidase 4 (GPX4) in tissues. These results suggested that berberine alleviated ferroptosis via the cGAS-STING pathway, thus exerting therapeutic effects on tendinopathy. To validate these findings further, we administered the ferroptosis inducer Imidazole Ketone Erastin (IKE) to evaluate the effects of berberine. IKE significantly diminished the therapeutic effects of berberine on tendinopathy, as indicated by the previously mentioned markers. Thus, berberine mitigated ferroptosis by inhibiting the cGAS-STING pathway, highlighting its potential in managing tendinopathy.
Collapse
Affiliation(s)
- Yeshuai Shen
- The Second Affiliated Hospital of Soochow University, China; The Affiliated Hospital of Xuzhou Medical University, China
| | - Wenting Wei
- The Affiliated Xuzhou Municipal Hospital of Xuzhou Medical University, China
| | - Yang Lu
- Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, 321 Zhongshan Road, Nanjing, 210008, Jiangsu, China
| | | | - Shaojie Yang
- Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, 321 Zhongshan Road, Nanjing, 210008, Jiangsu, China
| | - Huan Liu
- Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, 321 Zhongshan Road, Nanjing, 210008, Jiangsu, China
| | - Xingquan Xu
- Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, 321 Zhongshan Road, Nanjing, 210008, Jiangsu, China.
| | - Haibin Zhou
- The Second Affiliated Hospital of Soochow University, China.
| |
Collapse
|
8
|
Wang M, Jin G, Duan T, Li R, Gao Y, Yu M, Xu Y. Microglial phagocytosis and regulatory mechanisms: Key players in the pathophysiology of depression. Neuropharmacology 2025; 271:110383. [PMID: 39993469 DOI: 10.1016/j.neuropharm.2025.110383] [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: 09/08/2024] [Revised: 02/20/2025] [Accepted: 02/21/2025] [Indexed: 02/26/2025]
Abstract
Depression is a globally prevalent emotional disorder with a complex pathophysiology. Microglia are resident immune cells in the central nervous system, playing crucial roles in regulating inflammation, synaptic plasticity, immune phagocytosis, and other functions, thereby exerting significant impacts on neuropsychiatric disorders like depression. Increasing research indicates that abnormal phagocytic function of microglia in the brain is involved in depression, showing excessive or insufficient phagocytosis in different states. Here, we have provided a review of the signaling molecules involved in microglial phagocytosis in depression, including "eat me" signals such as phosphatidylserine (PS), complement, and "don't eat me" signals such as CD47, CD200 and related receptors. Furthermore, we discuss the regulatory effects of existing pharmaceuticals and dietary nutrients on microglial phagocytosis in depression, emphasizing the need for tailored modulation based on the varying phagocytic states of microglia. This review aims to facilitate a deeper understanding of the role of microglial phagocytosis in depression and provide a roadmap for potential therapeutic strategies for depression targeting microglial phagocytosis.
Collapse
Affiliation(s)
- Man Wang
- Department of Neurology, The Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu, 212001, China
| | - Guimin Jin
- Department of Neurology, The Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu, 212001, China
| | - Tingting Duan
- Department of Neurology, The Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu, 212001, China
| | - Run Li
- Department of Neurology, The Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu, 212001, China
| | - Yubin Gao
- Department of Neurology, The Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu, 212001, China
| | - Ming Yu
- Department of Neurology, The Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu, 212001, China.
| | - Yuhao Xu
- Department of Neurology, The Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu, 212001, China; Department of Neuroimaging Laboratory, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu, 212013, China.
| |
Collapse
|
9
|
Xue L, Liu R, Qiu T, Zhuang H, Li H, Zhang L, Yin R, Jiang T. Design, synthesis, and activity evaluation of novel STING inhibitors based on C170 and H151. Eur J Med Chem 2025; 290:117533. [PMID: 40157312 DOI: 10.1016/j.ejmech.2025.117533] [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: 12/31/2024] [Revised: 03/15/2025] [Accepted: 03/18/2025] [Indexed: 04/01/2025]
Abstract
Stimulating the STING signaling pathway for immune system activation is considered a promising strategy for cancer treatment. However, activating the STING pathway can lead to adverse effects, as aberrant activation or specific mutations in STING may result in autoimmune and inflammatory diseases. Therefore, the development of STING inhibitors is equally important. In this study, we first introduced hydroxyl groups into the STING inhibitors C170 and H151, creating functional sites for further modification. Then the introduction of various substituents resulted in the identification of more potent inhibitors, Y2 and HY2, which effectively suppressed the activation of the STING pathway in THP1 and RAW264.7 cells. Compounds Y2 and HY2 demonstrated potent anti-inflammatory effects in mice cisplatin-induced acute kidney injury models by inhibiting the STING pathway. Collectively, Y2 and HY2 warrant further investigation as novel anti-inflammatory agents.
Collapse
Affiliation(s)
- Liang Xue
- Key Laboratory of Marine Drugs, Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao, 266003, China; Laboratory for Marine Drugs and Bioproducts, Qingdao Marine Science and Technology Center, Qingdao, 266237, China
| | - Ruixue Liu
- Key Laboratory of Marine Drugs, Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao, 266003, China; Laboratory for Marine Drugs and Bioproducts, Qingdao Marine Science and Technology Center, Qingdao, 266237, China
| | - Tingting Qiu
- Key Laboratory of Marine Drugs, Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao, 266003, China; Laboratory for Marine Drugs and Bioproducts, Qingdao Marine Science and Technology Center, Qingdao, 266237, China
| | - Huiying Zhuang
- Key Laboratory of Marine Drugs, Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao, 266003, China; Laboratory for Marine Drugs and Bioproducts, Qingdao Marine Science and Technology Center, Qingdao, 266237, China
| | - Hongwei Li
- Key Laboratory of Marine Drugs, Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao, 266003, China; Laboratory for Marine Drugs and Bioproducts, Qingdao Marine Science and Technology Center, Qingdao, 266237, China
| | - Lican Zhang
- Key Laboratory of Marine Drugs, Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao, 266003, China; Laboratory for Marine Drugs and Bioproducts, Qingdao Marine Science and Technology Center, Qingdao, 266237, China
| | - Ruijuan Yin
- Key Laboratory of Marine Drugs, Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao, 266003, China; Laboratory for Marine Drugs and Bioproducts, Qingdao Marine Science and Technology Center, Qingdao, 266237, China; Marine Biomedical Research Institute of Qingdao, Qingdao, 266237, China.
| | - Tao Jiang
- Key Laboratory of Marine Drugs, Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao, 266003, China; Laboratory for Marine Drugs and Bioproducts, Qingdao Marine Science and Technology Center, Qingdao, 266237, China; Center for Innovative Drug Discovery, Greater Bay Area Institute of Precision Medicine (Guangzhou), Guangzhou, 511455, China.
| |
Collapse
|
10
|
Shmuel-Galia L, Jiang Z, Stine L, Cahill S, Ng SL, Wilson R, Kandasamy RK, Kurt-Jones EA, Ramanjulu JM, Bertin J, Kasparcova V, Pesiridis GS, Fitzgerald KA, Humphries F. Alternative cGAS signaling promotes herpes simplex encephalitis. Proc Natl Acad Sci U S A 2025; 122:e2423873122. [PMID: 40424128 DOI: 10.1073/pnas.2423873122] [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: 12/11/2024] [Accepted: 04/16/2025] [Indexed: 05/29/2025] Open
Abstract
During infection, foreign DNA is sensed by cyclic GMP-AMP synthase (cGAS) leading to the production of cGAMP, STING-dependent type I interferon and proinflammatory cytokine expression, and autophagy. To prevent a response to self-DNA, cGAS activity is tightly regulated. Dysregulation of cGAS underpins interferonopathies, such as Aicardi-Goutières syndrome, as well as Lupus and neurodegenerative diseases like Parkinson's disease. Thus, cGAS and its product cGAMP are therapeutic targets. However, if cGAS functions independently of cGAMP signaling is undefined. Here, we identified an alternative signaling pathway that cGAS engages independent of cGAMP synthesis. We demonstrate that alternative cGAS signaling promotes hyperexpression of CXCL1 and enhanced neutrophil recruitment that facilitates viral dissemination during herpes simplex encephalitis. Our study reports of an alternative cGAS response independent of cGAMP, highlighting a previously uncharacterized scaffold function for cGAS.
Collapse
Affiliation(s)
- Liraz Shmuel-Galia
- Division of Innate Immunity, Department of Medicine, University of Massachusetts Chan Medical School, Worcester, MA 01605
| | - Zhaozhao Jiang
- Division of Innate Immunity, Department of Medicine, University of Massachusetts Chan Medical School, Worcester, MA 01605
| | - Laurel Stine
- Division of Innate Immunity, Department of Medicine, University of Massachusetts Chan Medical School, Worcester, MA 01605
| | - Sara Cahill
- Division of Innate Immunity, Department of Medicine, University of Massachusetts Chan Medical School, Worcester, MA 01605
| | - Sze-Ling Ng
- Innate Immunity Research Unit, GlaxoSmithKline, Collegeville, PA 19426
| | - Ruth Wilson
- Division of Innate Immunity, Department of Medicine, University of Massachusetts Chan Medical School, Worcester, MA 01605
| | - Richard Kumaran Kandasamy
- Centre of Molecular Inflammation Research, and Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology, Trondheim 7491, Norway
| | - Evelyn A Kurt-Jones
- Division of Infectious Diseases and Immunology, Department of Medicine, University of Massachusetts Chan Medical, Worcester, MA 01605
| | - Joshi M Ramanjulu
- Innate Immunity Research Unit, GlaxoSmithKline, Collegeville, PA 19426
| | - John Bertin
- Immunology and Inflammation Therapeutic Area, Sanofi, Cambridge, MA 02139
| | - Viera Kasparcova
- Innate Immunity Research Unit, GlaxoSmithKline, Collegeville, PA 19426
| | - G Scott Pesiridis
- Innate Immunity Research Unit, GlaxoSmithKline, Collegeville, PA 19426
| | - Katherine A Fitzgerald
- Division of Innate Immunity, Department of Medicine, University of Massachusetts Chan Medical School, Worcester, MA 01605
| | - Fiachra Humphries
- Division of Innate Immunity, Department of Medicine, University of Massachusetts Chan Medical School, Worcester, MA 01605
| |
Collapse
|
11
|
Zhang X, Du H, Qiu T, Fu H, Dai J, Lian Q, Yan F, Guo D, Lin J, Xu S, Li D, Chen Q, Huang Z. Tanshinone IIA alleviates myocarditis in Trex1-D18N lupus-like mice by inhibiting the interaction between STING and SEC24C. Int Immunopharmacol 2025; 156:114659. [PMID: 40252465 DOI: 10.1016/j.intimp.2025.114659] [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: 01/14/2025] [Revised: 04/07/2025] [Accepted: 04/08/2025] [Indexed: 04/21/2025]
Abstract
The cyclic GMP-AMP synthase-stimulator of interferon genes (cGAS-STING) signaling pathway serves as a crucial component of the innate immune defense, playing a vital role in combating pathogen invasion. However, its dysregulation or abnormal activation can trigger the development of autoimmune diseases. This study demonstrated that Tanshinone IIA, a major lipid-soluble component of Salvia miltiorrhiza Bunge, can effectively inhibit the activation of the cGAS-STING signaling pathway. Mechanistically, Tanshinone IIA inhibits the transport of STING from the ER to the Golgi apparatus by weakening the interaction between STING and SEC24C, thereby preventing the activation of the cGAS-STING signaling pathway. Furthermore, Tanshinone IIA significantly ameliorated myocardial inflammation in WT and Trex1D18N/D18N mice. Our research indicates that Tanshinone IIA shows potential therapeutic value in alleviating autoimmune diseases by effectively inhibiting the abnormal activation of the cGAS-STING pathway.
Collapse
Affiliation(s)
- Xiaoxiong Zhang
- Department of Integrative Medicine, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, Fujian Province 350117, China; Fujian Key Laboratory of Innate Immune Biology, Biomedical Research Center of South China, College of Life Sciences, Fujian Normal University Qishan Campus, College Town, Fuzhou, Fujian Province 350117, China
| | - Hekang Du
- Fujian Key Laboratory of Innate Immune Biology, Biomedical Research Center of South China, College of Life Sciences, Fujian Normal University Qishan Campus, College Town, Fuzhou, Fujian Province 350117, China; Department of Pathology, The First Affiliated Hospital, Fujian Medical University, 20 Chazhong Road, Fuzhou 350005, China
| | - Tao Qiu
- Fujian Key Laboratory of Innate Immune Biology, Biomedical Research Center of South China, College of Life Sciences, Fujian Normal University Qishan Campus, College Town, Fuzhou, Fujian Province 350117, China
| | - Honggao Fu
- School of Basic Medical Sciences, Fujian Medical University, Fuzhou 350005, China
| | - Jiawei Dai
- Institutes of Biomedical Sciences, Fudan University, Shanghai 200030, China
| | - Qiying Lian
- Fujian Key Laboratory of Innate Immune Biology, Biomedical Research Center of South China, College of Life Sciences, Fujian Normal University Qishan Campus, College Town, Fuzhou, Fujian Province 350117, China
| | - Fang Yan
- Fujian Key Laboratory of Innate Immune Biology, Biomedical Research Center of South China, College of Life Sciences, Fujian Normal University Qishan Campus, College Town, Fuzhou, Fujian Province 350117, China
| | - Dong Guo
- Fujian Key Laboratory of Innate Immune Biology, Biomedical Research Center of South China, College of Life Sciences, Fujian Normal University Qishan Campus, College Town, Fuzhou, Fujian Province 350117, China
| | - Jinpei Lin
- Department of Integrative Medicine, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, Fujian Province 350117, China
| | - Shan Xu
- Fujian Key Laboratory of Innate Immune Biology, Biomedical Research Center of South China, College of Life Sciences, Fujian Normal University Qishan Campus, College Town, Fuzhou, Fujian Province 350117, China
| | - Daliang Li
- Fujian Key Laboratory of Innate Immune Biology, Biomedical Research Center of South China, College of Life Sciences, Fujian Normal University Qishan Campus, College Town, Fuzhou, Fujian Province 350117, China.
| | - Qi Chen
- Fujian Key Laboratory of Innate Immune Biology, Biomedical Research Center of South China, College of Life Sciences, Fujian Normal University Qishan Campus, College Town, Fuzhou, Fujian Province 350117, China.
| | - Zhengrong Huang
- Department of Integrative Medicine, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, Fujian Province 350117, China.
| |
Collapse
|
12
|
Zhou Q, Luo J, Chai X, Yang J, Zhong S, Zhang Z, Chang X, Wang H. Therapeutic targeting the cGAS-STING pathway associated with protein and gene: An emerging and promising novel strategy for aging-related neurodegenerative disease. Int Immunopharmacol 2025; 156:114679. [PMID: 40252469 DOI: 10.1016/j.intimp.2025.114679] [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: 02/04/2025] [Revised: 04/11/2025] [Accepted: 04/13/2025] [Indexed: 04/21/2025]
Abstract
Neurodegenerative diseases (NDDs) represent a rapidly escalating global health challenge, contributing significantly to the worldwide disease burden and posing substantial threats to public health systems across nations. Among the many risk factors for neurodegeneration, aging is the major risk factor. In the context of aging, multiple factors lead to the release of endogenous DNA (especially mitochondrial DNA, mtDNA), which is an important trigger for the activation of the cGAS-STING innate immune pathway. Recent studies have identified an increasing role for activation of the cGAS-STING signaling pathway as a driver of senescence-associated secretory phenotypes (SASPs) in aging and NDDs. The cGAS-STING pathway mediates the immune sensing of DNA and is a key driver of chronic inflammation and functional decline during the aging process. Blocking cGAS-STING signaling may reduce the inflammatory response by preventing mtDNA release and enhancing mitophagy. Targeted inhibition of the cGAS-STING pathway by biological macromolecules such as natural products shows promise in therapeutic strategies for age-related NDDs. This review aims to systematically and comprehensively introduces the role of the cGAS-STING pathway in age-related NDDs in the context of aging while revealing the molecular mechanisms of the cGAS-STING pathway and its downstream signaling pathways and to develop more targeted and effective therapeutic strategies for NDDs.
Collapse
Affiliation(s)
- Qiongli Zhou
- Department of Toxicology, School of Public Health, Lanzhou University, Gansu 730000, China
| | - Jinghao Luo
- Department of Toxicology, School of Public Health, Lanzhou University, Gansu 730000, China
| | - Xueting Chai
- Department of Toxicology, School of Public Health, Lanzhou University, Gansu 730000, China
| | - Jirui Yang
- Department of Toxicology, School of Public Health, Lanzhou University, Gansu 730000, China
| | - Shiyin Zhong
- Department of Toxicology, School of Public Health, Lanzhou University, Gansu 730000, China
| | - Zhimin Zhang
- Department of Toxicology, School of Public Health, Lanzhou University, Gansu 730000, China
| | - Xuhong Chang
- Department of Toxicology, School of Public Health, Lanzhou University, Gansu 730000, China
| | - Hui Wang
- Department of Toxicology, School of Public Health, Lanzhou University, Gansu 730000, China.
| |
Collapse
|
13
|
Chen Z, Cheng J, Li Z, Huang H, Ren Y, Zhou Y, Li J, Zhang Q, Duan X, Hu Y. Dual targeted ferritin-based delivery system blocks the crosstalk between cancer cells and cancer-associated fibroblasts to potentiate immunotherapy of colorectal cancer. J Control Release 2025:113877. [PMID: 40425094 DOI: 10.1016/j.jconrel.2025.113877] [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: 10/31/2024] [Revised: 05/05/2025] [Accepted: 05/20/2025] [Indexed: 05/29/2025]
Abstract
The crosstalk between cancer cells and cancer-associated fibroblasts (CAFs) plays a pivotal role in maintaining immunosuppressive tumor microenvironment (iTME) and promoting tumor progression and metastasis. Herein, we first verify that the presence of CAFs significantly promotes colorectal cancer progression and causes an iTME in mouse model and publicly available single-cell data. Then, we constructed a ferritin-based dual-targeting delivery system, named HFAI (mHFn-FAPI@ATMi/ICG), to prevent communications between cancer cells and CAFs to enhance colorectal cancer immunotherapy. Indeed, mouse-derived heavy chain ferritin (mHFn) was modified with fibroblast activation protein-α inhibitor (FAPI) to endow it dual targeting capability. In tumor cells, photosensitizer indocyanine green (ICG)-mediated photothermal therapy (PTT) and photodynamic therapy (PDT), together with ataxia-telangiectasia mutated inhibitors (ATMi)-mediated DNA repair inhibition, triggered DNA damage accumulation, activated cGAS/STING signaling pathway. Whereas in CAFs, ICG-mediated PTT/PDT eradicated CAFs, while ATMi normalized CAFs. The activation of the cGAS/STING pathway and the elimination of CAFs reshaped the iTME, promoted immune cell infiltration, and enhanced the immunotherapy of CAFs-rich tumors. Furthermore, HFAI serves as a promising probe for identifying CAFs-rich peritoneal metastasis tissues and enabling near-infrared fluorescence-guided accurate resection. Taken together, this dual-targeted nanocomposites showed a remarkably synergistic therapy effect against colorectal cancer by modulating tumor-stromal crosstalk.
