1
|
Zhao Z, Zhou J, Li X, Zhang T, Tian Z, Sun T, Jiang C. Manganese-based virus-mimicking nanomedicine with triple immunomodulatory functions inhibits breast cancer brain metastasis. Biomaterials 2025; 320:123262. [PMID: 40138963 DOI: 10.1016/j.biomaterials.2025.123262] [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: 12/09/2024] [Revised: 02/23/2025] [Accepted: 03/16/2025] [Indexed: 03/29/2025]
Abstract
Hindered by the challenges of blood-brain barrier (BBB) hindrance, tumor heterogeneity and immunosuppressive microenvironment, patients with breast cancer brain metastasis have yet to benefit from current clinical treatments, experiencing instead a decline in quality of life due to radiochemotherapy. While virus-mimicking nanosystems (VMN) mimicking viral infection processes show promise in treating peripheral tumors, the inability to modulate the immunosuppressive microenvironment limits the efficacy against brain metastasis. Accordingly, a VMN-based triple immunomodulatory strategy is initially proposed, aiming to activate innate and adaptive immune responses and reverse the immunosuppressive microenvironment. Here, manganese-based virus-mimicking nanomedicine (Vir-HD@HM) with intratumoral drug enrichment is engineered. Vir-HD@HM can induce the immune response through the activation of cGAS-STING by mimicking the in vivo infection process of herpesviruses. Meanwhile, DNAzyme mimicking the genome can rescue the epigenetic silencing of PTEN with the assistance of Mn2+, thus ameliorating the immunosuppressive metastatic microenvironment and achieving synergistic sensitizing therapeutic efficacy. In vivo experiments substantiate the efficacy of Vir-HD@HM in recruiting NK cells and CD8+ T cells to metastatic foci, inhibiting Treg cells infiltration, and prolonging murine survival without adjunctive radiochemotherapy. This study demonstrates that Vir-HD@HM with triple immunomodulation offers an encouraging therapeutic option for patients with brain metastasis.
Collapse
Affiliation(s)
- Zhenhao Zhao
- Department of Pharmaceutics, School of Pharmacy, Fudan University, Key Laboratory of Smart Drug Delivery, Ministry of Education, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai, 201203, China
| | - Jingyi Zhou
- Department of Pharmaceutics, School of Pharmacy, Fudan University, Key Laboratory of Smart Drug Delivery, Ministry of Education, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai, 201203, China
| | - Xuwen Li
- Department of Pharmaceutics, School of Pharmacy, Fudan University, Key Laboratory of Smart Drug Delivery, Ministry of Education, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai, 201203, China
| | - Tongyu Zhang
- Department of Pharmaceutics, School of Pharmacy, Fudan University, Key Laboratory of Smart Drug Delivery, Ministry of Education, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai, 201203, China
| | - Zonghua Tian
- Department of Pharmaceutics, School of Pharmacy, Fudan University, Key Laboratory of Smart Drug Delivery, Ministry of Education, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai, 201203, China
| | - Tao Sun
- Department of Pharmaceutics, School of Pharmacy, Fudan University, Key Laboratory of Smart Drug Delivery, Ministry of Education, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai, 201203, China
| | - Chen Jiang
- Department of Pharmaceutics, School of Pharmacy, Fudan University, Key Laboratory of Smart Drug Delivery, Ministry of Education, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai, 201203, China; Department of Digestive Diseases, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350212, China.
| |
Collapse
|
2
|
Zhang Z, Wang X, Zhao C, Zhu H, Liao X, Tsai HI. STING and metabolism-related diseases: Roles, mechanisms, and applications. Cell Signal 2025; 132:111833. [PMID: 40294833 DOI: 10.1016/j.cellsig.2025.111833] [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: 01/23/2025] [Revised: 04/08/2025] [Accepted: 04/22/2025] [Indexed: 04/30/2025]
Abstract
The stimulator of interferon genes (STING) pathway plays a critical role in innate immunity, acting as a central mediator that links cytosolic DNA sensing to inflammatory signaling. STING not only responds to cellular metabolic states but also actively regulates key metabolic processes, including glycolysis, lipid metabolism, and redox balance. This bidirectional interaction underscores the existence of a dynamic feedback mechanism between STING signaling and metabolic pathways, which is essential for maintaining cellular homeostasis. This review provides a comprehensive analysis, beginning with an in-depth overview of the classical STING signaling pathway, followed by a detailed examination of its reciprocal regulation of various metabolic pathways. Additionally, it explores the role and mechanisms of STING signaling in metabolic disorders, including obesity, diabetes, and atherosclerosis. By integrating these insights into the mutual regulation between STING and its metabolism, novel therapeutic strategies targeting this pathway in metabolic diseases have been proposed.
Collapse
Affiliation(s)
- Zhengyang Zhang
- Institute of Medical Imaging and Artificial Intelligence, Jiangsu University, Zhenjiang 212001, China; School of Medicine, Jiangsu University, Zhenjiang 212013, China
| | - Xirui Wang
- Department of Biomedical Engineering, School of Medical Imaging, Xuzhou Medical University, Xuzhou 221000, China
| | - Chuangchuang Zhao
- Institute of Medical Imaging and Artificial Intelligence, Jiangsu University, Zhenjiang 212001, China; School of Medicine, Jiangsu University, Zhenjiang 212013, China
| | - Haitao Zhu
- Institute of Medical Imaging and Artificial Intelligence, Jiangsu University, Zhenjiang 212001, China; Department of Medical Imaging, The Affiliated Hospital of Jiangsu University, Zhenjiang 212001, China
| | - Xiang Liao
- Institute of Medical Imaging and Artificial Intelligence, Jiangsu University, Zhenjiang 212001, China.
| | - Hsiang-I Tsai
- Institute of Medical Imaging and Artificial Intelligence, Jiangsu University, Zhenjiang 212001, China; Department of Medical Imaging, The Affiliated Hospital of Jiangsu University, Zhenjiang 212001, China.
| |
Collapse
|
3
|
Qiu W, Zheng Z, Wang J, Cai Y, Zou J, Huang Z, Yang P, Ye W, Jin M, Zhang D, Little PJ, Zhou Q, Liu Z. Targeting mitochondrial DNA-STING-NF-κB Axis-mediated microglia activation by cryptotanshinone alleviates ischemic retinopathy. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 142:156779. [PMID: 40279967 DOI: 10.1016/j.phymed.2025.156779] [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: 10/09/2024] [Revised: 03/27/2025] [Accepted: 04/17/2025] [Indexed: 04/29/2025]
Abstract
BACKGROUND Ischemic retinopathy, a leading cause of vision impairment, involves oxidative stress and dysregulated inflammation, with microglia playing a key role. Cryptotanshinone (CTS), a bioactive compound from Salvia miltiorrhiza, exhibits anti-inflammatory and antioxidant properties and thus has the potential for development as a therapeutic agent. However, the actual mechanism of action of CTS in ischemic retinopathy is not known. Overactivation of the STING pathway in microglia is critical in ischemic retinopathy pathogenesis and a potential target of CTS. PURPOSE This study aimed to explore whether CTS alleviates ischemic retinopathy by modulating microglial STING signaling. METHODS Oxygen-induced retinopathy (OIR) mice and hypoxia-induced microglial cells were used. CTS efficacy in ischemic retinopathy was evaluated at multiple stages using fluorescein fundus angiography, electroretinogram, H&E staining, and Western blotting of relevant proteins. Network pharmacology and RNA sequencing identified STING as a key target. Furthermore, surface plasmon resonance (SPR), molecular docking, and site-directed mutagenesis were systematically employed to elucidate the precise binding interface between CTS and the STING protein. STING activation and knockout models were employed to further investigate the mechanisms of action of CTS. RESULTS CTS treatment reduced microglial activation and pathological retinal angiogenesis, and protected both retinal function and structure in OIR mice. Network pharmacology, RNA sequencing, and experimental validation demonstrated a significant link between the protective effect of CTS and the inhibition of STING signaling. Mechanistically, CTS suppressed cytosolic mtDNA release, blocked STING translocation from the ER to the Golgi, and enhanced lysosomal STING degradation. These CTS-mediated effects were abolished by STING activation and absent in Sting-deficient OIR mice. Notably, CTS combined with anti-VEGF therapy showed synergistic efficacy in suppressing pathological retinal neovascularization. CONCLUSION CTS, a natural inhibitor of STING, alleviated ischemic retinopathy by inhibiting the mtDNA-STING-NF-κB signaling pathway via multifaceted mechanisms in microglia.