Collapse
Affiliation(s)
- Zhian Chen
- Zhongshan City People's Hospital, Zhongshan 528403, China; Department of General Surgery, Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Tumor, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Jinmei Cheng
- Cancer Research Institute, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong 510515, China
| | - Zhenhao Li
- Department of General Surgery, Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Tumor, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Huilin Huang
- Department of General Surgery, Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Tumor, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Yingxin Ren
- Department of General Surgery, Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Tumor, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Yang Zhou
- Department of General Surgery, Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Tumor, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Jinhui Li
- Cancer Research Institute, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong 510515, China
| | - Qianbing Zhang
- Cancer Research Institute, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong 510515, China.
| | - Xiaopin Duan
- Cancer Research Institute, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong 510515, China.
| | - Yanfeng Hu
- Department of General Surgery, Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Tumor, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China.
| |
Collapse
|
14
|
Wang Z, Liu J, Mou Y, Zhou X, Liao W, Li Y, Liu Y, Tang J. Disruption of cholesterol homeostasis triggers NLRP3-cGAS-STING axis-dependent hepatic fibrosis and honokiol intervention effects. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 143:156904. [PMID: 40449452 DOI: 10.1016/j.phymed.2025.156904] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Revised: 05/16/2025] [Accepted: 05/23/2025] [Indexed: 06/03/2025]
Abstract
BACKGROUND Maintaining cholesterol homeostasis is crucial for sustaining human health and physiological function. Although the detrimental effects of chronic cholesterol overload on hepatic injury and fibrosis are well documented, the molecular mechanisms driving this pathology remain incompletely understood. PURPOSE This study investigates the mechanistic role of chronic cholesterol overload in driving liver fibrosis and evaluates the therapeutic efficacy of honokiol as a targeted intervention. STUDY DESIGN AND METHODS High-cholesterol models induced by cholesterol and 25-hydroxycholesterol in human HepG2 cells or induced by cholesterol crystals in mouse bone marrow-derived macrophages were established. We also examined the effect of cholesterol on the livers of mice following a 20-week regimen of high-cholesterol diets. RESULTS Excess cholesterol interfered with normal cholesterol metabolism both in vitro and in vivo, and led to liver damage and fibrosis in vivo. Further research showed that cholesterol exposure triggered NLRP3 inflammasome activation and programmed cell death called pyroptosis; induced an increase in mitochondrial ROS and a disruption of intracellular redox homeostasis, followed by the opening of the mitochondrial permeability transition pore; and finally induced cellular DNA damage, resulting in the translocation of the double-stranded DNA fragment into the cytoplasm and the activation of the DNA-sensing adaptor STING. The activation of the NLRP3-cGAS-STING axis initiated the downstream cascade reaction and up-regulated the expression of pro-inflammatory cytokines, including IL-1β, TNF-α, and IFN-β, thus facilitating liver damage and fibrosis. Furthermore, honokiol, an active ingredient in Magnolia officinalis, could alleviate liver damage and fibrosis by blocking NLRP3 inflammasome activation, pyroptosis, and the cGAS-STING pathway. CONCLUSION Systematic evidence shows that cholesterol induces liver fibrosis through the activation of the NLRP3-cGAS-STING signaling axis and that honokiol demonstrates interventional efficacy in mitigating this process.
Collapse
Affiliation(s)
- Zhilei Wang
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu 610075, China.
| | - Jingwen Liu
- Department of Oncology, Chongqing Traditional Chinese Medicine Hospital, Chongqing, 400021, China
| | - Yu Mou
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu 610075, China
| | - Xianglu Zhou
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu 610075, China
| | - Wenhao Liao
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu 610075, China
| | - Yuchen Li
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu 610075, China
| | - Yong Liu
- Department of Oncology, Chongqing Traditional Chinese Medicine Hospital, Chongqing, 400021, China.
| | - Jianyuan Tang
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu 610075, China.
| |
Collapse
|
15
|
Dhapola R, Paidlewar M, Kumari S, Sharma P, Vellingiri B, Medhi B, HariKrishnaReddy D. cGAS-STING and neurodegenerative diseases: A molecular crosstalk and therapeutic perspective. Int Immunopharmacol 2025; 159:114902. [PMID: 40403503 DOI: 10.1016/j.intimp.2025.114902] [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: 02/21/2025] [Revised: 05/05/2025] [Accepted: 05/15/2025] [Indexed: 05/24/2025]
Abstract
Neurodegenerative disorders such as Alzheimer's disease (AD), Parkinson's disease (PD), Huntington's disease (HD), Amyotrophic Lateral Sclerosis (ALS), Multiple Sclerosis (MS) and Frontotemporal Dementia (FTD) share key pathological features, including neuroinflammation, oxidative stress, mitochondrial dysfunction, autophagic dysfunction, and DNA damage. By identifying cytosolic DNA and triggering the type I interferon response, the cyclic GMP-AMP synthase (cGAS)-stimulator of interferon genes (STING) pathway regulates neuroinflammation. Dysregulated cGAS-STING signaling has been linked to neuroinflammation and neuronal degeneration across multiple neurodegenerative conditions. In many neurodegenerative disorders, neuroinflammation is mediated by the cGAS-STING pathway. Mitochondrial malfunction and impaired autophagy cause cytosolic DNA buildup in Huntington's, Parkinson's, and Alzheimer's diseases, which activates cGAS-STING and drives chronic inflammation. This pathway is triggered by TDP-43 pathology and nucleic acid dysregulation in ALS and FTD, which leads to neuronal destruction. Both central demyelination and peripheral immunological responses are linked to cGAS-STING activation in multiple sclerosis. Various inhibitors, such as RU.521, H-151, and naturally occurring compounds like metformin, potentially attenuate cGAS-STING-mediated neuroinflammation and associated pathologies. H-151 significantly decreased the expression of pro-inflammatory markers in murine macrophage J774 cells activated with cGAMP: TNF-α by 68 %, IFN-β by 84 %, and CXCL10 by 96 %. cGAS-STING inhibitors target neuroinflammation, offering a disease-modifying approach unlike current symptomatic treatments. However, challenges like blood-brain barrier penetration, off-target effects, and immune suppression hinder clinical translation, necessitating optimized drug delivery and immune modulation. With a focus on its potential for future clinical applications, this review explores the role of the cGAS-STING pathway in neurodegeneration and new treatment approaches.
Collapse
Affiliation(s)
- Rishika Dhapola
- Advanced Pharmacology and Neuroscience Laboratory, Department of Pharmacology, School of Health Sciences, Central University of Punjab, Ghudda, Bathinda 151401, Punjab, India
| | - Mohit Paidlewar
- Advanced Pharmacology and Neuroscience Laboratory, Department of Pharmacology, School of Health Sciences, Central University of Punjab, Ghudda, Bathinda 151401, Punjab, India
| | - Sneha Kumari
- Advanced Pharmacology and Neuroscience Laboratory, Department of Pharmacology, School of Health Sciences, Central University of Punjab, Ghudda, Bathinda 151401, Punjab, India
| | - Prajjwal Sharma
- Advanced Pharmacology and Neuroscience Laboratory, Department of Pharmacology, School of Health Sciences, Central University of Punjab, Ghudda, Bathinda 151401, Punjab, India
| | - Balachandar Vellingiri
- Human Cytogenetics and Stem Cell Laboratory, Department of Zoology, School of Basic Sciences, Central University of Punjab, 151401 Bathinda, Punjab, India
| | - Bikash Medhi
- Department of Pharmacology, Post Graduate Institute of Medical Education and Research, Chandigarh, 160012, India
| | - Dibbanti HariKrishnaReddy
- Advanced Pharmacology and Neuroscience Laboratory, Department of Pharmacology, School of Health Sciences, Central University of Punjab, Ghudda, Bathinda 151401, Punjab, India.
| |
Collapse
|
16
|
Moosavi Zenooz A, Eterafi M, Azarmi Giglou S, Safarzadeh E. Embracing cancer immunotherapy with manganese particles. Cell Oncol (Dordr) 2025:10.1007/s13402-025-01070-9. [PMID: 40397376 DOI: 10.1007/s13402-025-01070-9] [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: 02/15/2025] [Accepted: 05/05/2025] [Indexed: 05/22/2025] Open
Abstract
A substance integral to the sustenance and functionality of virtually all forms of life is manganese (Mn), classified as an essential trace metal. Its significance lies in its pivotal role in facilitating metabolic processes crucial for survival. Additionally, Mn exerts influence over various biological functions including bone formation and maintenance, as well as regulation within systems governing immunity, nervous signaling, and digestion. Manganese nanoparticles (Mn-NP) stand out as a beacon of promise within the realm of immunotherapy, their focus honed on intricate mechanisms such as triggering immune pathways, igniting inflammasomes, inducing immunogenic cell death (ICD), and sculpting the nuances of the tumor microenvironment. These minuscule marvels have dazzled researchers with their potential in reshaping the landscape of cancer immunotherapy - serving as potent vaccine enhancers, efficient drug couriers, and formidable allies when paired with immune checkpoint inhibitors (ICIs) or cutting-edge photodynamic/photothermal therapies. Herein, we aim to provide a comprehensive review of recent advances in the application of Mn and Mn-NP in the immunotherapy of cancer. We hope that this review will display an insightful view of Mn-NPs and provide guidance for design and application of them in immune-based cancer therapies.
Collapse
Affiliation(s)
- Ali Moosavi Zenooz
- Cancer Immunology and Immunotherapy Research Center, Ardabil University of Medical Sciences, Ardabil, Iran
- Students Research Committee, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Majid Eterafi
- Cancer Immunology and Immunotherapy Research Center, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Soheil Azarmi Giglou
- Students Research Committee, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Elham Safarzadeh
- Cancer Immunology and Immunotherapy Research Center, Ardabil University of Medical Sciences, Ardabil, Iran.
- Department of Microbiology, Parasitology and Immunology, School of Medicine, Ardabil University of Medical Sciences, Ardabil, 5166614711, Iran.
| |
Collapse
|
17
|
Wang Y, Li Z, Li Z, Song Y, Li J, Yuan L, Wang C, Lai F, Yan R, Xiao W, Wang J. Zebrafish fkbp5 attenuates antiviral innate immunity by autophagic degradation of transcription factor irf7. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2025:vkaf089. [PMID: 40391431 DOI: 10.1093/jimmun/vkaf089] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/15/2024] [Accepted: 03/25/2025] [Indexed: 05/21/2025]
Abstract
Activation of the type I interferon (IFN-I) signaling pathway is crucial for protecting host cells against viral infections. IFN-I production requires the transcription factors IFN regulatory factor 3 (IRF3) and IRF7, and its regulation must be finely tuned to both combat infection effectively and prevent excessive immunopathology. Here, we report that selective autophagy mediated by zebrafish FK506-binding protein 5 (Fkbp5), a PPIase (peptidyl-prolyl isomerase) promotes the degradation of Irf7 and Irf3, thereby inhibiting virus-induced type I IFN production. Quantitative real-time reverse-transcription polymerase chain reaction experiments indicate that zebrafish fkbp5 is induced by viral infection. Moreover, disrupting fkbp5 in AB-line zebrafish using CRISPR/Cas9 enhances survival rates and reduces viral messenger RNA levels compared with wild-type zebrafish. In cell culture, using promoter analysis and quantitative real-time reverse-transcription polymerase chain reaction, we found fkbp5 overexpression significantly attenuates cellular antiviral capacity and facilitates viral proliferation. Mechanistically, we found that fkbp5 inhibits Irf3/7-induced IFN activation, which depends on the binding of Fkbp5 to the Irf3 or IRF association domain of Irf7 via co-immunoprecipitation and Western blot assays. Furthermore, Fkbp5 induces the autophagic degradation of Irf3 and Irf7 independent of its PPIase activity. Blocking autophagy in vivo and in vitro restores the regulation of the RLR (RIG-I-like receptor) pathway by fkbp5. These findings reveal a critical role for zebrafish fkbp5 in suppressing the activation of Irf7 and Irf3 for IFN signaling and antiviral immune responses.
Collapse
Affiliation(s)
- Yanyi Wang
- Key Laboratory of Breeding Biotechnology and Sustainable Aquaculture, Hubei Hongshan Laboratory Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, P. R. China
- Laboratory for Marine Biology and Biotechnology, Qingdao Marine Science and Technology Center, Qingdao, P. R. China
- University of Chinese Academy of Sciences, Beijing, P. R. China
| | - Zhi Li
- Key Laboratory of Breeding Biotechnology and Sustainable Aquaculture, Hubei Hongshan Laboratory Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, P. R. China
- Laboratory for Marine Biology and Biotechnology, Qingdao Marine Science and Technology Center, Qingdao, P. R. China
- University of Chinese Academy of Sciences, Beijing, P. R. China
| | - Ziyi Li
- Key Laboratory of Breeding Biotechnology and Sustainable Aquaculture, Hubei Hongshan Laboratory Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, P. R. China
- Laboratory for Marine Biology and Biotechnology, Qingdao Marine Science and Technology Center, Qingdao, P. R. China
- University of Chinese Academy of Sciences, Beijing, P. R. China
| | - Yanan Song
- Key Laboratory of Breeding Biotechnology and Sustainable Aquaculture, Hubei Hongshan Laboratory Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, P. R. China
- Laboratory for Marine Biology and Biotechnology, Qingdao Marine Science and Technology Center, Qingdao, P. R. China
- University of Chinese Academy of Sciences, Beijing, P. R. China
| | - Jun Li
- Key Laboratory of Breeding Biotechnology and Sustainable Aquaculture, Hubei Hongshan Laboratory Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, P. R. China
- Laboratory for Marine Biology and Biotechnology, Qingdao Marine Science and Technology Center, Qingdao, P. R. China
- University of Chinese Academy of Sciences, Beijing, P. R. China
| | - Le Yuan
- Key Laboratory of Breeding Biotechnology and Sustainable Aquaculture, Hubei Hongshan Laboratory Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, P. R. China
- Laboratory for Marine Biology and Biotechnology, Qingdao Marine Science and Technology Center, Qingdao, P. R. China
- University of Chinese Academy of Sciences, Beijing, P. R. China
| | - Chunling Wang
- Key Laboratory of Breeding Biotechnology and Sustainable Aquaculture, Hubei Hongshan Laboratory Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, P. R. China
| | - Fuxiang Lai
- Key Laboratory of Breeding Biotechnology and Sustainable Aquaculture, Hubei Hongshan Laboratory Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, P. R. China
- Laboratory for Marine Biology and Biotechnology, Qingdao Marine Science and Technology Center, Qingdao, P. R. China
- University of Chinese Academy of Sciences, Beijing, P. R. China
| | - Runkun Yan
- Key Laboratory of Breeding Biotechnology and Sustainable Aquaculture, Hubei Hongshan Laboratory Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, P. R. China
- Laboratory for Marine Biology and Biotechnology, Qingdao Marine Science and Technology Center, Qingdao, P. R. China
- University of Chinese Academy of Sciences, Beijing, P. R. China
| | - Wuhan Xiao
- Key Laboratory of Breeding Biotechnology and Sustainable Aquaculture, Hubei Hongshan Laboratory Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, P. R. China
- Laboratory for Marine Biology and Biotechnology, Qingdao Marine Science and Technology Center, Qingdao, P. R. China
- University of Chinese Academy of Sciences, Beijing, P. R. China
- Innovation of Seed Design, Chinese Academy of Sciences, Wuhan, P. R. China
| | - Jing Wang
- Key Laboratory of Breeding Biotechnology and Sustainable Aquaculture, Hubei Hongshan Laboratory Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, P. R. China
- Laboratory for Marine Biology and Biotechnology, Qingdao Marine Science and Technology Center, Qingdao, P. R. China
- University of Chinese Academy of Sciences, Beijing, P. R. China
- Innovation of Seed Design, Chinese Academy of Sciences, Wuhan, P. R. China
| |
Collapse
|
18
|
Jiang H, Zeng Y, Ning W, Hong J, Zhu M, Li P, Ye F, Chen Z, Chen H, Chen W, Li G, Huang H. Ketogenesis instigates immune suppression in enzalutamide resistant prostate cancer via OTUD7B β-hydroxybutyrylation. Cancer Lett 2025; 625:217808. [PMID: 40393641 DOI: 10.1016/j.canlet.2025.217808] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2025] [Revised: 05/08/2025] [Accepted: 05/17/2025] [Indexed: 05/22/2025]
Abstract
Next-generation androgen receptor inhibitors are the primary treatment for metastatic prostate cancer. Unfortunately, the majority of patients rapidly develop resistance. Resistance to enzalutamide has been linked to the emergence of an immunosuppressive tumor, although the underlying mechanisms remain poorly understood. In this study, we observed a marked overexpression of enzymes involved in the ketogenic pathway in enzalutamide-induced castration-resistant prostate cancer, which contributed to immune desertification and resistance to immunotherapy. Mechanistically, upregulation of the ketogenic pathway led to the accumulation of β-hydroxybutyrate, which promoted β-hydroxybutyrylation of the cell cycle-regulated deubiquitinase OTUD7B at lysine 511. This modification impaired the degradation of APC/C substrates, resulting in a subsequent reduction in cytoplasmic double-stranded DNA accumulation, thereby attenuating cGAS-STING activation and interferon expression. These findings shed light on the metabolic adaptations and immune escape driven by androgen receptor signaling inhibitors, potentially informing the development of more effective and durable therapeutic approaches in the near future.
Collapse
Affiliation(s)
- Haoran Jiang
- Department of Urology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China; Institute of Urology, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China; Zhejiang Key Laboratory of Critical Care Medicine,Wenzhou, 325000,Zhejiang China; Translational Medicine Laboratory, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China; Key Laboratory of Novel Nuclide Technologies on Precision Diagnosis and Treatment & Clinical Transformation of Wenzhou City, Wenzhou, 325035, Zhejiang, China; Department of Radiation Oncology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China
| | - Yuan Zeng
- Department of Gastroenterology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China
| | - Weiqiang Ning
- Department of Urology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China; Institute of Urology, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China
| | - Junkai Hong
- Department of Urology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China; Institute of Urology, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China
| | - Moyang Zhu
- Department of Urology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China; Institute of Urology, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China; Translational Medicine Laboratory, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China
| | - Ping Li
- Center for Wound Repair and Regeneration, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China
| | - Fangdie Ye
- Fudan Institute of Urology, Huashan Hospital, Fudan University, Shanghai, 200000, China
| | - Zhifa Chen
- Department of Urology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China; Institute of Urology, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China
| | - Haoran Chen
- Department of Urology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China; Institute of Urology, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China
| | - Wei Chen
- Department of Urology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China; Institute of Urology, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China.
| | - Gang Li
- Department of Radiation Oncology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China.
| | - Hang Huang
- Department of Urology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China; Institute of Urology, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China; Zhejiang Key Laboratory of Critical Care Medicine,Wenzhou, 325000,Zhejiang China.
| |
Collapse
|
19
|
Nouri HR, Schaunaman N, Kraft M, Numata M, Vercelli D, Chu HW. Tollip deficiency enhances mitophagy and reduces STING activation in influenza A virus-infected mice. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2025:vkaf058. [PMID: 40375803 DOI: 10.1093/jimmun/vkaf058] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/19/2024] [Accepted: 02/13/2025] [Indexed: 05/18/2025]
Abstract
Toll-interacting protein (Tollip) is an intracellular adaptor protein with diverse functions including regulation of autophagy of mitochondria-mitophagy. Tollip deficiency promotes viral infection, but whether mitophagy is involved remains unclear. We sought to determine if mitophagy and associated signaling such as mitochondrial DNA (mtDNA) release and activation of stimulator of interferon genes (STING) contribute to worsened viral infection due to Tollip deficiency. Wild-type and Tollip knockout (KO) C57/BL6 mice were intranasally infected with influenza A virus (IAV), and then treated with or without a STING agonist 2'3'cGAMP for 4 d. PINK1 (an initiator of mitophagy) KO mouse tracheal epithelial cells (mTECs) or PINK1 KO mice were infected with IAV to reveal the role of mitophagy in viral infection. In IAV-infected mice, Tollip deficiency enhanced lung mitophagy (more PINK1 and BNIP3L, but less p62), and decreased release of mtDNA. Furthermore, Tollip deficiency suppressed STING activation and the antiviral response (eg IFN-β and MX1), and increased viral load. In IAV-infected Tollip KO mice, 2'3'cGAMP activated STING and increased antiviral response coupled with less virus. PINK1-deficient mice increased lung release of mtDNA and augmented STING activation and antiviral responses. PINK1 deficiency in mTECs increased STING activation and significantly decreased the viral load. Our findings suggest that enhanced mitophagy due to Tollip deficiency reduces mtDNA release and STING activation during viral infection, resulting in decreased antiviral responses. Reduction of mitophagy and/or STING activation may open novel avenues for therapeutic intervention in human subjects with Tollip deficiency and viral infection.
Collapse
Affiliation(s)
- Hamid Reza Nouri
- Department of Medicine, National Jewish Health, Denver, CO, United States
| | | | - Monica Kraft
- Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Mari Numata
- Department of Medicine, National Jewish Health, Denver, CO, United States
| | - Donata Vercelli
- Department of Cellular and Molecular Medicine, Asthma and Airway Disease Research Center, University of Arizona, Tucson, AZ, United States
| | - Hong Wei Chu
- Department of Medicine, National Jewish Health, Denver, CO, United States
| |
Collapse
|
20
|
Pan Y, Zhao X, Chen Q, Zhao T, Ma Y, Wu H, Xiang Y, Jiang P, Li W, Yan Q, Mao S, Tao Y, Wang L, Zhu Y, Xu G. Faecalibacterium Prausnitzii extracellular vesicles regulating macrophage differentiation via homologous recombination repair in colitis model. Microbiol Res 2025; 298:128217. [PMID: 40393169 DOI: 10.1016/j.micres.2025.128217] [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: 02/08/2025] [Revised: 05/05/2025] [Accepted: 05/08/2025] [Indexed: 05/22/2025]
Abstract
Inflammatory Bowel Disease (IBD) is characterized by chronic inflammation influenced by the depletion of beneficial gut microbiota, a critical factor in disease onset and progression. This study investigates the therapeutic potential of extracellular vesicles (EVs) derived from Faecalibacterium prausnitzii (F.p EVs), a commensal bacterium whose reduction is linked to IBD. Our research demonstrates that F.p EVs are preferentially taken up by macrophages, where they exert their anti-inflammatory effects through the enhancement of homologous recombination (HR) repair mechanisms. Specifically, F.p EVs upregulate the expression of key proteins involved in HR repair, such as BRCA1 and BRCA2, thereby reducing DNA damage and inhibiting the cGAS-STING pathway, which is central to the inflammatory response. This modulation of macrophage function results in decreased pro-inflammatory cytokine production and enhanced intestinal barrier integrity. By elucidating these mechanisms, our study provides a clear understanding of how F.p EVs can be used to target fundamental aspects of IBD pathology, laying the groundwork for the development of more effective and targeted therapies.
Collapse
Affiliation(s)
- Yinya Pan
- Department of Gastroenterology, Department of Pharmacy, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing 210008, China
| | - Xinlu Zhao
- Department of Gastroenterology, Nanjing Drum Tower Hospital, Affiliated Hospital of Nanjing Medical University, Nanjing 210008, China
| | - Qiongyun Chen
- Department of Gastroenterology, Taikang Xianlin Drum Tower Hospital, Nanjing, Jiangsu 21008, China
| | - Tao Zhao
- Department of Gastroenterology, Taikang Xianlin Drum Tower Hospital, Nanjing, Jiangsu 21008, China
| | - Yichun Ma
- Department of Gastroenterology, Nanjing Drum Tower Hospital Clinical College of Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, Jiangsu 21008, China
| | - Hai Wu
- Department of Gastroenterology, Department of Pharmacy, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing 210008, China
| | - Ying Xiang
- Department of Gastroenterology, Nanjing Drum Tower Hospital Clinical College of Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, Jiangsu 21008, China
| | - Ping Jiang
- Department of Gastroenterology, Department of Pharmacy, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing 210008, China
| | - Wenjun Li
- Department of Gastroenterology, Department of Pharmacy, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing 210008, China
| | - Qiong Yan
- Department of Gastroenterology, Nanjing Drum Tower Hospital Clinical College of Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, Jiangsu 21008, China
| | - Shangtao Mao
- Department of Gastroenterology, Department of Pharmacy, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing 210008, China
| | - Yufei Tao
- Department of Gastroenterology, Department of Pharmacy, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing 210008, China
| | - Lei Wang
- Department of Gastroenterology, Department of Pharmacy, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing 210008, China.
| | - Yun Zhu
- Department of Gastroenterology, Department of Pharmacy, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing 210008, China; Nanjing Medical Center for Clinical Pharmacy, Nanjing 210008, China.
| | - Guifang Xu
- Department of Gastroenterology, Department of Pharmacy, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing 210008, China; Department of Gastroenterology, Nanjing Drum Tower Hospital, Affiliated Hospital of Nanjing Medical University, Nanjing 210008, China; Department of Gastroenterology, Taikang Xianlin Drum Tower Hospital, Nanjing, Jiangsu 21008, China; Department of Gastroenterology, Nanjing Drum Tower Hospital Clinical College of Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, Jiangsu 21008, China.
| |
Collapse
|
21
|
Sun M, Yang Z, Tang F, Li F, Ye Q, Sun C, Liang J. Alcoholic cirrhosis-associated immune dysfunction: What does it imply for us? Ann Hepatol 2025:101927. [PMID: 40379022 DOI: 10.1016/j.aohep.2025.101927] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/14/2025] [Revised: 03/19/2025] [Accepted: 04/02/2025] [Indexed: 05/19/2025]
Abstract
Alcoholic cirrhosis is a leading cause of chronic advanced liver disease. With the gradual eradication of viral hepatitis and the rising levels of alcohol consumption, the incidence of alcoholic cirrhosis is expected to increase steadily. Alcohol is primarily metabolized in the gastrointestinal tract, producing toxic metabolites that enter the portal vein circulation and are subsequently transported to the liver. Excessive alcohol intake activates the microsomal ethanol oxidation system and disrupts the intestinal microbiota-driven microenvironment dictated by intestinal microbiota, and increase intestinal permeability, all of which trigger severe systemic inflammatory responses and impaired immune function. This phenomenon, known as cirrhosis-associated immune dysfunction (CAID), is closely linked to the severity of cirrhosis and can significantly influence disease progression, potentially leading to multi-organ failure. This narrative review sheds light on the relationship between alcoholic cirrhosis and CAID, focusing on tailored interventions to modify immune response and modulate gut microbiota composition in hopes of mitigating the development and deterioration of alcoholic cirrhosis.
Collapse
Affiliation(s)
- Mingyu Sun
- Department of Hepatology, Tianjin Key Laboratory of Extracorporeal Life Support for Critical Diseases, Nankai University Affiliated Third Center Hospital, Tianjin 300170, China
| | - Ziyi Yang
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Fei Tang
- Department of Hepatology, Tianjin Key Laboratory of Extracorporeal Life Support for Critical Diseases, Nankai University Affiliated Third Center Hospital, Tianjin 300170, China
| | - Fenghui Li
- Department of Hepatology, Tianjin Key Laboratory of Extracorporeal Life Support for Critical Diseases, Nankai University Affiliated Third Center Hospital, Tianjin 300170, China
| | - Qing Ye
- Department of Hepatology, Tianjin Key Laboratory of Extracorporeal Life Support for Critical Diseases, Nankai University Affiliated Third Center Hospital, Tianjin 300170, China
| | - Chao Sun
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Tianjin 300052, China; Department of Gastroenterology, Tianjin Medical University General Hospital Airport Hospital, Tianjin 300308, China.
| | - Jing Liang
- Department of Hepatology, Tianjin Key Laboratory of Extracorporeal Life Support for Critical Diseases, Nankai University Affiliated Third Center Hospital, Tianjin 300170, China.
| |
Collapse
|
22
|
Wang XT, Zhu X, Lian ZH, Liu Q, Yan HH, Qiu Y, Ge XY. AUP1 and UBE2G2 complex targets STING signaling and regulates virus-induced innate immunity. mBio 2025; 16:e0060225. [PMID: 40237449 PMCID: PMC12077101 DOI: 10.1128/mbio.00602-25] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2025] [Accepted: 03/18/2025] [Indexed: 04/18/2025] Open
Abstract
Stimulator of interferon genes (STING) is an endoplasmic reticulum (ER) signaling adaptor that is essential for the host immune response triggered by DNA pathogens. Precise regulation of STING is crucial for maintaining a balanced immune response and preventing harmful autoinflammation. Activation of STING requires its translocation from the ER to the Golgi apparatus. However, the mechanisms that maintain STING in its resting state remain largely unclear. Here, we find that deficiency of the ancient ubiquitous protein 1 (AUP1) causes spontaneous activation of STING and enhances the expression of type I interferons (IFNs) under resting conditions. Furthermore, deficiency of UBE2G2, a cofactor of AUP1, also promotes the abnormal activation of STING. AUP1 deficiency significantly enhances STING signaling induced by DNA virus, and AUP1 deficiency exhibits increased resistance to DNA virus infection in vitro and in vivo. Mechanistically, AUP1 may form a complex with UBE2G2 to interact with STING, preventing its exit from the ER membrane. Notably, infection with the RNA virus vesicular stomatitis virus (VSV) promotes the accumulation of lipid droplets (LDs) and AUP1 proteins. Additionally, AUP1 deficiency markedly inhibits the replication of VSV because AUP1 deficiency reduces lipid accumulation and alters the expression of lipid metabolism genes, such as carnitine palmitoyltransferase 1A (CPT1A), monoglyceride lipase (MGLL), and sterol regulatory element-binding transcription factor 1 (SREBF1). This study uncovers the essential roles of AUP1 in the STING signaling pathway and lipid metabolism pathway, highlighting its dual role in regulating virus replication.IMPORTANCEThe stimulator of interferon genes (STING) signaling cascade plays an essential role in coordinating innate immunity against DNA pathogens and autoimmunity. Precise regulation of the innate immune response is essential for maintaining homeostasis. In this study, we demonstrate that ancient ubiquitous protein 1 (AUP1) and UBE2G2 act as negative regulators of the innate immune response by targeting STING. Notably, AUP1 interacts with STING to retain STING in the endoplasmic reticulum (ER), preventing STING translocation and thereby limiting STING signaling in the resting state. In addition, deficiency of AUP1 markedly inhibits the replication of DNA virus and RNA virus. Our findings provide new insights into the regulation of STING signaling and confirm AUP1 has a dual role in regulating virus replication.
Collapse
Affiliation(s)
- Xin-Tao Wang
- Hunan Provincial Key Laboratory of Medical Virology, Institute of Pathogen Biology and Immunology, College of Biology, Hunan University, Changsha, Hunan, China
| | - Xi Zhu
- Hunan Provincial Key Laboratory of Medical Virology, Institute of Pathogen Biology and Immunology, College of Biology, Hunan University, Changsha, Hunan, China
| | - Zhong-Hao Lian
- Hunan Provincial Key Laboratory of Medical Virology, Institute of Pathogen Biology and Immunology, College of Biology, Hunan University, Changsha, Hunan, China
| | - Qian Liu
- Hunan Provincial Key Laboratory of Medical Virology, Institute of Pathogen Biology and Immunology, College of Biology, Hunan University, Changsha, Hunan, China
| | - Hui-Hui Yan
- Hunan Provincial Key Laboratory of Medical Virology, Institute of Pathogen Biology and Immunology, College of Biology, Hunan University, Changsha, Hunan, China
| | - Ye Qiu
- Hunan Provincial Key Laboratory of Medical Virology, Institute of Pathogen Biology and Immunology, College of Biology, Hunan University, Changsha, Hunan, China
| | - Xing-Yi Ge
- Hunan Provincial Key Laboratory of Medical Virology, Institute of Pathogen Biology and Immunology, College of Biology, Hunan University, Changsha, Hunan, China
| |
Collapse
|
23
|
Zhu L, Tang Z, Jiang W, Dong Y, Li X, Huang K, Wu T, Xu L, Guo W, Gu Y. Cholesterol biosynthesis induced by radiotherapy inhibits cGAS-STING activation and contributes to colorectal cancer treatment resistance. Exp Mol Med 2025:10.1038/s12276-025-01457-6. [PMID: 40355720 DOI: 10.1038/s12276-025-01457-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2024] [Revised: 02/04/2025] [Accepted: 03/10/2025] [Indexed: 05/14/2025] Open
Abstract
Radiotherapy-induced DNA damage can lead to apoptotic cell death and trigger an anti-tumor immune response via the cyclic GMP-AMP synthase-stimulator of interferon genes (cGAS-STING) pathway, which senses cytoplasmic double-stranded DNA. However, radiotherapy resistance poses a significant challenge in treating cancers, including colorectal cancer (CRC). Understanding the mechanisms underlying this resistance is crucial for developing effective therapies. Here we report that radiotherapy enhances cholesterol synthesis, which subsequently inhibits the cGAS-STING pathway, leading to radiotherapy resistance. Mechanistically, 3-hydroxy-3-methylglutaryl-CoA reductase (HMGCR) levels increase rapidly in response to radiation, resulting in increased cholesterol synthesis. This increased cholesterol sequesters STING in the endoplasmic reticulum, hindering its activation and downstream interferon signaling. Elevated HMGCR and cholesterol levels correlate with poor prognosis and reduced response to radiation therapy in patients with CRC. Importantly, pharmacological inactivation of HMGCR significantly enhanced radiotherapy responsiveness in animal models, dependent on cGAS-STING signaling-mediated anti-tumor responses. Our findings reveal that radiotherapy-induced cholesterol inhibits cGAS-STING signaling, facilitating tumor immune escape. Therefore, combining statins with radiotherapy represents a promising therapeutic strategy for treating CRC.