Collapse
Affiliation(s)
- Wanlu Qiu
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research, International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Discovery of Chinese Ministry of Education (MOE), College of Pharmacy, Jinan University, Guangzhou 510632, China; Department of Ophthalmology, the First Affiliated Hospital, Jinan University, Guangzhou 510006, China
| | - Zhihua Zheng
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research, International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Discovery of Chinese Ministry of Education (MOE), College of Pharmacy, Jinan University, Guangzhou 510632, China; The Affiliated Guangdong Second Provincial General Hospital, Postdoctoral Research Station of Traditional Chinese Medicine, School of Pharmacy, Jinan University, Guangzhou, 510632, China
| | - Jiaojiao Wang
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research, International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Discovery of Chinese Ministry of Education (MOE), College of Pharmacy, Jinan University, Guangzhou 510632, China; Key Laboratory of Big Data Mining and Precision Drug Design of Guangdong Medical University, Key Laboratory of Computer-Aided Drug Design of Dongguan City, Key Laboratory for Research and Development of Natural Drugs of Guangdong Province, School of Pharmacy, Guangdong Medical University, Dongguan, 523808, China.
| | - Youran Cai
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research, International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Discovery of Chinese Ministry of Education (MOE), College of Pharmacy, Jinan University, Guangzhou 510632, China; Department of Ophthalmology, the First Affiliated Hospital, Jinan University, Guangzhou 510006, China
| | - Jiami Zou
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research, International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Discovery of Chinese Ministry of Education (MOE), College of Pharmacy, Jinan University, Guangzhou 510632, China
| | - Ziqing Huang
- Department of Ophthalmology, the First Affiliated Hospital, Jinan University, Guangzhou 510006, China
| | - Pinglian Yang
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research, International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Discovery of Chinese Ministry of Education (MOE), College of Pharmacy, Jinan University, Guangzhou 510632, China
| | - Weile Ye
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research, International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Discovery of Chinese Ministry of Education (MOE), College of Pharmacy, Jinan University, Guangzhou 510632, China
| | - Mei Jin
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research, International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Discovery of Chinese Ministry of Education (MOE), College of Pharmacy, Jinan University, Guangzhou 510632, China
| | - Dongmei Zhang
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research, International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Discovery of Chinese Ministry of Education (MOE), College of Pharmacy, Jinan University, Guangzhou 510632, China
| | - Peter J Little
- School of Pharmacy, Pharmacy Australia Centre of Excellence, The University of Queensland, Woolloongabba, QLD 4102, Australia; Department of Pharmacy, Guangzhou Xinhua University, Guangzhou, 510520, China
| | - Qing Zhou
- Department of Ophthalmology, the First Affiliated Hospital, Jinan University, Guangzhou 510006, China.
| | - Zhiping Liu
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research, International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Discovery of Chinese Ministry of Education (MOE), College of Pharmacy, Jinan University, Guangzhou 510632, China.
| |
Collapse
|
4
|
Zhang Z, Yang J, Zhou Q, Zhong S, Liu J, Zhang X, Chang X, Wang H. The cGAS-STING-mediated ROS and ferroptosis are involved in manganese neurotoxicity. J Environ Sci (China) 2025; 152:71-86. [PMID: 39617588 DOI: 10.1016/j.jes.2024.05.003] [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: 01/31/2024] [Revised: 04/12/2024] [Accepted: 05/01/2024] [Indexed: 12/12/2024]
Abstract
Manganese (Mn) has been characterized as an environmental pollutant. Excessive releases of Mn due to human activities have increased Mn levels in the environment over the years, posing a threat to human health and the environment. Long-term exposure to high concentrations of Mn can induce neurotoxicity. Therefore, toxicological studies on Mn are of paramount importance. Mn induces oxidative stress through affecting the level of reactive oxygen species (ROS), and the overabundance of ROS further triggers ferroptosis. Additionally, Mn2+ was found to be a novel activator of the cyclic guanosine-adenosine synthase (cGAS)-stimulator of interferon genes (STING) pathway in the innate immune system. Thus, we speculate that Mn exposure may promote ROS production by activating the cGAS-STING pathway, which further induces oxidative stress and ferroptosis, and ultimately triggers Mn neurotoxicity. This review discusses the mechanism between Mn-induced oxidative stress and ferroptosis via activation of the cGAS-STING pathway, which may offer a prospective direction for future in-depth studies on the mechanism of Mn neurotoxicity.
Collapse
Affiliation(s)
- Zhimin Zhang
- Department of Toxicology, School of Public Health, Lanzhou University, Lanzhou 730000, China
| | - Jirui Yang
- Department of Toxicology, School of Public Health, Lanzhou University, Lanzhou 730000, China
| | - Qiongli Zhou
- Department of Toxicology, School of Public Health, Lanzhou University, Lanzhou 730000, China
| | - Shiyin Zhong
- Department of Toxicology, School of Public Health, Lanzhou University, Lanzhou 730000, China
| | - Jingjing Liu
- Department of Toxicology, School of Public Health, Lanzhou University, Lanzhou 730000, China
| | - Xin Zhang
- Department of Toxicology, School of Public Health, Lanzhou University, Lanzhou 730000, China
| | - Xuhong Chang
- Department of Toxicology, School of Public Health, Lanzhou University, Lanzhou 730000, China
| | - Hui Wang
- Department of Toxicology, School of Public Health, Lanzhou University, Lanzhou 730000, China.
| |
Collapse
|
5
|
Gu Z, Li L, Xu P, Li C, Liu B, Zhu P, Xie W, Zhang LW, Wang Y, Wang Y. Multimodal Regulation of Dendritic Cells via Mineralized Vaccines for Postsurgical Tumor Relapse Prevention. ACS NANO 2025. [PMID: 40401399 DOI: 10.1021/acsnano.5c02846] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/23/2025]
Abstract
The regulation of dendritic cell (DC) function is pivotal for augmenting the immune response of traditional tumor therapy, particularly in mitigating postsurgical tumor relapse. Unfortunately, DC functionality is often compromised in the postsurgical microenvironment, leading to immune tolerance and tumor progression. To address these challenges, we developed a personalized cancer vaccine, Cell@CaP, which utilizes inactivated autologous tumor cells coated with a calcium phosphate shell. This innovative vaccine incorporates cyclic GMP-AMP (cGAMP), a STING agonist, and calcium ions (Ca2+) to boost DCs activation and antigen presentation. The Cell@CaP vaccine leverages high mechanical hardness, intact tumor antigens, and the release of cGAMP and Ca2+ to activate DCs through multiple pathways. Upon interaction with DCs, the vaccine triggers the Piezo1 mechanosensitive channel, inducing a Ca2+ influx. This mechanical stimulation, combined with the release of cGAMP and tumor antigens, activates the cGAS-STING, PI3K-Akt, and RhoA-MYPT1 signaling pathways. These synergistic effects enhance the expression of costimulatory molecules and promote the secretion of pro-inflammatory cytokines (interferon-β and tumor necrosis factor-α). In a postsurgical osteosarcoma model, the combination of Cell@CaP with radiotherapy significantly inhibited primary tumor relapse and suppressed distant lung metastases. This multimodal vaccine strategy integrates mechanical, chemical, and immunological cues into a single platform, offering a promising approach to enhance DCs functionality and activate robust antitumor immunity. The study highlights the potential of Cell@CaP as a personalized immunotherapy for preventing postsurgical tumor relapse.
Collapse
Affiliation(s)
- Zenghui Gu
- State Key Laboratory of Radiation Medicine and Protection, The Fourth Affiliated Hospital of Soochow University, School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou 215123, China
- Department of Orthopedic, the Second Affiliated Hospital of Soochow University, 1055 Sanxiang Road, Suzhou 215000, China
| | - Liubing Li
- State Key Laboratory of Radiation Medicine and Protection, The Fourth Affiliated Hospital of Soochow University, School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou 215123, China
- Department of Orthopedic, the Second Affiliated Hospital of Soochow University, 1055 Sanxiang Road, Suzhou 215000, China
| | - Pei Xu
- State Key Laboratory of Radiation Medicine and Protection, The Fourth Affiliated Hospital of Soochow University, School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou 215123, China
| | - Chenze Li
- State Key Laboratory of Radiation Medicine and Protection, The Fourth Affiliated Hospital of Soochow University, School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou 215123, China
| | - Bo Liu
- State Key Laboratory of Radiation Medicine and Protection, The Fourth Affiliated Hospital of Soochow University, School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou 215123, China
| | - Peiyao Zhu