Collapse
Affiliation(s)
- Lijun Zhu
- Department of Oncology, The First Affiliated Hospital with Nanjing Medical University, The First Clinical Medical College of Nanjing Medical University, Nanjing Medical University, Nanjing, China
| | - Zhaohui Tang
- State Key Laboratory of Pharmaceutical Biotechnology, Department of Gastroenterology, Nanjing Drum Tower Hospital, School of Life Science, Nanjing University, Nanjing, China
| | - Wen Jiang
- Department of Oncology, The First Affiliated Hospital with Nanjing Medical University, The First Clinical Medical College of Nanjing Medical University, Nanjing Medical University, Nanjing, China
| | - Yuwen Dong
- Department of Oncology, The First Affiliated Hospital with Nanjing Medical University, The First Clinical Medical College of Nanjing Medical University, Nanjing Medical University, Nanjing, China
| | - Xiaofei Li
- Department of Oncology, The First Affiliated Hospital with Nanjing Medical University, The First Clinical Medical College of Nanjing Medical University, Nanjing Medical University, Nanjing, China
| | - Kai Huang
- State Key Laboratory of Pharmaceutical Biotechnology, Department of Gastroenterology, Nanjing Drum Tower Hospital, School of Life Science, Nanjing University, Nanjing, China
| | - Tiancong Wu
- Department of Gastroenterology and Hepatology, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China.
| | - Lingyan Xu
- Department of Oncology, The First Affiliated Hospital with Nanjing Medical University, The First Clinical Medical College of Nanjing Medical University, Nanjing Medical University, Nanjing, China.
| | - Wenjie Guo
- State Key Laboratory of Pharmaceutical Biotechnology, Department of Gastroenterology, Nanjing Drum Tower Hospital, School of Life Science, Nanjing University, Nanjing, China.
| | - Yanhong Gu
- Department of Oncology, The First Affiliated Hospital with Nanjing Medical University, The First Clinical Medical College of Nanjing Medical University, Nanjing Medical University, Nanjing, China.
| |
Collapse
|
24
|
Peng S, Hou X, Liu J, Huang F. Advances in polymer nanomaterials targeting cGAS-STING pathway for enhanced cancer immunotherapy. J Control Release 2025; 381:113560. [PMID: 40023225 DOI: 10.1016/j.jconrel.2025.02.056] [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: 09/13/2024] [Revised: 02/17/2025] [Accepted: 02/19/2025] [Indexed: 03/04/2025]
Abstract
Cyclic guanosine monophosphate-adenosine monophosphate synthase (cGAS)-stimulator of interferon genes (STING) pathway has been recognized as a promising target for cancer immunotherapy. Although various STING agonists have been developed, their clinical applications are still severely impeded by various issues, such as non-specific accumulation, adverse effects, rapid clearance, etc. In recent years, the emergence of nanomaterials has profoundly revolutionized STING agonists delivery, which promote tumor-targeted delivery, boost the immunotherapeutic effects and reduce systemic toxicity of STING agonists. In particular, polymer nanomaterials possess inherent advantages including controllable structure, tunable function and degradability. These properties afford them the capacity to serve as delivery vehicles for small-molecule STING agonists. Furthermore, the superior characteristics of polymer nanomaterials can enable their utilization as a novel STING agonist to stimulate anti-tumor immunity. In this review, the molecular mechanisms of cGAS-STING pathway activation are discussed. The recent development of small-molecules STING agonists is described. Then polymer nanomaterials are discussed as carriers for STING agonists in cancer immunotherapy, including polymersomes, polymer micelles, polymer capsules, and polymer nanogels. Additionally, polymer nanomaterials are identified as a novel class of STING agonists for efficient cancer immunotherapy, encompassing both polymer materials and polymer-STING agonists conjugates. The review also presents the combination of polymer-based cGAS-STING immunotherapy with chemotherapy, radiotherapy, phototherapy (both photodynamic and photothermal), chemodynamic therapy, and other therapeutic strategies. Furthermore, the discussion highlights recent advancements targeting the cGAS-STING pathway in clinically approved polymer nanomaterials and corresponding potent innovations. Finally, the potential challenges and perspectives of polymer nanomaterials for activating cGAS-STING pathway are outlined, emphasizing the critical scientific issue and hoping to offer guidance for their clinical translation.
Collapse
Affiliation(s)
- Shiyu Peng
- State Key Laboratory of Advanced Medical Materials and Devices, Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Key Laboratory of Radiopharmacokinetics for Innovative Drugs, Tianjin Institutes of Health Science, Institute of Radiation Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300192, China
| | - Xiaoxue Hou
- State Key Laboratory of Advanced Medical Materials and Devices, Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Key Laboratory of Radiopharmacokinetics for Innovative Drugs, Tianjin Institutes of Health Science, Institute of Radiation Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300192, China
| | - Jianfeng Liu
- State Key Laboratory of Advanced Medical Materials and Devices, Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Key Laboratory of Radiopharmacokinetics for Innovative Drugs, Tianjin Institutes of Health Science, Institute of Radiation Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300192, China.
| | - Fan Huang
- State Key Laboratory of Advanced Medical Materials and Devices, Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Key Laboratory of Radiopharmacokinetics for Innovative Drugs, Tianjin Institutes of Health Science, Institute of Radiation Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300192, China.
| |
Collapse
|
25
|
Grizzi F, Martin-Perez M, Rai G, Saeed AFUH, Raman A, Bordoloi D. Editorial: Innate immune pathways as targets for developing therapeutic intervention against human cancers. Front Immunol 2025; 16:1595279. [PMID: 40416962 PMCID: PMC12098504 DOI: 10.3389/fimmu.2025.1595279] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2025] [Accepted: 04/24/2025] [Indexed: 05/27/2025] Open
Affiliation(s)
- Fabio Grizzi
- Department of Immunology and Inflammation, IRCCS Humanitas Research Hospital, Milan, Italy
- Department of Biomedical Sciences, Humanitas University, Milan, Italy
| | - Miguel Martin-Perez
- Department of Cell Biology, Physiology and Immunology, University of Barcelona, Barcelona, Spain
| | - Geeta Rai
- Department of Molecular & Human Genetics, Institute of Science, Banaras Hindu University, Varanasi, India
| | | | - Ayush Raman
- National Cancer Institute, NIH, Bethesda, MD, United States
| | - Devivasha Bordoloi
- Department of Biological Sciences, Indian Institute of Science Education and Research (IISER) Bhopal, Bhopal, India
| |
Collapse
|
26
|
Losarwar S, Pancholi B, Babu R, Garabadu D. Mitochondria-dependent innate immunity: A potential therapeutic target in Flavivirus infection. Int Immunopharmacol 2025; 154:114551. [PMID: 40158432 DOI: 10.1016/j.intimp.2025.114551] [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: 01/16/2025] [Revised: 03/10/2025] [Accepted: 03/22/2025] [Indexed: 04/02/2025]
Abstract
Mitochondria, known as the powerhouse of cells, play a crucial role in host innate immunity during flavivirus infections such as Dengue, Zika, West Nile, and Japanese Encephalitis Virus. Mitochondrial antiviral signaling protein (MAVS) resides on the outer mitochondrial membrane which is triggered by viral RNA recognition by RIG-I-like receptors (RLRs). This activation induces IRF3 and NF-κB signaling, resulting in type I interferon (IFN) production and antiviral responses. Upon flavivirus infection, mitochondrial stress and dysfunction may lead to the release of mitochondrial DNA (mtDNA) into the cytoplasm, which serves as a damage-associated molecular pattern (DAMP). Cytosolic mtDNA is sensed by cGAS (cyclic GMP-AMP synthase), leading to the activation of the STING (Stimulator of Interferon Genes) pathway to increase IFN production and expand inflammation. Flaviviral proteins control mitochondrial morphology by controlling mitochondrial fission (MF) and fusion (MFu), disrupting mitochondrial dynamics (MD) to inhibit MAVS signaling and immune evasion. Flaviviral proteins also cause oxidative stress, resulting in the overproduction of reactive oxygen species (ROS), which triggers NLRP3 inflammasome activation and amplifies inflammation. Additionally, flaviviruses drive metabolic reprogramming by shifting host cell metabolism from oxidative phosphorylation (OxPhos) to glycolysis and fatty acid synthesis, creating a pro-replicative environment that supports viral replication and persistence. Thus, the present review explores the complex interaction between MAVS, mtDNA, and the cGAS-STING pathway, which is key to the innate immune response against flavivirus infections. Understanding these mechanisms opens new avenues in therapeutic interventions in targeting mitochondrial pathways to enhance antiviral immunity and mitigate viral infection.
Collapse
Affiliation(s)
- Saurabh Losarwar
- Department of Pharmacology, Central University of Punjab, Bhatinda 151401, India
| | | | - Raja Babu
- Department of Pharmacology, Central University of Punjab, Bhatinda 151401, India
| | - Debapriya Garabadu
- Department of Pharmacology, Central University of Punjab, Bhatinda 151401, India.
| |
Collapse
|
27
|
Hou S, Chang J, Xing C, Ye Z, Li W, Zhang Y, Zheng Z, Xiao J, Li S. Design, Synthesis, and Biological Evaluation of Selective STING Synergists That Enhance cGAMP-STING Pathway Activation without Inherent Agonist Activity. J Med Chem 2025; 68:9407-9430. [PMID: 40298091 DOI: 10.1021/acs.jmedchem.4c03131] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/30/2025]
Abstract
The cGAS-STING pathway is pivotal for innate immunity and antitumor responses. However, the challenge of selectively targeting the diseased tissue without harming the healthy tissue has impeded the development of STING agonists. In this article, we tackle this issue by developing novel STING synergists that target the STING C-terminal domain pocket. Our findings indicate that agonist 12B can boost the cGAMP-STING pathway synergistically. Through reverse optimization of 12B, we synthesized three series of compounds, with compounds 55, 66, and 67 emerging as selective STING synergists that amplify cGAMP-induced pathway activation without inherent agonist properties. Compound 67 emerged as the most potent (EC50 = 20.53 μM), displaying a broad binding affinity across STING-CTD alleles and potent antitumor efficacy in vivo. Notably, it exhibited excellent safety profiles in both in vitro and in vivo models, along with favorable pharmacokinetics. These findings highlight the therapeutic potential of novel STING synergists for cancer immunotherapy.
Collapse
Affiliation(s)
- Shi Hou
- State Key Laboratory of National Security Specially Needed Medicines, Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China
| | - Jiajia Chang
- State Key Laboratory of National Security Specially Needed Medicines, Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China
| | - Cheng Xing
- State Key Laboratory of National Security Specially Needed Medicines, Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China
| | - Ze Ye
- State Key Laboratory of National Security Specially Needed Medicines, Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China
| | - Wei Li
- State Key Laboratory of National Security Specially Needed Medicines, Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China
| | - Ying Zhang
- Department of Medicinal Chemistry, School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Zhibing Zheng
- State Key Laboratory of National Security Specially Needed Medicines, Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China
| | - Junhai Xiao
- State Key Laboratory of National Security Specially Needed Medicines, Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China
| | - Song Li
- State Key Laboratory of National Security Specially Needed Medicines, Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China
| |
Collapse
|
28
|
Chandramoorthy HC, Saleh RO, Altalbawy FMA, Mohammed JS, Ganesan S, Kundlas M, Premkumar J, Ray S, Mustafa YF, Abbas JK. Deciphering cGAS-STING signaling: implications for tumor immunity and hepatocellular carcinoma. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025:10.1007/s00210-025-04240-6. [PMID: 40332552 DOI: 10.1007/s00210-025-04240-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/12/2025] [Accepted: 04/28/2025] [Indexed: 05/08/2025]
Abstract
Hepatocellular carcinoma (HCC) is the most prevalent form of primary liver cancer and poses a significant global health challenge due to its rising incidence and associated mortality. Recent advancements in understanding the cytosolic DNA sensing, the cyclic GMP-AMP synthase-stimulator of interferon genes (cGAS-STING) pathway have illuminated its critical role in the immune response to HCC. This narrative review deciphers the multifaceted involvement of cGAS-STING in HCC, mainly its function in detecting cytosolic DNA and initiating type I interferon (IFN-I) responses, which are pivotal for antitumor immunity. This immune response is crucial for combating pathogens and can play a role in tumor surveillance. In the context of HCC, the tumor microenvironment (TME) can exhibit immune resistance, which complicates the effectiveness of therapies like immune checkpoint blockade. However, activation of the cGAS-STING pathway has been shown to stimulate antitumor immune responses, enhancing the activity of dendritic cells and cytotoxic T lymphocytes. There is ongoing research into STING agonists as a treatment strategy for HCC, with some studies indicating promising results in prolonging survival and enhancing the immune response against tumors. By summarizing current knowledge and identifying research gaps, this review aims to provide a comprehensive overview of cGAS-STING signaling in HCC and its future directions, emphasizing its potential as a therapeutic target in the fight against HCC. Understanding these mechanisms could pave the way for innovative immunotherapeutic approaches that enhance the efficacy of existing treatments and improve patient prognosis.
Collapse
Affiliation(s)
- Harish C Chandramoorthy
- Department of Microbiology & Clinical Parasitology, College of Medicine & Central Research Laboratories, King Khalid University, Abha, Saudi Arabia
| | - Raed Obaid Saleh
- Department of Medical Laboratories Techniques, College of Health and Medical Techniques, University of Al Maarif, Al Anbar, 31001, Iraq.
| | - Farag M A Altalbawy
- Department of Chemistry, University College of Duba, University of Tabuk, Tabuk, Saudi Arabia
| | - Jaafaru Sani Mohammed
- Medical Analysis Department, Faculty of Applied Science, Tishk International University, Erbil, Iraq
| | - Subbulakshmi Ganesan
- Department of Chemistry and Biochemistry, School of Sciences, JAIN (Deemed to Be University), Bangalore, Karnataka, India
| | - Mayank Kundlas
- Centre for Research Impact & Outcome, Chitkara University Institute of Engineering and Technology, Chitkara University, Rajpura, 140401, Punjab, India
| | - J Premkumar
- Department of Biomedical, Sathyabama Institute of Science and Technology, Chennai, Tamil Nadu, India
| | - Subhashree Ray
- Department of Biochemistry, IMS and SUM Hospital, Siksha 'O' Anusandhan (Deemed to Be University), Bhubaneswar, Odisha, 751003, India
| | - Yasser Fakri Mustafa
- Department of Pharmaceutical Chemistry, College of Pharmacy, University of Mosul, Mosul, 41001, Iraq
| | - Jamal K Abbas
- Department of Pharmaceutical, Al-Nisour University College, Nisour Seq. Karkh, Baghdad, Iraq
| |
Collapse
|
29
|
Zhu J, Ruan X, Mangione MC, Parra P, Chen G, Su X, Luo X, Cao DJ. The cGAS-STING pathway promotes acute ischemia-induced neutropoiesis and neutrophil priming in the bone marrow. Basic Res Cardiol 2025:10.1007/s00395-025-01111-2. [PMID: 40332608 DOI: 10.1007/s00395-025-01111-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/13/2025] [Revised: 04/10/2025] [Accepted: 04/15/2025] [Indexed: 05/08/2025]
Abstract
Our previous work demonstrated that the DNA-sensing receptor cyclic GMP-AMP synthase (cGAS) negatively affects post-infarct repair by promoting pro-inflammatory macrophages. However, whether cGAS and its downstream partner STING (Stimulator of Interferon Genes) regulate neutrophil production and function in the context of acute myocardial ischemia remains unclear. This study investigated the role of the cGAS-STING pathway in neutropoiesis (neutrophil production and differentiation) and examined whether ischemia primes neutrophils in the bone marrow via this pathway, enhancing their functionality and contributing to cardiac inflammatory injury. Using myocardial infarction (MI) models in wild-type (WT), Cgas-/-, and Sting-/- mice, we analyzed neutrophils from the bone marrow, peripheral blood, and infarcted tissue. Additionally, we generated neutrophil-specific conditional knockouts of Cgas and performed adoptive transfer experiments with Cgas-deficient neutrophils. RNA sequencing revealed that ischemia increased neutrophil production in the bone marrow and activated pathways involved in cytokine signaling, phagocytosis, chemotaxis, and degranulation. Inhibiting the cGAS-STING pathway reduced neutrophil production by decreasing lineage committed neutrophil precursors including early neutrophil precursors (eNP) and preNeu and downregulated ischemia-induced pathways. Neutrophil conditional Cgas deletion or adoptive transfer of Cgas-deficient neutrophils improved survival but did not significantly impact ischemia-induced remodeling. In conclusion, we demonstrate for the first time that ischemia enhanced neutrophil functionality before recruitment to infarcted tissue, and the cGAS-STING pathway played an important role in neutrophil production and priming. Furthermore, our findings demonstrate a neutrophil-specific role of cGAS in promoting cardiac rupture and mortality in MI. This study provides a more comprehensive understanding of the cGAS-STING pathway in acute ischemia and may support the translation of cGAS-STING modulators, an emerging therapeutic field.
Collapse
Affiliation(s)
- Jiankun Zhu
- Department of Internal Medicine, Cardiology Division, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Xinjia Ruan
- Research Center of Biostatistics and Computational Pharmacy, China Pharmaceutical University, Nanjing, 211198, People's Republic of China
| | - MariaSanta C Mangione
- Department of Internal Medicine, Cardiology Division, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Pablo Parra
- Department of Internal Medicine, Cardiology Division, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Guo Chen
- Department of Internal Medicine, Cardiology Division, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Xiaoping Su
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Xiang Luo
- Department of Internal Medicine, Cardiology Division, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Dian J Cao
- Department of Internal Medicine, Cardiology Division, University of Texas Southwestern Medical Center, Dallas, TX, USA.
- Department of Internal Medicine, Cardiology Division, Dallas VA Medical Center, Dallas, TX, USA.
| |
Collapse
|
30
|
Hyslop K, Ki KK, Naidoo R, O'Brien D, Prabhu A, Gill D, Anstey C, Rapchuk IL, McDonald CI, Marshall L, Pearse I, Fraser JF, Suen JY, Passmore MR. Cell-free mitochondrial DNA may predict the risk of post-operative complications and outcomes in surgical aortic valve replacement patients. Sci Rep 2025; 15:15857. [PMID: 40328845 PMCID: PMC12056145 DOI: 10.1038/s41598-025-00382-y] [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: 09/11/2024] [Accepted: 04/28/2025] [Indexed: 05/08/2025] Open
Abstract
Surgical aortic valve replacement (AVR) is the standard of care for severe aortic valve disease; however, post-operative complications continue to pose challenges. Inflammation caused by increased release of cell-free mitochondrial DNA (cf-mtDNA) during surgery may be a contributing factor. The aim of this pilot study was to investigate the relationship between cf-mtDNA release and the risk of inflammatory-mediated post-operative complications in adult AVR patients. Plasma was collected from patients undergoing an AVR with cardiopulmonary bypass (CPB) at baseline, intra-operatively and post ICU admission at 6, 12 and 18 h. Cf-mtDNA was quantified and inflammatory biomarker expression in leukocytes and plasma was assessed. Cf-mtDNA levels increased 16-fold at the end of CPB with a corresponding increase in average fragment size. Cf-mtDNA levels may be associated with post-operative bleeding, infection, hepatic failure and hospital length of stay. Gene expression and plasma analysis revealed altered levels of markers associated with inflammation and innate immune responses. Several markers were associated with post-operative complications and outcomes. Our results indicate that cf-mtDNA levels, including average fragment sizes, are increased following initiation of CPB in AVR patients, and may be associated with post-operative complications and outcomes. However, larger studies are needed to validate these findings.