- State Key Laboratory of Radiation Medicine and Protection, The Fourth Affiliated Hospital of Soochow University, School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou 215123, China
- Department of Orthopedic, the Second Affiliated Hospital of Soochow University, 1055 Sanxiang Road, Suzhou 215000, China
| | - Wei Xie
- State Key Laboratory of Radiation Medicine and Protection, The Fourth Affiliated Hospital of Soochow University, School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou 215123, China
| | - Leshuai W Zhang
- State Key Laboratory of Radiation Medicine and Protection, The Fourth Affiliated Hospital of Soochow University, School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou 215123, China
| | - Yangyun Wang
- State Key Laboratory of Radiation Medicine and Protection, The Fourth Affiliated Hospital of Soochow University, School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou 215123, China
| | - Yong Wang
- State Key Laboratory of Radiation Medicine and Protection, The Fourth Affiliated Hospital of Soochow University, School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou 215123, China
- Department of Orthopedic, the Second Affiliated Hospital of Soochow University, 1055 Sanxiang Road, Suzhou 215000, China
| |
Collapse
|
6
|
Timilsina S, Huang JY, Abdelfattah N, Medina D, Singh D, Abdulsahib S, Subbarayalu P, Do TP, Venkata PP, Nirzhor S, Prochnau J, Bhandari M, Zheng S, Chen Y, Huang G, Mukherjee N, Hromas R, Sung P, Kaklamani V, Vadlamudi R, Zhang N, Rao MK. Epigenetic silencing of DNA sensing pathway by FOXM1 blocks stress ligand-dependent antitumor immunity and immune memory. Nat Commun 2025; 16:3967. [PMID: 40295473 PMCID: PMC12037779 DOI: 10.1038/s41467-025-59186-3] [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: 04/19/2024] [Accepted: 04/14/2025] [Indexed: 04/30/2025] Open
Abstract
The interplay between tumor cells and the microenvironment significantly influences cancer progression. Here, we report a significant role of the transcription factor FOXM1 in shaping the tumor immune landscape. Single-cell sequencing reveals that tumor-intrinsic FOXM1 creates an immune-suppressive tumor microenvironment by inhibiting expression of stress ligands (including ULBP1) on cancer cells, thereby blocking NKG2D-NKG2DL interactions critical for priming natural killer- and T cell-mediated cytotoxicity of cancer cells. FOXM1 suppresses ULBP1 expression by epigenetically silencing the DNA-sensing protein STING using a DNMT1-UHRF1 complex, which in turn inhibits the unfolded protein response protein CHOP from activating ULBP1. Importantly, cancer patients with higher levels of FOXM1 and DNMT1, and lower levels of STING and ULBP1, have worse survival and are less responsive to immunotherapy. Collectively, our findings provide key insight into how a tumor-intrinsic transcription factor epigenetically shapes the tumor immune microenvironment, with strong implications for refining existing and designing new cancer immunotherapies.
Collapse
Affiliation(s)
| | - Jian Yu Huang
- Greehey Children's Cancer Research Institute, San Antonio, TX, USA
| | - Nourhan Abdelfattah
- Department of Neurology, Houston Methodist Research Institute, Houston, TX, USA
| | - Daisy Medina
- Greehey Children's Cancer Research Institute, San Antonio, TX, USA
- Department of Cell Systems and Anatomy, UT Health San Antonio, San Antonio, TX, USA
| | - Deepika Singh
- Greehey Children's Cancer Research Institute, San Antonio, TX, USA
- Department of Cell Systems and Anatomy, UT Health San Antonio, San Antonio, TX, USA
| | - Shahad Abdulsahib
- Greehey Children's Cancer Research Institute, San Antonio, TX, USA
- Department of Cell Systems and Anatomy, UT Health San Antonio, San Antonio, TX, USA
| | - Panneerdoss Subbarayalu
- Greehey Children's Cancer Research Institute, San Antonio, TX, USA
- Department of Cell Systems and Anatomy, UT Health San Antonio, San Antonio, TX, USA
| | - Trong Phat Do
- Greehey Children's Cancer Research Institute, San Antonio, TX, USA
- Department of Cell Systems and Anatomy, UT Health San Antonio, San Antonio, TX, USA
| | - Prabhakar Pitta Venkata
- Greehey Children's Cancer Research Institute, San Antonio, TX, USA
- Department of Cell Systems and Anatomy, UT Health San Antonio, San Antonio, TX, USA