Collapse
Affiliation(s)
- Kieran Hyslop
- Critical Care Research Group, The Prince Charles Hospital, Brisbane, Australia
- Faculty of Medicine, University of Queensland, Brisbane, Australia
| | - Katrina K Ki
- Critical Care Research Group, The Prince Charles Hospital, Brisbane, Australia
- Faculty of Medicine, University of Queensland, Brisbane, Australia
| | - Rishendran Naidoo
- Department of Cardiothoracic Surgery, The Prince Charles Hospital, Brisbane, Australia
| | - Donalee O'Brien
- Department of Cardiothoracic Surgery, The Prince Charles Hospital, Brisbane, Australia
| | - Anil Prabhu
- Department of Cardiothoracic Surgery, The Prince Charles Hospital, Brisbane, Australia
| | - Denzil Gill
- Faculty of Medicine, University of Queensland, Brisbane, Australia
- Adult Intensive Care Services, The Prince Charles Hospital, Brisbane, Australia
| | - Chris Anstey
- Faculty of Medicine, University of Queensland, Brisbane, Australia
- School of Medicine, Griffith University, Birtinya, Australia
| | - Ivan L Rapchuk
- Faculty of Medicine, University of Queensland, Brisbane, Australia
- Department of Anaesthesia and Perfusion, The Prince Charles Hospital, Brisbane, Australia
| | - Charles I McDonald
- Department of Anaesthesia and Perfusion, The Prince Charles Hospital, Brisbane, Australia
| | - Lachlan Marshall
- Department of Cardiothoracic Surgery, The Prince Charles Hospital, Brisbane, Australia
- Department of Cardiac Surgery, Princess Alexandra Hospital, Brisbane, Australia
| | - India Pearse
- Critical Care Research Group, The Prince Charles Hospital, Brisbane, Australia
| | - John F Fraser
- Critical Care Research Group, The Prince Charles Hospital, Brisbane, Australia
- Faculty of Medicine, University of Queensland, Brisbane, Australia
| | - Jacky Y Suen
- Critical Care Research Group, The Prince Charles Hospital, Brisbane, Australia
- Faculty of Medicine, University of Queensland, Brisbane, Australia
| | - Margaret R Passmore
- Critical Care Research Group, The Prince Charles Hospital, Brisbane, Australia.
- Faculty of Medicine, University of Queensland, Brisbane, Australia.
| |
Collapse
|
31
|
Kang S, Vu TH, Kim C, Truong AD, Hong YH. Exosomes derived from African swine fever virus-infected pigs mediate immune responses through NF-κB and JAK-STAT signaling pathways. Res Vet Sci 2025; 191:105687. [PMID: 40347599 DOI: 10.1016/j.rvsc.2025.105687] [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: 01/29/2024] [Revised: 10/14/2024] [Accepted: 05/03/2025] [Indexed: 05/14/2025]
Abstract
African swine fever (ASF) virus (ASFV) is an infectious disease that affects the pig industry, causing up to 85 % morbidity and 100 % mortality. To date, there are no available vaccines against ASFV. Exosomes are extracellular vesicles that are released from most cell types. Exosomes carry components such as nucleic acids, lipids, and proteins that play a vital role in cell-to-cell communication. This study investigated the effect of exosomes derived from the serum of ASFV-infected pigs on a porcine macrophage cell line. Exosomes derived from the serum of pigs infected with ASFV contained ASFV structural proteins (p30 and p72). Expression levels of interferon (IFN)-α, IFN-γ, IL-6, and CXCL8 in porcine macrophage cells were affected by exposure to exosomes derived from the serum of ASFV-infected pigs. Nuclear factor-κB (NF-κB) and Janus kinase/signal transducers and activators of transcription (JAK/STAT) signaling pathway play an important role in the immune response to ASFV infection. Exosomes derived from ASFV-infected pigs affected mRNA and protein levels of NF-κB, tank binding kinase 1 (TBK1), JAK1, JAK2, and STAT1, suggesting that exosomes derived from ASFV-infected pigs mediate antiviral response by modulating the expression of inflammatory cytokines and activity of the NF-κB and JAK-STAT signaling pathways. The present study provides novel information about the immunomodulatory effects of exosome derived from pigs infected with ASFV, improving our understanding of ASFV pathogenesis and the host immune response to ASFV infection.
Collapse
Affiliation(s)
- Suyeon Kang
- Department of Animal Science and Technology, Chung-Ang University, Anseong 17546, Republic of Korea
| | - Thi Hao Vu
- Department of Animal Science and Technology, Chung-Ang University, Anseong 17546, Republic of Korea
| | - Chaeeun Kim
- Department of Animal Science and Technology, Chung-Ang University, Anseong 17546, Republic of Korea
| | - Anh Duc Truong
- Department of Biochemistry and Immunology, National Institute of Veterinary Research, 86 Truong Chinh, Dong Da, Hanoi 100000, Viet Nam
| | - Yeong Ho Hong
- Department of Animal Science and Technology, Chung-Ang University, Anseong 17546, Republic of Korea.
| |
Collapse
|
32
|
Jeyachandran AV, Zaiss AK, Chakravarty N, Singh S, Delgado Y, Paravastu R, Satheeshkumar N, Gerald E, Jeysankar A, Thomas J, Fuller L, Lee N, Taylor C, Joshi S, Parcells M, French SW, Date A, Bouhaddou M, Garcia G, Kumar A, Damoiseaux R, Arumugaswami V. Drug screen reveals new potent host-targeted antivirals against Mpox virus. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.05.02.651913. [PMID: 40400715 PMCID: PMC12094526 DOI: 10.1101/2025.05.02.651913] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 05/29/2025]
Abstract
Mpox virus (MPXV), a re-emerging zoonotic threat, has caused outbreaks in non-endemic regions through respiratory, sexual, and close-contact transmission. The increased transmissibility of Clade IIb fueled the 2022 global outbreak, with 2024 Clade Ib spread in the Democratic Republic of Congo further escalating concern. Both outbreaks were declared public health emergencies by the WHO. Although tecovirimat (TPOXX) has been used off-label for Mpox, its limited effectiveness highlights the critical need for newer antivirals for MPXV. We conducted high-throughput antiviral drug screening using a host-directed kinase inhibitor library composed of 2,750 compounds against 2022 Clade IIb MPXV. Our primary screen identified 138 compounds preventing MPXV cytopathic effects, including multiple inhibitors of EGFR, PI3K-mTOR, and Ras/Raf, as well as apoptosis and autophagy regulators. Secondary and tertiary screenings yielded a shortlist of potent, nontoxic antiviral compounds that inhibited MPXV replication. Three selected compounds, IRAK4-IN-6, SM-7368, and KRAS inhibitor-10, reduced MPXV-induced cell death in primary human epidermal keratinocytes. IRAK4-IN-6 and SM-7368 were also found to modulate NF-κB and STING signaling. Furthermore, these compounds were found effective in reducing skin lesions and viral burden in a mouse model of MPXV skin infection. Together, our study reveals new classes of antiviral compounds against MPXV, offering promising candidates for future clinical development.
Collapse
Affiliation(s)
- Arjit Vijey Jeyachandran
- Department of Molecular and Medical Pharmacology, University of California, Los Angeles, CA, USA
| | - Anne K. Zaiss
- Department of Molecular and Medical Pharmacology, University of California, Los Angeles, CA, USA
- Department of Orthopaedic Surgery, University of California, Los Angeles, CA, USA
| | - Nikhil Chakravarty
- Department of Molecular and Medical Pharmacology, University of California, Los Angeles, CA, USA
- Department of Medicine, California University of Science and Medicine, Colton, CA, USA
| | - Sneha Singh
- Department of Ophthalmology, Visual and Anatomical Sciences, Wayne State University, Detroit, MI, USA
| | - Yennifer Delgado
- Department of Microbiology, Immunology, and Molecular Genetics, University of California, Los Angeles, CA, USA
- Institute for Quantitative and Computational Biosciences, University of California, Los Angeles, CA, USA
| | - Ramya Paravastu
- Department of Molecular and Medical Pharmacology, University of California, Los Angeles, CA, USA
| | - Nivedha Satheeshkumar
- Department of Molecular and Medical Pharmacology, University of California, Los Angeles, CA, USA
| | - Ephrem Gerald
- Department of Molecular and Medical Pharmacology, University of California, Los Angeles, CA, USA
| | - Aakash Jeysankar
- Department of Molecular and Medical Pharmacology, University of California, Los Angeles, CA, USA
| | - Joshua Thomas
- Department of Molecular and Medical Pharmacology, University of California, Los Angeles, CA, USA
| | - Lilly Fuller
- Department of Molecular and Medical Pharmacology, University of California, Los Angeles, CA, USA
| | - Noella Lee
- Department of Molecular and Medical Pharmacology, University of California, Los Angeles, CA, USA
| | - Cameron Taylor
- California NanoSystems Institute, University of California, Los Angeles, CA, USA
| | - Shantanu Joshi
- Ahmanson-Lovelace Brain Mapping Center, Department of Neurology, University of California, Los Angeles, CA, USA
| | - Mark Parcells
- Department of Animal and Food Sciences, Department of Biological Sciences, University of Delaware, Newark, DE 19716, USA
| | - Samuel W. French
- Department of Cellular and Molecular Pathology, University of California, Los Angeles, CA, USA
| | - Abhijit Date
- Department of Pharmacology and Toxicology, University of Arizona, Tucson, AZ, USA
| | - Mehdi Bouhaddou
- Department of Microbiology, Immunology, and Molecular Genetics, University of California, Los Angeles, CA, USA
- Institute for Quantitative and Computational Biosciences, University of California, Los Angeles, CA, USA
| | - Gustavo Garcia
- Department of Molecular and Medical Pharmacology, University of California, Los Angeles, CA, USA
| | - Ashok Kumar
- Department of Ophthalmology, Visual and Anatomical Sciences, Wayne State University, Detroit, MI, USA
| | - Robert Damoiseaux
- Department of Molecular and Medical Pharmacology, University of California, Los Angeles, CA, USA
- Department of Bioengineering, Samueli School of Engineering, University of California, Los Angeles, CA, USA
- Eli & Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, CA, USA
| | - Vaithilingaraja Arumugaswami
- Department of Molecular and Medical Pharmacology, University of California, Los Angeles, CA, USA
- California NanoSystems Institute, University of California, Los Angeles, CA, USA
- Eli & Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, CA, USA
| |
Collapse
|
33
|
Yang X, Lv L, Zhang Y, Zhang Z, Zeng S, Zhang X, Wang Q, Dorf M, Li S, Fu B. ATP2A2 regulates STING1/MITA-driven signal transduction including selective autophagy. Autophagy 2025:1-16. [PMID: 40265346 DOI: 10.1080/15548627.2025.2496786] [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: 11/25/2024] [Revised: 04/08/2025] [Accepted: 04/18/2025] [Indexed: 04/24/2025] Open
Abstract
STING1/MITA not only induces innate immune responses but also triggers macroautophagy/autophagy to selectively degrade signaling molecules. However, the molecular mechanisms regulating STING1-mediated selective autophagy remain unclear. Here, we first report that ATP2A2 directly interacts with STING1, regulating STING1-mediated innate immune response by modulating its polymerization and trafficking, thereby inhibiting DNA virus infection. Notably, while screening for reticulophagy receptors involved in STING1-mediated selective autophagy, we identified SEC62 as an important receptor protein in STING1-mediated reticulophagy. Mechanistically, SEC62 strengthens its interaction with STING1 upon activation and concurrently facilitates STING1-mediated reticulophagy upon starvation, which are dependent on ATP2A2. Furthermore, knocking down SEC62 in WT cells inhibits STING1-mediated MAP1LC3B/LC3B lipidation and autophagosome formation, an effect that is lost in ATP2A2 knockout cells, suggesting that SEC62's role in STING1-mediated selective autophagy is ATP2A2 dependent. Thus, our findings identify the reticulophagy receptor SEC62 as a novel receptor protein regulating STING1-mediated selective autophagy, providing new insight into the mechanism regarding a reticulophagy receptor in the process of STING1-induced selective autophagy.Abbrevations: aa: amino acids; AP-MS: affinity tag purification-mass spectrometry; ATP2A1: ATPase sarcoplasmic/endoplasmic reticulum Ca2+ transporting 1; ATP2A2: ATPase sarcoplasmic/endoplasmic reticulum Ca2+ transporting 2; ATP2A3: ATPase sarcoplasmic/endoplasmic reticulum Ca2+ transporting 3; CANX: calnexin; CCPG1: cell cycle progression 1; CGAS: cyclic GMP-AMP synthase; ctDNA: calf thymus DNA; dsRNA: double-stranded RNA; diABZI: diamidobenzimidazole; ER: endoplasmic reticulum; ERGIC: ER-Golgi intermediate compartment; EBSS: Earle's Balanced Salt Solution; EV: empty vector; FL: full length; GOLGA2/GM130: golgin A2; HSV-1: herpes simplex virus type 1; IRF3: interferon regulatory factor 3; IFNs: type I interferons; ISD: interferon stimulatory DNA; KO: knockout; MAVS: mitochondrial antiviral signaling protein; MOI: multiplicity of infection; poly(I:C): polyinosinic-polycytidylic acid; NBR1: NBR1 autophagy cargo receptor; PRR: pattern recognition receptor; reticulophagy: selective autophagic degradation of the ER; RETREG1/FAM134B: reticulophagy regulator 1; RIGI: RNA sensor RIG-I; RTN3L: reticulon 3; SEC62: SEC62 homolog, preprotein translocation factor; SeV: Sendai virus; STIM1: stromal interaction molecule 1; STING1/MITA: stimulator of interferon response cGAMP interactor 1; TBK1: TANK binding kinase 1; TEX264: testis expressed 264, ER-phagy receptor; TMX1: thioredoxin related transmembrane protein 1; VSV: vesicular stomatitis virus; VACV: vaccinia virus; ZMPSTE24: zinc metallopeptidase STE24.
Collapse
Affiliation(s)
- Xue Yang
- Department of Rheumatology and Immunology, State Key Laboratory of Virology and Biosafety, Zhongnan Hospital, Wuhan University, Wuhan, China
- Frontier Science Center for Immunology and Metabolism, Medical Research Institute, School of Medicine, Wuhan University, Wuhan, China
| | - Linyue Lv
- Department of Rheumatology and Immunology, State Key Laboratory of Virology and Biosafety, Zhongnan Hospital, Wuhan University, Wuhan, China
- Frontier Science Center for Immunology and Metabolism, Medical Research Institute, School of Medicine, Wuhan University, Wuhan, China
| | - Yuelan Zhang
- Department of Rheumatology and Immunology, State Key Laboratory of Virology and Biosafety, Zhongnan Hospital, Wuhan University, Wuhan, China
- Frontier Science Center for Immunology and Metabolism, Medical Research Institute, School of Medicine, Wuhan University, Wuhan, China
| | - Zhuyou Zhang
- Department of Rheumatology and Immunology, State Key Laboratory of Virology and Biosafety, Zhongnan Hospital, Wuhan University, Wuhan, China
- Frontier Science Center for Immunology and Metabolism, Medical Research Institute, School of Medicine, Wuhan University, Wuhan, China
| | - Shaowei Zeng
- Department of Rheumatology and Immunology, State Key Laboratory of Virology and Biosafety, Zhongnan Hospital, Wuhan University, Wuhan, China
- Frontier Science Center for Immunology and Metabolism, Medical Research Institute, School of Medicine, Wuhan University, Wuhan, China
| | - Xinyi Zhang
- Department of Rheumatology and Immunology, State Key Laboratory of Virology and Biosafety, Zhongnan Hospital, Wuhan University, Wuhan, China
- Frontier Science Center for Immunology and Metabolism, Medical Research Institute, School of Medicine, Wuhan University, Wuhan, China
| | - Qinyang Wang
- Department of Rheumatology and Immunology, State Key Laboratory of Virology and Biosafety, Zhongnan Hospital, Wuhan University, Wuhan, China
- Frontier Science Center for Immunology and Metabolism, Medical Research Institute, School of Medicine, Wuhan University, Wuhan, China
| | - Martin Dorf
- Department of Microbiology & Immunobiology, Harvard Medical School, Boston, Massachusetts, USA
| | - Shitao Li
- Department of Microbiology and Immunology, Tulane University, New Orleans, LA, USA
| | - Bishi Fu
- Department of Rheumatology and Immunology, State Key Laboratory of Virology and Biosafety, Zhongnan Hospital, Wuhan University, Wuhan, China
- Frontier Science Center for Immunology and Metabolism, Medical Research Institute, School of Medicine, Wuhan University, Wuhan, China
| |
Collapse
|
34
|
Bai X, Guo YR, Zhao ZM, Li XY, Dai DQ, Zhang JK, Li YS, Zhang CD. Macrophage polarization in cancer and beyond: from inflammatory signaling pathways to potential therapeutic strategies. Cancer Lett 2025; 625:217772. [PMID: 40324582 DOI: 10.1016/j.canlet.2025.217772] [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: 02/26/2025] [Revised: 04/30/2025] [Accepted: 05/02/2025] [Indexed: 05/07/2025]
Abstract
Macrophages are innate immune cells distributed throughout the body that play vital roles in organ development, tissue homeostasis, and immune surveillance. Macrophages acquire a binary M1/M2 polarized phenotype through signaling cascades upon sensing different signaling molecules in the environment, thereby playing a core role in a series of immune tasks, rendering precise regulation essential. M1/M2 macrophage phenotypes regulate inflammatory responses, while controlled activation of inflammatory signaling pathways is involved in regulating macrophage polarization. Among the relevant signaling pathways, we focus on the six well-characterized NF-κB, MAPK, JAK-STAT, PI3K/AKT, inflammasome, and cGAS-STING inflammatory pathways, and elucidate their roles and crosstalk in macrophage polarization. Furthermore, the effects of many environmental signals that influence macrophage polarization are investigated by modulating these pathways in vivo and in vitro. We thus detail the physiological and pathophysiological status of these six inflammatory signaling pathways and involvement in regulating macrophage polarization in cancer and beyond, as well as describe potential therapeutic approaches targeting these signaling pathways. In this review, the latest research advances in inflammatory signaling pathways regulating macrophage polarization are reviewed, as targeting these inflammatory signaling pathways provides suitable strategies to intervene in macrophage polarization and various tumor and non-tumor diseases.
Collapse
Affiliation(s)
- Xiao Bai
- Department of Surgical Oncology, The Fourth Affiliated Hospital of China Medical University, Shenyang 110032, China
| | - Yun-Ran Guo
- Health Sciences Institute of China Medical University, Shenyang 110122, China
| | - Zhe-Ming Zhao
- Department of Surgical Oncology, The Fourth Affiliated Hospital of China Medical University, Shenyang 110032, China
| | - Xin-Yun Li
- Clinical Medicine, The Fourth Affiliated Hospital of China Medical University, Shenyang 110032, China
| | - Dong-Qiu Dai
- Department of Surgical Oncology, The Fourth Affiliated Hospital of China Medical University, Shenyang 110032, China; Cancer Center, The Fourth Affiliated Hospital of China Medical University, Shenyang 110032, China.
| | - Jia-Kui Zhang
- Department of Surgical Oncology, The Fourth Affiliated Hospital of China Medical University, Shenyang 110032, China.
| | - Yong-Shuang Li
- Department of Surgical Oncology, The Fourth Affiliated Hospital of China Medical University, Shenyang 110032, China.
| | - Chun-Dong Zhang
- Department of Surgical Oncology, The Fourth Affiliated Hospital of China Medical University, Shenyang 110032, China; Central Laboratory, The Fourth Affiliated Hospital of China Medical University, Shenyang 110032, China.
| |
Collapse
|
35
|
Tao K, Tao K, Wang J. The potential mechanisms of extracellular vesicles in transfusion-related adverse reactions: Recent advances. Transfus Clin Biol 2025; 32:205-227. [PMID: 40180029 DOI: 10.1016/j.tracli.2025.03.006] [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: 01/15/2025] [Revised: 03/15/2025] [Accepted: 03/25/2025] [Indexed: 04/05/2025]
Abstract
Blood transfusion is an irreplaceable clinical treatment. Blood components are differentiated and stored according to specific guidelines. Storage temperatures and times vary depending on the blood component, but they all release extracellular vesicles (EVs) during storage. Although blood transfusions can be life-saving, they can also cause many adverse transfusion reactions, among which the effects of EVs are of increasing interest to researchers. EVs are submicron particles that vary in size, composition, and surface biomarkers, are encapsulated by a lipid bilayer, and are not capable of self-replication. EVs released by blood cells are important contributors to pathophysiologic states through proinflammatory, coagulant, and immunosuppressive effects, which in turn promote or inhibit the associated disease phenotype. Therefore, this review explores the potential mechanisms of hematopoietic-derived EVs in transfusion-associated adverse reactions and discusses the potential of the latest proteomics tools to be applied to the analysis of EVs in the field of transfusion medicine with a view to reducing the risk of blood transfusion.
Collapse
Affiliation(s)
- Keyi Tao
- Panzhihua University, Panzhihua 617000 Sichuan, China
| | - Keran Tao
- Institute of Medicine and Nursing, Hubei University of Medicine, Shiyan 442000 Hubei, China
| | - Jing Wang
- Southwest Medical University, Luzhou 646000 Sichuan, China; Department of Blood Transfusion, The Affiliated Hospital of Southwest Medical University, Luzhou Sichuan, 646000 China.
| |
Collapse
|
36
|
de Necochea Campion R, Pesqueira M, Vallejos P, McCullough C, Bloesch A, LaRosa SP. A lectin affinity plasmapheresis device removes extracellular vesicles and microRNAs from renal perfusates following controlled oxygenated rewarming of discarded donor kidneys. Transpl Immunol 2025; 90:102215. [PMID: 40024312 DOI: 10.1016/j.trim.2025.102215] [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: 11/12/2024] [Revised: 02/26/2025] [Accepted: 02/27/2025] [Indexed: 03/04/2025]
Abstract
Kidney transplantation is considered the benchmark treatment for end-stage kidney disease patients, yet the scarcity of suitable kidneys poses a significant hindrance for patients and healthcare providers. One approach is to extend the criteria for the use of kidneys from deceased brain death and deceased circulatory death donors. Use of these organs, especially from these extended criteria donors, is associated with ischemia reperfusion injury and resultant delayed graft function as well as increased rates of allograft rejection. To lessen these complications as well as increase the time of organ viability assessment, machine perfusion has been evaluated on recovered kidneys. In this study we examined the immunogenic molecular content of perfusates from discarded organs that had undergone Controlled Oxygenated Rewarming (COR). Perfusates were analyzed for extracellular vesicles (EVs), DNA (Deoxyribonucleic acid), and microRNAs. These perfusates were then pumped over a plasma separator containing a lectin affinity resin. Following treatment, a significant diminution in extracellular vesicles, dsDNA (double-stranded DNA) associated with EVs, and microRNAs (miRNA) were observed. Specifically, in three out of the four renal perfusates analyzed there was significant removal of small EVs (<200 nm) and vesicles loaded with dsDNA (p < 0.05). Notably, depletion of larger EVs (100-500 nm) was found to be significant in all treated perfusates (p < 0.01). NanoString analysis of miRNA found 5 species potentially involved in renal dysfunction (hsa-let 7a-5p, hsa-miR-148b-3p, hsa-miR-148a-3p, hsa-miR-29b-3pb and hsa-miR-99a5p) to be significantly depleted in treated renal perfusates (p ≤ 0.05). These results support a future study incorporating this treatment method into a dynamic machine perfusion circuit to explore if reduction of these mediators is associated with improved function of retrieved kidneys.
Collapse
Affiliation(s)
| | - Miguel Pesqueira
- Aethlon Medical Inc., 11555 Sorrento Valley Rd., San Diego, CA, United States of America
| | - Paul Vallejos
- Aethlon Medical Inc., 11555 Sorrento Valley Rd., San Diego, CA, United States of America
| | - Cameron McCullough
- Aethlon Medical Inc., 11555 Sorrento Valley Rd., San Diego, CA, United States of America
| | - Alessio Bloesch
- Aethlon Medical Inc., 11555 Sorrento Valley Rd., San Diego, CA, United States of America
| | - Steven P LaRosa
- Aethlon Medical Inc., 11555 Sorrento Valley Rd., San Diego, CA, United States of America
| |
Collapse
|
37
|
Zhang M, Ji Y, Liu M, Dai Y, Zhang H, Tong S, Cai Y, Liu M, Qu N. Nano-delivery of STING agonists: Unraveling the potential of immunotherapy. Acta Biomater 2025; 197:104-120. [PMID: 40164370 DOI: 10.1016/j.actbio.2025.03.054] [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: 12/24/2024] [Revised: 03/10/2025] [Accepted: 03/28/2025] [Indexed: 04/02/2025]
Abstract
The cyclic GMP-AMP synthetase-interferon gene stimulator (cGAS-STING) pathway possesses tremendous potential in immune responses, viral defense, and anti-tumor treatment. Currently, an increasing number of nanocarriers are being engineered to convey STING agonists, with the goal of booSTING the conveying capacity of cGAS-STING agonists and augment the therapeutic potency of STING agonists. In this review, we explore the mechanisms of cGAS-STING activators, the application of different nanocarriers in the STING pathway, and the application of nanocarriers in anti-tumor therapy, antiviral therapy and autoimmune diseases. Additionally, we also discuss the adverse effects of STING pathway activation and the challenges encountered in nano delivery, we hope that future research will delve into the development of new nanocarriers and the clinical translation of nanocarriers in STING-mediated immunotherapy. STATEMENT OF SIGNIFICANCE: The cyclic GMP-AMP synthetase-interferon gene stimulator (cGAS-STING) pathway possesses tremendous potential in immune responses, viral defense, and anti-tumor treatment. In this review, we first explore the activation mechanism of cGAS-STING signal pathway and the diverse array of nanocarriers that have been employed in the context of the STING pathway, such as natural carrier, lipid nanoparticles, polymeric nanoparticles, and inorganic nanoparticles, highlighting their unique properties and the challenges they present in clinical applications. Furthermore, we discuss the research progress regarding nanocarriers in STING-mediated immunotherapy, such as the application of nanocarriers in anti-tumor therapy, antiviral therapy and autoimmune diseases therapy. Finally, the side effects of STING pathway activation and the issues encountered in nano delivery will be discussed, hoping that future research will delve into the development of new nanocarriers and the clinical translation of nanocarriers in STING-mediated immunotherapy.
Collapse
Affiliation(s)
- Meng Zhang
- School of Pharmaceutical Science, Liaoning University, Shenyang 110036, PR China
| | - Yating Ji
- School of Pharmaceutical Science, Liaoning University, Shenyang 110036, PR China
| | - Mingxia Liu
- School of Pharmaceutical Science, Liaoning University, Shenyang 110036, PR China
| | - Yixin Dai
- School of Pharmaceutical Science, Liaoning University, Shenyang 110036, PR China
| | - Hongxia Zhang
- School of Pharmaceutical Science, Liaoning University, Shenyang 110036, PR China
| | - Shiyu Tong
- School of Pharmaceutical Science, Liaoning University, Shenyang 110036, PR China
| | - Yuqing Cai
- School of Pharmaceutical Science, Liaoning University, Shenyang 110036, PR China
| | - Mengjiao Liu
- Institute for Experimental Molecular Imaging, RWTH Aachen University Clinic, Forckenbeckstrasse 55, Aachen 52074, Germany
| | - Na Qu
- School of Pharmaceutical Science, Liaoning University, Shenyang 110036, PR China.
| |
Collapse
|
38
|
Wang C, Lin X, Guan S, Wu Q, Liang S. Dihydroartemisinin Attenuates Radiation-Induced Lung Injury by Inhibiting the cGAS/STING/NF-κB Signaling Pathway. Drug Dev Res 2025; 86:e70090. [PMID: 40285509 PMCID: PMC12032573 DOI: 10.1002/ddr.70090] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2025] [Revised: 03/26/2025] [Accepted: 04/12/2025] [Indexed: 04/29/2025]
Abstract
Dihydroartemisinin (DHA) is a derivative of artemisinin, which affects inflammation, oxidative stress, and immune regulation. However, the mechanism underlying its effects remains largely unknown. This study aims to explore the mechanism by which DHA affects radiation-induced lung injury (RILI), providing new insights for lung radiotherapy. To elucidate its mechanism of action, C57BL/6 J mice were irradiated with 15 Gy whole chest. RILI was evaluated by qRT-PCR, ELISA, histology, Western blot analysis, immunohistochemistry, and RILI signaling cascade studies. In addition, small interfering RNAs were employed to knockdown cGAS proteins in the cGAS-STING signaling pathway in the human bronchial epithelium cell line (BEAS-2B). Both In Vivo and Vitro experiments were conducted to investigate the specific mechanism by which DHA alleviated RILI. We observed the activation of the cGAS-STING pathway, along with the phosphorylation of the downstream target NF-κB and an increase in inflammatory factor levels in the mouse model following radiation exposure. In the cell model, irradiation also triggered the activation of the cGAS-STING signaling pathway and its downstream targets, leading to elevated levels of inflammatory factors. Notably, knocking down the cGAS using small interfering RNA in the BEAS-2B cells significantly alleviated RILI in the cell model. Our study elucidated the mechanism of DHA reducing RILI through the cGAS/STING/NF-κB signaling pathway, and revealed that the GAS/STING/NF-κB axis may be a potential therapeutic target for RILI.
Collapse
Affiliation(s)
- Cailan Wang
- Department of Radiation OncologyGuangxi Medical University Cancer HospitalNanningChina
| | - Xinyi Lin
- Department of Radiation OncologyGuangxi Medical University Cancer HospitalNanningChina
| | - Shichun Guan
- Department of Radiation OncologyGuangxi Medical University Cancer HospitalNanningChina
| | - Qiaoyuan Wu
- Department of Radiation OncologyGuangxi Medical University Cancer HospitalNanningChina
| | - Shixiong Liang
- Department of Radiation OncologyGuangxi Medical University Cancer HospitalNanningChina
| |
Collapse
|
39
|
Sharma M, Wagh P, Shinde T, Trimbake D, Tripathy AS. Exploring the Role of Pattern Recognition Receptors as Immunostimulatory Molecules. Immun Inflamm Dis 2025; 13:e70150. [PMID: 40396589 DOI: 10.1002/iid3.70150] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Revised: 12/09/2024] [Accepted: 01/21/2025] [Indexed: 05/22/2025] Open
Abstract
BACKGROUND Pattern recognition receptors (PRRs) are the receptors of the innate immune system that play a vital role in initiating innate immune response. PRRs recognize pathogen associated molecular patterns (PAMPs) and activate immune cells through a signaling cascade. Due to this remarkable ability to recognize pathogenic microbes and elucidation of an immune response in a well-organized manner, PRR agonizts are likely to have great potential as vaccine adjuvants. Recent advancements in vaccine development raised concerns regarding the reduced immunogenicity of various vaccines, questioning the vaccine efficacy. In such cases, the use of an adjuvant becomes crucial. Understanding the structure and downstream signaling of PRRs will provide the possibility of developing a novel therapeutic approach. METHOD The rapidly evolving field of immunology and vaccinology, coupled with the increasing focus on PRRs in disease therapy, demands a comprehensive overview. In this review, we provide all-inclusive and contemporary gist on PRRs and the applications of their agonizts. We explored the potential of PRR agonizts as vaccine adjuvant. The current review integrates the basic understanding of PRRs and recent findings highlighting emerging trends of the same. RESULT Our review highlights that combining multiple PRR agonizts could offer synergistic benefits. This approach might prove advantageous and could potentially enhance vaccine efficacy and reduce the need for excessive immunogens. CONCLUSION A comprehensive understanding of PRR subset, agonizts of PRR and their application in vaccine adjuvant. This knowledge will be significant in formulating vaccine approaches.
Collapse
Affiliation(s)
- Meenal Sharma
- Department of Dengue and Chikungunya, Indian Council of Medical Research-National Institute of Virology, Pune, India
| | - Priyanka Wagh
- Department of Dengue and Chikungunya, Indian Council of Medical Research-National Institute of Virology, Pune, India
| | - Tanvi Shinde
- Department of Dengue and Chikungunya, Indian Council of Medical Research-National Institute of Virology, Pune, India
| | - Diptee Trimbake
- Department of Dengue and Chikungunya, Indian Council of Medical Research-National Institute of Virology, Pune, India
| | - Anuradha S Tripathy
- Department of Dengue and Chikungunya, Indian Council of Medical Research-National Institute of Virology, Pune, India
| |
Collapse
|
40
|
Yuan Y, Li Q, Yan G, Qian Y, Guo W, Li S, Wang F, Shang W, Zhu Z, Ge D, Wang Y, Liu Y. Targeting KMT5C Suppresses Lung Cancer Progression and Enhances the Efficacy of Immunotherapy. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2407575. [PMID: 40126333 PMCID: PMC12097080 DOI: 10.1002/advs.202407575] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/05/2024] [Revised: 02/24/2025] [Indexed: 03/25/2025]
Abstract
The immune evasion is one major challenge for cancer immunotherapy. Despite considerable advancements in immune checkpoint blockade (ICB) therapies for the advanced non-small cell lung cancer (NSCLC) patients, only a minority of patients receive long-term survival benefit. Here, this work demonstrates that lysine methyltransferase 5C (KMT5C) is a crucial promoter of the NSCLC progression and immune evasion. This work first observes that upregulation of KMT5C in NSCLC correlated with cancer progression and poor patient prognosis. Notably, KMT5C knockdown in NSCLC cells suppress tumor growth and metastasis in mice. Mechanistically, this work demonstrates that KMT5C activated the DNA repair response to inhibit the STING-IRF3 pathway, downstream type I IFN signaling, and CCL5 secretion, leading to the downregulation of CD8+ T cell infiltration and function in NSCLC, ultimately facilitating tumor immune evasion and tumor progression. Importantly, both the pharmacological inhibitor A196 and the genetic inhibition of KMT5C could synergize with anti-PD-1 therapy in the lung cancer mouse model. Clinically, high expression levels of KMT5C in patients with NSCLC are associated with a lower response rate and worse clinical outcomes to ICB therapy. Therefore, these findings identify a previously unknown functional link between KMT5C and tumor immune evasion, and demonstrate that targeting KMT5C may be a potential therapeutic approach for enhancing the efficacy of NSCLC patients to ICB therapy.
Collapse
Affiliation(s)
- Yunfeng Yuan
- Department of Thoracic SurgeryZhongshan HospitalFudan UniversityShanghai200032China
| | - Qianyu Li
- Department of Liver SurgeryClinical Stem Cell Research CenterRen Ji HospitalShanghai Jiao Tong University School of MedicineShanghai200127China
| | - Guoquan Yan
- Institute of Biomedical SciencesShanghai Medical CollegeFudan UniversityShanghai200032China
| | - Yifei Qian
- Department of Liver SurgeryClinical Stem Cell Research CenterRen Ji HospitalShanghai Jiao Tong University School of MedicineShanghai200127China
| | - Wenyun Guo
- Department of Liver SurgeryClinical Stem Cell Research CenterRen Ji HospitalShanghai Jiao Tong University School of MedicineShanghai200127China
| | - Songling Li
- Department of Liver SurgeryClinical Stem Cell Research CenterRen Ji HospitalShanghai Jiao Tong University School of MedicineShanghai200127China
| | - Fan Wang
- Department of Liver SurgeryClinical Stem Cell Research CenterRen Ji HospitalShanghai Jiao Tong University School of MedicineShanghai200127China
| | - Wanjing Shang
- Lymphocyte Biology SectionLaboratory of Immune System BiologyNational Institute of Allergy and infectious DiseasesNational Institutes of HealthBethesdaMD20814USA
| | - Zijun Zhu
- Department of Liver SurgeryClinical Stem Cell Research CenterRen Ji HospitalShanghai Jiao Tong University School of MedicineShanghai200127China
| | - Di Ge
- Department of Thoracic SurgeryZhongshan HospitalFudan UniversityShanghai200032China
| | - Yanan Wang
- Department of Laboratory MedicineRen Ji HospitalShanghai Jiao Tong University School of MedicineShanghai200127China
| | - Yanfeng Liu
- Department of Liver SurgeryClinical Stem Cell Research CenterRen Ji HospitalShanghai Jiao Tong University School of MedicineShanghai200127China
- Shanghai Engineering Research Center of Transplantation and ImmunologyShanghai Institute of TransplantationShanghai200127China
| |
Collapse
|
41
|
Yu B, Zhang W, Shao Z, Chen X, Cen Y, Liu Y, Chen Y, Li X, Liang Z, Li S, Chen X. Self-promoted tumor-targeting nanomedicine activates STING-driven antitumor immunity via photodynamic DNA damage and PARP inhibition. Chem Sci 2025:d5sc01953b. [PMID: 40321187 PMCID: PMC12044612 DOI: 10.1039/d5sc01953b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2025] [Accepted: 04/12/2025] [Indexed: 05/08/2025] Open
Abstract
The activation of antitumor immunity through strategically designed nanomedicine presents a promising approach to overcome the limitations of conventional cancer therapies. In this work, bioinformatic analysis found an abnormal poly(ADP-ribose) polymerase-1 (PARP-1) expression in breast cancer, linked to the cyclic GMP-AMP synthase (cGAS)-stimulator of the interferon gene (STING) pathway and immune suppression. PARP-1 inhibitor screening revealed olaparib (Ola) as a promising candidate, enhancing DNA damage and potentiating the immunotherapeutic response. Consequently, a self-promoted tumor-targeting nanomedicine (designated as PN-Ola) was proposed to activate STING-driven antitumor immunity through photodynamic DNA damage and PARP inhibition. PN-Ola was composed of a programmed death-ligand 1 (PD-L1) targeting amphiphilic peptide-photosensitizer conjugate (C16-K(PpIX)-WHRSYYTWNLNT), which effectively encapsulates Ola. Notably, PN-Ola demonstrated selective accumulation in tumor cells that overexpress PD-L1, while concurrently enhancing PD-L1 expression, thereby establishing a self-promoting mechanism for improved drug accumulation within tumor cells. Meanwhile, the photodynamic therapy (PDT) effects of PN-Ola would result in oxidative DNA damage and subsequent accumulation of DNA fragments. Additionally, the PARP inhibition provided by PN-Ola disrupted the DNA repair pathways in tumor cells, leading to a boosted release of DNA fragments that further stimulated STING-driven antitumor immunity. The synergistic mechanism of PN-Ola effectively activates the immunotherapeutic response by enhancing T cell activation and infiltration, leading to the eradication of metastatic tumors without inducing side effects. This study presents a promising strategy to overcome targeting ligand heterogeneity while activating systemic antitumor immunity for the effective eradication of metastatic tumors.
Collapse
Affiliation(s)
- Baixue Yu
- The Fifth Affiliated Hospital, Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, The NMPA and State Key Laboratory of Respiratory Disease, The School of Pharmaceutical Sciences, Guangzhou Medical University Guangzhou 511436 P. R. China
| | - Wei Zhang
- The Fifth Affiliated Hospital, Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, The NMPA and State Key Laboratory of Respiratory Disease, The School of Pharmaceutical Sciences, Guangzhou Medical University Guangzhou 511436 P. R. China
| | - Zhouchuan Shao
- The Fifth Affiliated Hospital, Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, The NMPA and State Key Laboratory of Respiratory Disease, The School of Pharmaceutical Sciences, Guangzhou Medical University Guangzhou 511436 P. R. China
| | - Xiayun Chen
- The Fifth Affiliated Hospital, Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, The NMPA and State Key Laboratory of Respiratory Disease, The School of Pharmaceutical Sciences, Guangzhou Medical University Guangzhou 511436 P. R. China
| | - Yi Cen
- The Fifth Affiliated Hospital, Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, The NMPA and State Key Laboratory of Respiratory Disease, The School of Pharmaceutical Sciences, Guangzhou Medical University Guangzhou 511436 P. R. China
| | - Yibin Liu
- The Fifth Affiliated Hospital, Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, The NMPA and State Key Laboratory of Respiratory Disease, The School of Pharmaceutical Sciences, Guangzhou Medical University Guangzhou 511436 P. R. China
| | - Ying Chen
- The Fifth Affiliated Hospital, Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, The NMPA and State Key Laboratory of Respiratory Disease, The School of Pharmaceutical Sciences, Guangzhou Medical University Guangzhou 511436 P. R. China
| | - Xinxuan Li
- The Fifth Affiliated Hospital, Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, The NMPA and State Key Laboratory of Respiratory Disease, The School of Pharmaceutical Sciences, Guangzhou Medical University Guangzhou 511436 P. R. China
| | - Ziqi Liang
- The Fifth Affiliated Hospital, Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, The NMPA and State Key Laboratory of Respiratory Disease, The School of Pharmaceutical Sciences, Guangzhou Medical University Guangzhou 511436 P. R. China
| | - Shiying Li
- The Fifth Affiliated Hospital, Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, The NMPA and State Key Laboratory of Respiratory Disease, The School of Pharmaceutical Sciences, Guangzhou Medical University Guangzhou 511436 P. R. China
- Department of Diagnostic Radiology, Yong Loo Lin School of Medicine, National University of Singapore 119074 Singapore
| | - Xiaoyuan Chen
- Department of Diagnostic Radiology, Yong Loo Lin School of Medicine, National University of Singapore 119074 Singapore
- Department of Chemical and Biomolecular Engineering, College of Design and Engineering, National University of Singapore 117575 Singapore
- Department of Biomedical Engineering, College of Design and Engineering, National University of Singapore 117575 Singapore
- Department of Pharmacy and Pharmaceutical Sciences, Faculty of Science, National University of Singapore 117544 Singapore
- Clinical Imaging Research Centre, Centre for Translational Medicine, Yong Loo Lin School of Medicine, National University of Singapore 117599 Singapore
- Nanomedicine Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore 117597 Singapore
- Theranostics Center of Excellence (TCE), Yong Loo Lin School of Medicine, National University of Singapore 138667 Singapore
- Institute of Molecular and Cell Biology, Agency for Science, Technology, and Research (A*STAR) 138673 Singapore
| |
Collapse
|
42
|
Wang X, Xu P, Bentley-DeSousa A, Hancock-Cerutti W, Cai S, Johnson BT, Tonelli F, Shao L, Talaia G, Alessi DR, Ferguson SM, De Camilli P. The bridge-like lipid transport protein VPS13C/PARK23 mediates ER-lysosome contacts following lysosome damage. Nat Cell Biol 2025; 27:776-789. [PMID: 40211074 DOI: 10.1038/s41556-025-01653-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Accepted: 03/06/2025] [Indexed: 04/12/2025]
Abstract
Based on genetic studies, lysosome dysfunction is thought to play a pathogenetic role in Parkinson's disease. Here we show that VPS13C, a bridge-like lipid-transport protein and a Parkinson's disease gene, is a sensor of lysosome stress or damage. Following lysosome membrane perturbation, VPS13C rapidly relocates from the cytosol to the surface of lysosomes where it tethers their membranes to the ER. This recruitment depends on Rab7 and requires a signal at the damaged lysosome surface that releases an inhibited state of VPS13C, which hinders access of its VAB domain to lysosome-bound Rab7. Although another Parkinson's disease protein, LRRK2, is also recruited to stressed or damaged lysosomes, its recruitment occurs at much later stages and by different mechanisms. Given the role of VPS13 proteins in bulk lipid transport, these findings suggest that lipid delivery to lysosomes by VPS13C is part of an early protective response to lysosome damage.
Collapse
Affiliation(s)
- Xinbo Wang
- Department of Neuroscience, Yale University School of Medicine, New Haven, CT, USA
- Department of Cell Biology, Yale University School of Medicine, New Haven, CT, USA
- Howard Hughes Medical Institute, Yale University School of Medicine, New Haven, CT, USA
- Program in Cellular Neuroscience, Neurodegeneration, and Repair, Yale University School of Medicine, New Haven, CT, USA
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, USA
| | - Peng Xu
- Department of Neuroscience, Yale University School of Medicine, New Haven, CT, USA
- Department of Cell Biology, Yale University School of Medicine, New Haven, CT, USA
- Howard Hughes Medical Institute, Yale University School of Medicine, New Haven, CT, USA
- Program in Cellular Neuroscience, Neurodegeneration, and Repair, Yale University School of Medicine, New Haven, CT, USA
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, USA
| | - Amanda Bentley-DeSousa
- Department of Neuroscience, Yale University School of Medicine, New Haven, CT, USA
- Department of Cell Biology, Yale University School of Medicine, New Haven, CT, USA
- Program in Cellular Neuroscience, Neurodegeneration, and Repair, Yale University School of Medicine, New Haven, CT, USA
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, USA
| | - William Hancock-Cerutti
- Department of Neuroscience, Yale University School of Medicine, New Haven, CT, USA
- Department of Cell Biology, Yale University School of Medicine, New Haven, CT, USA
- Howard Hughes Medical Institute, Yale University School of Medicine, New Haven, CT, USA
- Program in Cellular Neuroscience, Neurodegeneration, and Repair, Yale University School of Medicine, New Haven, CT, USA
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, USA
| | - Shujun Cai
- Department of Neuroscience, Yale University School of Medicine, New Haven, CT, USA
- Department of Cell Biology, Yale University School of Medicine, New Haven, CT, USA
- Howard Hughes Medical Institute, Yale University School of Medicine, New Haven, CT, USA
- Program in Cellular Neuroscience, Neurodegeneration, and Repair, Yale University School of Medicine, New Haven, CT, USA
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, USA
| | - Benjamin T Johnson
- Department of Neuroscience, Yale University School of Medicine, New Haven, CT, USA
- Department of Cell Biology, Yale University School of Medicine, New Haven, CT, USA
- Howard Hughes Medical Institute, Yale University School of Medicine, New Haven, CT, USA
- Program in Cellular Neuroscience, Neurodegeneration, and Repair, Yale University School of Medicine, New Haven, CT, USA
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, USA
| | - Francesca Tonelli
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, USA
- MRC Protein Phosphorylation and Ubiquitylation Unit, School of Life Sciences, University of Dundee, Dundee, UK
| | - Lin Shao
- Center for Neurodevelopment and Plasticity, Wu Tsai Institute, Yale University, New Haven, CT, USA
| | - Gabriel Talaia
- Department of Neuroscience, Yale University School of Medicine, New Haven, CT, USA
- Department of Cell Biology, Yale University School of Medicine, New Haven, CT, USA
- Program in Cellular Neuroscience, Neurodegeneration, and Repair, Yale University School of Medicine, New Haven, CT, USA
| | - Dario R Alessi
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, USA
- MRC Protein Phosphorylation and Ubiquitylation Unit, School of Life Sciences, University of Dundee, Dundee, UK
| | - Shawn M Ferguson
- Department of Neuroscience, Yale University School of Medicine, New Haven, CT, USA
- Department of Cell Biology, Yale University School of Medicine, New Haven, CT, USA
- Program in Cellular Neuroscience, Neurodegeneration, and Repair, Yale University School of Medicine, New Haven, CT, USA
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, USA
| | - Pietro De Camilli
- Department of Neuroscience, Yale University School of Medicine, New Haven, CT, USA.
- Department of Cell Biology, Yale University School of Medicine, New Haven, CT, USA.
- Howard Hughes Medical Institute, Yale University School of Medicine, New Haven, CT, USA.
- Program in Cellular Neuroscience, Neurodegeneration, and Repair, Yale University School of Medicine, New Haven, CT, USA.
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, USA.
| |
Collapse
|
43
|
Khemraj P, Kuznyetsova A, Hood DA. Adaptations in mitochondrial quality control and interactions with innate immune signaling within skeletal muscle: A narrative review. JOURNAL OF SPORT AND HEALTH SCIENCE 2025:101049. [PMID: 40318804 DOI: 10.1016/j.jshs.2025.101049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/03/2024] [Revised: 02/14/2025] [Accepted: 03/17/2025] [Indexed: 05/07/2025]
Abstract
Skeletal muscle health and function are essential determinants of metabolic health, physical performance, and overall quality of life. The quality of skeletal muscle is heavily dependent on the complex mitochondrial reticulum that contributes toward its unique adaptability. It is now recognized that mitochondrial perturbations can activate various innate immune pathways, such as the nucleotide-binding oligomerization domain (NOD)-like receptor protein 3 (NLRP3) inflammasome complex by propagating inflammatory signaling in response to damage-associated molecular patterns (DAMPs). The NLRP3 inflammasome is a multimeric protein complex and is a prominent regulator of innate immunity and cell death by mediating the activation of caspase-1, pro-inflammatory cytokines interleukin-1β and interleukin-18 and pro-pyroptotic protein gasdermin-D. While several studies have begun to demonstrate the relationship between various mitochondrial DAMPs (mtDAMPs) and NLRP3 inflammasome activation, the influence of various metabolic states on the production of these DAMPs and subsequent inflammatory profile remains poorly understood. This narrative review aimed to address this by highlighting the effects of skeletal muscle use and disuse on mitochondrial quality mechanisms including mitochondrial biogenesis, fusion, fission and mitophagy. Secondly, this review summarized the impact of alterations in mitochondrial quality control mechanisms following muscle denervation, aging, and exercise training in relation to NLRP3 inflammasome activation. By consolidating the current body of literature, this work aimed to further the understanding of innate immune signaling within skeletal muscle, which can highlight areas for future research and therapeutic strategies to regulate NLRP3 inflammasome activation during divergent metabolic conditions.
Collapse
Affiliation(s)
- Priyanka Khemraj
- Muscle Health Research Centre, School of Kinesiology and Health Science, York University, Toronto M3J 1P3, Canada
| | - Anastasiya Kuznyetsova
- Muscle Health Research Centre, School of Kinesiology and Health Science, York University, Toronto M3J 1P3, Canada
| | - David A Hood
- Muscle Health Research Centre, School of Kinesiology and Health Science, York University, Toronto M3J 1P3, Canada.
| |
Collapse
|
44
|
Schmidleithner L, Stüve P, Feuerer M. Transposable elements as instructors of the immune system. Nat Rev Immunol 2025:10.1038/s41577-025-01172-3. [PMID: 40301669 DOI: 10.1038/s41577-025-01172-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/27/2025] [Indexed: 05/01/2025]
Abstract
Transposable elements (TEs) are mobile repetitive nucleic acid sequences that have been incorporated into the genome through spontaneous integration, accounting for almost 50% of human DNA. Even though most TEs are no longer mobile today, studies have demonstrated that they have important roles in different biological processes, such as ageing, embryonic development, and cancer. TEs influence these processes through various mechanisms, including active transposition of TEs contributing to ongoing evolution, transposon transcription generating RNA or protein, and by influencing gene regulation as enhancers. However, how TEs interact with the immune system remains a largely unexplored field. In this Perspective, we describe how TEs might influence different aspects of the immune system, such as innate immune responses, T cell activation and differentiation, and tissue adaptation. Furthermore, TEs can serve as a source of neoantigens for T cells in antitumour immunity. We suggest that TE biology is an important emerging field of immunology and discuss the potential to harness the TE network therapeutically, for example, to improve immunotherapies for cancer and autoimmune and inflammatory diseases.
Collapse
Affiliation(s)
| | - Philipp Stüve
- Leibniz Institute for Immunotherapy, Regensburg, Germany
| | - Markus Feuerer
- Leibniz Institute for Immunotherapy, Regensburg, Germany.
- Chair for Immunology, University Regensburg, Regensburg, Germany.
| |
Collapse
|
45
|
Pham TL, Sharma R, Neupane C, Gao F, Cha GH, Kim H, Nam MH, Lee SE, Yang S, Sim H, Lee S, Hur GM, Kim HW, Park JB. Neuronal STING-GAT1 signaling maintains paclitaxel-induced neuropathic pain in the spinal cord. Pain 2025:00006396-990000000-00886. [PMID: 40310867 DOI: 10.1097/j.pain.0000000000003593] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2024] [Accepted: 02/10/2025] [Indexed: 05/03/2025]
Abstract
ABSTRACT Stimulator of interferon genes (STING), a pivotal immune regulator, has emerged as a contributor to nociception, yet its role in chronic pains remains still unknown. Here, we demonstrate that STING plays a dual role in normal and neuropathic pain in mature male rodents. Stimulator of interferon genes maintains type I interferon (IFN-I) level restraining pain sensitivity in normal and sham control, while activated STING/interferon regulatory factor 3 (IRF3) signaling increases the expression of gamma-aminobutyric acid (GABA) transporter 1 (GAT1) in the spinal cord (SC), thus, generating paclitaxel (PTX)-induced peripheral neuropathy. Genetic interference of STING (STING-/- mice) attenuated PTX-induced mechanical hypersensitivity with attenuated PTX-induced GAT1 increase, preventing PTX-induced increase in tonic GABAA inhibition of the spinal dorsal horn neurons. Stimulator of interferon genes regulates GAT expression through a TANK-binding kinase 1 (TBK1)-IRF3 signaling pathway, with IRF3 as a crucial transcription factor. Silencing neuronal STING, as opposed to its astrocytic counterpart, effectively restrained the PTX-induced mechanical hypersensitivity and GAT1 increase in the SC. Pharmacological inhibition of STING (H-151) efficiently diminished the TBK1/IRF3/GAT1 signaling pathway to alleviate PTX-induced mechanical hypersensitivity. Our findings show that STING-IRF3 serves a dual role: suppressing physiological nociception through IFN-I and acting as a transcriptional regulator of GAT1, contributing to chemotherapy-induced neuropathic pain.
Collapse
Affiliation(s)
- Thuy Linh Pham
- Laboratory of Veterinary Pharmacology, College of Veterinary Medicine and Research Institute for Veterinary Science, Seoul National University, Seoul, Korea
- Department of Physiology and Medical Science, College of Medicine and Brain Research Institute, Chungnam National University, Daejeon, South Korea
- Department of Obstetrics and Gynecology, Viet Tiep Friendship Hospital, Hai Phong, Vietnam
| | - Ramesh Sharma
- Laboratory of Veterinary Pharmacology, College of Veterinary Medicine and Research Institute for Veterinary Science, Seoul National University, Seoul, Korea
- Department of Physiology and Medical Science, College of Medicine and Brain Research Institute, Chungnam National University, Daejeon, South Korea
| | - Chiranjivi Neupane
- Laboratory of Veterinary Pharmacology, College of Veterinary Medicine and Research Institute for Veterinary Science, Seoul National University, Seoul, Korea
- Department of Physiology and Medical Science, College of Medicine and Brain Research Institute, Chungnam National University, Daejeon, South Korea
| | - Feifei Gao
- Department of Infectious Biology, Chungnam National University, Daejeon, South Korea
| | - Guang-Ho Cha
- Department of Infectious Biology, Chungnam National University, Daejeon, South Korea
| | - Hyunjin Kim
- Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul, Republic of Korea
| | - Min-Ho Nam
- Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul, Republic of Korea
| | - Seung Eun Lee
- Research Animal Resource Center, Korea Institute of Science and Technology (KIST), Seoul, Republic of Korea
| | - Sunjung Yang
- Laboratory of Veterinary Pharmacology, College of Veterinary Medicine and Research Institute for Veterinary Science, Seoul National University, Seoul, Korea
| | - Hunju Sim
- Laboratory of Veterinary Pharmacology, College of Veterinary Medicine and Research Institute for Veterinary Science, Seoul National University, Seoul, Korea
| | - Sanghoon Lee
- Laboratory of Veterinary Pharmacology, College of Veterinary Medicine and Research Institute for Veterinary Science, Seoul National University, Seoul, Korea
| | - Gang Min Hur
- Pharmacology and Medical Science, Chungnam National University, Daejeon, South Korea
| | - Hyun-Woo Kim
- Department of Physiology and Medical Science, College of Medicine and Brain Research Institute, Chungnam National University, Daejeon, South Korea
| | - Jin Bong Park
- Laboratory of Veterinary Pharmacology, College of Veterinary Medicine and Research Institute for Veterinary Science, Seoul National University, Seoul, Korea
| |
Collapse
|
46
|
Dipalo LL, Mikkelsen JG, Gijsbers R, Carlon MS. Trojan Horse-Like Vehicles for CRISPR-Cas Delivery: Engineering Extracellular Vesicles and Virus-Like Particles for Precision Gene Editing in Cystic Fibrosis. Hum Gene Ther 2025. [PMID: 40295092 DOI: 10.1089/hum.2024.258] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/30/2025] Open
Abstract
The advent of genome editing has kindled the hope to cure previously uncurable, life-threatening genetic diseases. However, whether this promise can be ultimately fulfilled depends on how efficiently gene editing agents can be delivered to therapeutically relevant cells. Over time, viruses have evolved into sophisticated, versatile, and biocompatible nanomachines that can be engineered to shuttle payloads to specific cell types. Despite the advances in safety and selectivity, the long-term expression of gene editing agents sustained by viral vectors remains a cause for concern. Cell-derived vesicles (CDVs) are gaining traction as elegant alternatives. CDVs encompass extracellular vesicles (EVs), a diverse set of intrinsically biocompatible and low-immunogenic membranous nanoparticles, and virus-like particles (VLPs), bioparticles with virus-like scaffold and envelope structures, but devoid of genetic material. Both EVs and VLPs can efficiently deliver ribonucleoprotein cargo to the target cell cytoplasm, ensuring that the editing machinery is only transiently active in the cell and thereby increasing its safety. In this review, we explore the natural diversity of CDVs and their potential as delivery vectors for the clustered regularly interspaced short palindromic repeats (CRISPR) machinery. We illustrate different strategies for the optimization of CDV cargo loading and retargeting, highlighting the versatility and tunability of these vehicles. Nonetheless, the lack of robust and standardized protocols for CDV production, purification, and quality assessment still hinders their widespread adoption to further CRISPR-based therapies as advanced "living drugs." We believe that a collective, multifaceted effort is urgently needed to address these critical issues and unlock the full potential of genome-editing technologies to yield safe, easy-to-manufacture, and pharmacologically well-defined therapies. Finally, we discuss the current clinical landscape of lung-directed gene therapies for cystic fibrosis and explore how CDVs could drive significant breakthroughs in in vivo gene editing for this disease.
Collapse
Affiliation(s)
- Laudonia Lidia Dipalo
- Department of Chronic Diseases and Metabolism, Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE), KU Leuven, Leuven, Belgium
| | | | - Rik Gijsbers
- Department of Pharmaceutical and Pharmacological Sciences, Advanced Disease Modelling, Targeted Drug Discovery, and Gene Therapy (ADVANTAGE) labs, KU Leuven, Leuven, Belgium
- Leuven Viral Vector Core, group Biomedical Sciences, KU Leuven, Leuven, Belgium
| | - Marianne S Carlon
- Department of Chronic Diseases and Metabolism, Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE), KU Leuven, Leuven, Belgium
| |
Collapse
|
47
|
Rao K, Zhang X, Luo Y, Xia Q, Jin Y, He J. Lactylation orchestrates ubiquitin-independent degradation of cGAS and promotes tumor growth. Cell Rep 2025; 44:115441. [PMID: 40106438 DOI: 10.1016/j.celrep.2025.115441] [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: 07/12/2024] [Revised: 01/06/2025] [Accepted: 02/26/2025] [Indexed: 03/22/2025] Open
Abstract
Lactate extensively associates with metabolic reprogramming, signal transduction, and immune modulation. Nevertheless, the regulatory role of lactate in immune sensing of cytosolic DNA remains uncertain. Here, we report that lactate serves as an initiator to facilitate proteasomal degradation of cyclic GMP-AMP synthase (cGAS) independent of ubiquitin, thus repressing the production of interferon and contributing to tumor growth. Mechanistically, lactylation of K21 stimulates cGAS translocation from the nucleus to the proteasome for degradation, which is compromised by phosphorylation of PSMA4 S188 via disrupting its association with cGAS. Concurrently, lactylation of K415 rewires PIK3CB activity and impairs ULK1-driven phosphorylation of PSMA4 S188. Physiologically, lactylation of cGAS sustains tumor growth. Expression of cGAS correlates with the antitumor effect of the LDHA inhibitor FX11. Finally, the lactate-cGAS axis indicates a prognostic outcome of lung adenocarcinoma. Collectively, these findings not only put forth a mechanism of cGAS degradation but also unravel the clinical relevance of cGAS lactylation.
Collapse
Affiliation(s)
- Keqiang Rao
- Department of Liver Surgery, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200120, China; Shanghai Institute of Transplantation, Shanghai 200120, China; Shanghai Engineering Research Center of Transplantation and Immunology, Shanghai 200120, China
| | - Xinchao Zhang
- Department of Pathology, College of Basic Medical Sciences, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Yi Luo
- Department of Liver Surgery, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200120, China; Shanghai Institute of Transplantation, Shanghai 200120, China; Shanghai Engineering Research Center of Transplantation and Immunology, Shanghai 200120, China
| | - Qiang Xia
- Department of Liver Surgery, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200120, China; Shanghai Institute of Transplantation, Shanghai 200120, China; Shanghai Engineering Research Center of Transplantation and Immunology, Shanghai 200120, China.
| | - Yuting Jin
- Department of Liver Surgery, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200120, China; Shanghai Institute of Transplantation, Shanghai 200120, China; Shanghai Engineering Research Center of Transplantation and Immunology, Shanghai 200120, China.
| | - Jing He
- Department of Liver Surgery, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200120, China.
| |
Collapse
|
48
|
Zhu J, Lin Q, Zhu H, Xie S, Nie S. Toxicity mechanism analysis of cGAS-STING-TBK1 signaling pathway small molecule modulator based on network toxicology and molecular docking strategy: quinacrine acetate as an example. Front Chem 2025; 13:1584588. [PMID: 40331038 PMCID: PMC12052562 DOI: 10.3389/fchem.2025.1584588] [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: 02/27/2025] [Accepted: 03/25/2025] [Indexed: 05/08/2025] Open
Abstract
Objective This study aims to investigate the toxicity characteristics and mechanisms of quinacrine acetate, a small molecule modulator of the cGAS-STING-TBK1 signaling pathway, and to establish and validate the application value of network toxicology analysis strategy. Methods ProTox and ADMETlab platforms were used to evaluate the toxic effects of quinacrine acetate on human tissues and organs. Potential targets associated with quinacrine acetate toxicity were identified through ChEMBL, STITCH, GeneCards, OMIM, and TD databases. GO and KEGG analyses were employed to elucidate related functions and molecular mechanisms. STRING and Cytoscape software were utilized to identify key hub genes, while molecular docking validation was performed using the CB-Dock2 database. Based on toxicity analysis results, COPD was selected as a disease model, and GEO database was used to analyze the expression characteristics, immune correlation, and drug target value of hub genes in COPD. Results ProTox and ADMETlab analyses revealed that quinacrine acetate exhibited significant toxicity to the respiratory system (toxicity level 4, risk coefficient 0.959). Through integrated multi-database analysis, 14 potential targets related to quinacrine acetate-induced respiratory system toxicity were identified. GO and KEGG pathway analyses indicated that quinacrine acetate-induced respiratory toxicity was primarily mediated through metabolic pathways. Network analysis via STRING and Cytoscape identified AKT1, PLA2G4A, and ALOX5 as three core targets. Molecular docking results confirmed strong binding affinity between quinacrine acetate and these core targets. In COPD patients, PLA2G4A and ALOX5 showed significantly upregulated expression, with hub gene ROC curve AUC value reaching 0.829, demonstrating good diagnostic value. Further immune correlation analysis revealed that ALOX5 and PLA2G4A were closely associated with various immune cell expressions and served as targets for multiple drugs including histamine, melittin, and formic acid. Conclusion This study demonstrates that quinacrine acetate may influence the progression and risk of respiratory system diseases by regulating metabolic pathways. The findings provide not only a theoretical foundation for understanding the molecular mechanisms of quinacrine acetate-induced respiratory toxicity but also new perspectives and methodological references for evaluating the toxic effects of small molecule compounds in respiratory diseases. Therefore, we demonstrates the practical application value of network toxicology as an efficient predictive tool for identifying potential toxicity targets and pathways, which can guide subsequent experimental validation and provide mechanistic insights that traditional toxicology approaches might miss.
Collapse
Affiliation(s)
- Jinchao Zhu
- School of Health Science and Engineering University of Shanghai for Science and Technology, Shanghai, China
| | - Qingyuan Lin
- Department of Pathology, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Honglin Zhu
- School of Health Science and Engineering University of Shanghai for Science and Technology, Shanghai, China
| | - Siqi Xie
- Sichuan Cancer Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Shengdong Nie
- School of Health Science and Engineering University of Shanghai for Science and Technology, Shanghai, China
| |
Collapse
|
49
|
Li JX, Zhang J, Li CH, Li YF, Chen HM, Li T, Zhang Q, Kong BH, Wang PH. Human papillomavirus E1 proteins inhibit RIG-I/MDA5-MAVS, TLR3-TRIF, cGAS-STING, and JAK-STAT signaling pathways to evade innate antiviral immunity. Front Immunol 2025; 16:1549766. [PMID: 40330484 PMCID: PMC12052760 DOI: 10.3389/fimmu.2025.1549766] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2024] [Accepted: 03/25/2025] [Indexed: 05/08/2025] Open
Abstract
Human papillomavirus (HPV) is a major etiological agent of both malignant and benign lesions, with high-risk types, such as HPV16 and HPV18, being strongly linked to cervical cancer, while low-risk types like HPV11 are associated with benign conditions. While viral proteins such as E6 and E7 are well-established regulators of immune evasion, the role of E1 in modulating the host antiviral responses remains insufficiently characterized. This study investigates the immunomodulatory functions of HPV16 and HPV11 E1 in suppressing innate antiviral immune signaling pathways. Through a combination of RT-qPCR and luciferase reporter assays, we demonstrate that E1 suppresses the production of interferons and interferon-stimulated genes triggered by viral infections and the activation of RIG-I/MDA5-MAVS, TLR3-TRIF, cGAS-STING, and JAK-STAT pathways. Co-immunoprecipitation assays reveal that E1 interacts directly with key signaling molecules within these pathways. E1 also impairs TBK1 and IRF3 phosphorylation and obstructs the nuclear translocation of IRF3, thereby broadly suppressing IFN responses. Additionally, E1 disrupts the JAK-STAT pathway by binding STAT1, which prevents the assembly and nuclear localization of the ISGF3 complex containing STAT1, STAT2, and IRF9, thereby further diminishing antiviral response. These findings establish E1 as a pivotal regulator of immune evasion and suggest its potential as a novel therapeutic target to enhance antiviral immunity in HPV-associated diseases.
Collapse
MESH Headings
- Humans
- Immunity, Innate
- Signal Transduction/immunology
- Interferon-Induced Helicase, IFIH1/metabolism
- Interferon-Induced Helicase, IFIH1/immunology
- DEAD Box Protein 58/metabolism
- DEAD Box Protein 58/immunology
- Membrane Proteins/metabolism
- Membrane Proteins/immunology
- Nucleotidyltransferases/metabolism
- Nucleotidyltransferases/immunology
- Toll-Like Receptor 3/metabolism
- Toll-Like Receptor 3/immunology
- Immune Evasion
- Papillomavirus Infections/immunology
- Papillomavirus Infections/virology
- Human papillomavirus 16/immunology
- Receptors, Immunologic
- Oncogene Proteins, Viral/immunology
- Oncogene Proteins, Viral/metabolism
- Adaptor Proteins, Vesicular Transport/metabolism
- Adaptor Proteins, Vesicular Transport/immunology
- Adaptor Proteins, Signal Transducing/metabolism
- Adaptor Proteins, Signal Transducing/immunology
- Human papillomavirus 11/immunology
- HEK293 Cells
- STAT Transcription Factors/metabolism
- Interferon Regulatory Factor-3
- Human Papillomavirus Viruses
Collapse
Affiliation(s)
- Jin-Xin Li
- Department of Infectious Disease and Hepatology, The Second Hospital of Shandong University, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Jing Zhang
- Department of Infectious Disease and Hepatology, The Second Hospital of Shandong University, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Cheng-Hao Li
- Key Laboratory for Experimental Teratology of Ministry of Education and Advanced Medical Research Institute, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Yun-Fang Li
- Key Laboratory for Experimental Teratology of Ministry of Education and Advanced Medical Research Institute, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Hui-Min Chen
- Key Laboratory for Experimental Teratology of Ministry of Education and Advanced Medical Research Institute, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Tao Li
- Department of Infectious Disease and Hepatology, The Second Hospital of Shandong University, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Qing Zhang
- Department of Obstetrics and Gynecology, Qilu Hospital, Shandong University, Jinan, China
- Gynecologic Oncology Key Laboratory of Shandong Province, Qilu Hospital, Shandong University, Jinan, China
| | - Bei-Hua Kong
- Department of Obstetrics and Gynecology, Qilu Hospital, Shandong University, Jinan, China
- Gynecologic Oncology Key Laboratory of Shandong Province, Qilu Hospital, Shandong University, Jinan, China
| | - Pei-Hui Wang
- Department of Infectious Disease and Hepatology, The Second Hospital of Shandong University, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
- Key Laboratory for Experimental Teratology of Ministry of Education and Advanced Medical Research Institute, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| |
Collapse
|
50
|
Du H, Cui D, Hu S, Zhou X, Lin X, Fu X, Feng S, Xu S, Jian W, Guo Y, Zhang S, Chen Q. The induction of type I interferonopathy in Trex1-P212fs mice is mediated by activation of the cGAS-STING pathway. Int J Biol Macromol 2025; 310:143414. [PMID: 40268028 DOI: 10.1016/j.ijbiomac.2025.143414] [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: 08/26/2024] [Revised: 04/03/2025] [Accepted: 04/20/2025] [Indexed: 04/25/2025]
Abstract
The cGAS-STING pathway is crucial for immune tolerance, pathogen resistance, and tumor immunity. Knocking out the cGAS gene can reverse the type I interferonopathy seen in Trex1-/- and Trex1D18N/D18N mice. TREX1, a key DNA-specific exonuclease in mammalian cells, degrades cytoplasmic DNA to prevent excessive immune activation. Mutations in TREX1 are linked to various autoimmune diseases. In prior research, we generated a Trex1-P212fs mouse model associated with systemic lupus erythematosus (SLE) using CRISPR-Cas9 gene editing. This model displays systemic inflammation that mirrors numerous characteristics of both Aicardi-Goutières syndrome (AGS) and SLE in humans. In this study, we found that the TREX1-P212fs mutation resulted in reduced dsDNA enzyme activity. DNA accumulation was present in the cytoplasm of Trex1P212fs/P212fs MEFs. Nonetheless, the role of the cGAS-STING pathway in mediating the disease phenotype in Trex1-P212fs mice associated with SLE has yet to be elucidated. We observed that cGas knockout mitigated systemic inflammation, lymphocyte proliferation, vasculitis, renal disease, and spontaneous T cell activation in Trex1-P212fs mice. Similarly, inhibition of STING with C-176 treatment ameliorated the disease phenotype in Trex1-P212fs mice. These findings elucidate the pathogenesis of TREX1-P212fs-associated type I interferonopathy and offer potential therapeutic targets for their treatment.
Collapse
Affiliation(s)
- Hekang Du
- Department of Pathology, The First Affiliated Hospital, Fujian Medical University, 20 Chazhong Road, Fuzhou 350005, China; Fujian Key Laboratory of Innate Immune Biology, Biomedical Research Center of South China, College of Life Sciences, Fujian Normal University Qishan Campus, College Town, Fuzhou, Fujian Province 350117, China.; Department of Pathology, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, 999 Huashan Road, Fuzhou 350212, China
| | - Dongya Cui
- Fujian Key Laboratory of Innate Immune Biology, Biomedical Research Center of South China, College of Life Sciences, Fujian Normal University Qishan Campus, College Town, Fuzhou, Fujian Province 350117, China
| | - Shun Hu
- Department of Pathology, The First Affiliated Hospital, Fujian Medical University, 20 Chazhong Road, Fuzhou 350005, China; Department of Pathology, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, 999 Huashan Road, Fuzhou 350212, China
| | - Xueyuan Zhou
- Fujian Key Laboratory of Innate Immune Biology, Biomedical Research Center of South China, College of Life Sciences, Fujian Normal University Qishan Campus, College Town, Fuzhou, Fujian Province 350117, China
| | - Xiaofang Lin
- Fujian Key Laboratory of Innate Immune Biology, Biomedical Research Center of South China, College of Life Sciences, Fujian Normal University Qishan Campus, College Town, Fuzhou, Fujian Province 350117, China
| | - Xiaodan Fu
- Department of Pathology, The First Affiliated Hospital, Fujian Medical University, 20 Chazhong Road, Fuzhou 350005, China; Department of Pathology, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, 999 Huashan Road, Fuzhou 350212, China
| | - Sisi Feng
- Department of Pathology, The First Affiliated Hospital, Fujian Medical University, 20 Chazhong Road, Fuzhou 350005, China; Department of Pathology, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, 999 Huashan Road, Fuzhou 350212, China
| | - Shan Xu
- Fujian Key Laboratory of Innate Immune Biology, Biomedical Research Center of South China, College of Life Sciences, Fujian Normal University Qishan Campus, College Town, Fuzhou, Fujian Province 350117, China
| | - Wentin Jian
- Department of Pathology, The First Affiliated Hospital, Fujian Medical University, 20 Chazhong Road, Fuzhou 350005, China; Department of Pathology, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, 999 Huashan Road, Fuzhou 350212, China
| | - Yuanli Guo
- Department of Pathology, The First Affiliated Hospital, Fujian Medical University, 20 Chazhong Road, Fuzhou 350005, China; Department of Pathology, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, 999 Huashan Road, Fuzhou 350212, China
| | - Sheng Zhang
- Department of Pathology, The First Affiliated Hospital, Fujian Medical University, 20 Chazhong Road, Fuzhou 350005, China; Department of Pathology, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, 999 Huashan Road, Fuzhou 350212, China..
| | - Qi Chen
- Fujian Key Laboratory of Innate Immune Biology, Biomedical Research Center of South China, College of Life Sciences, Fujian Normal University Qishan Campus, College Town, Fuzhou, Fujian Province 350117, China..
| |
Collapse
|