| | - Saif Nirzhor
- Greehey Children's Cancer Research Institute, San Antonio, TX, USA
- Department of Cell Systems and Anatomy, UT Health San Antonio, San Antonio, TX, USA
| | - Jack Prochnau
- Greehey Children's Cancer Research Institute, San Antonio, TX, USA
| | - Mukund Bhandari
- Department of Pathology, UT Southwestern Medical Center, Dallas, TX, USA
| | - Siyuan Zheng
- Greehey Children's Cancer Research Institute, San Antonio, TX, USA
- Department of Population Health Sciences, UT Health San Antonio, San Antonio, TX, USA
| | - Yidong Chen
- Greehey Children's Cancer Research Institute, San Antonio, TX, USA
- Department of Population Health Sciences, UT Health San Antonio, San Antonio, TX, USA
| | - Gang Huang
- Department of Cell Systems and Anatomy, UT Health San Antonio, San Antonio, TX, USA
| | | | - Robert Hromas
- Department of Medicine, UT Health, San Antonio, TX, USA
| | - Patrick Sung
- Greehey Children's Cancer Research Institute, San Antonio, TX, USA
- Department of Biochemistry & Structural Biology, UT Health San Antonio, San Antonio, TX, USA
| | | | - Ratna Vadlamudi
- Department of Obstetrics and Gynecology, UT Health San Antonio, San Antonio, TX, USA
- Audie L. Murphy Division, South Texas Veterans Health Care System, San Antonio, TX, USA
| | - Nu Zhang
- Audie L. Murphy Division, South Texas Veterans Health Care System, San Antonio, TX, USA
- Department of Microbiology, Immunology & Molecular Genetics, UT Health, San Antonio, TX, USA
| | - Manjeet K Rao
- Greehey Children's Cancer Research Institute, San Antonio, TX, USA.
- Department of Neurology, Houston Methodist Research Institute, Houston, TX, USA.
| |
Collapse
|
7
|
He K, Chen D, Zhu D, Zheng W, Lyu L, Zhang M, Chen Z, Wang X, Hu Y, Fu B. Drug-loaded indocyanine green J-aggregates activate metalloimmunotherapy for sustained photothermal therapy of hepatocellular carcinoma. J Nanobiotechnology 2025; 23:317. [PMID: 40287682 PMCID: PMC12032755 DOI: 10.1186/s12951-025-03353-7] [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: 01/10/2025] [Accepted: 03/24/2025] [Indexed: 04/29/2025] Open
Abstract
Hepatocellular carcinoma (HCC) poses a significant therapeutic challenge, driving the need for novel treatment strategies. This study investigates the combination of photothermal therapy (PTT) and metalloimmunotherapy for HCC treatment using Co + diABZI@J-dICG nanoparticles. Indocyanine green (ICG), an FDA-approved near-infrared (NIR) dye, is dimerized into J-aggregates to enhance PTT by improving light absorption and photothermal efficiency. The cGAS-STING pathway, a key mediator of innate immunity, is activated by the STING agonist diABZI, while cobalt ions (Co2+) further enhance immune responses. The Co + diABZI@J-dICG nanoparticles take advantage of ICG's hepatotropic properties for sustained tumor accumulation and immune activation, resulting in significant tumor growth inhibition and reduced HCC recurrence following hepatectomy.
Collapse
Affiliation(s)
- Kaiming He
- Department of Hepatic Surgery and Liver Transplantation Center, The Third Affiliated Hospital of Sun Yat-sen University, Organ Transplantation Institute of Sun Yat-sen University, Organ Transplantation Research Center of Guangdong Province, Guangdong Province Engineering Laboratory for Transplantation Medicine, Guangzhou, 510630, China
- Guangdong Provincial Key Laboratory of Liver Disease Research, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510630, China
| | - Desheng Chen
- Department of Hepatic Surgery and Liver Transplantation Center, The Third Affiliated Hospital of Sun Yat-sen University, Organ Transplantation Institute of Sun Yat-sen University, Organ Transplantation Research Center of Guangdong Province, Guangdong Province Engineering Laboratory for Transplantation Medicine, Guangzhou, 510630, China
- Guangdong Provincial Key Laboratory of Liver Disease Research, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510630, China
| | - Dongzi Zhu
- Department of General Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
| | - Wenjie Zheng
- Department of Hepatic Surgery and Liver Transplantation Center, The Third Affiliated Hospital of Sun Yat-sen University, Organ Transplantation Institute of Sun Yat-sen University, Organ Transplantation Research Center of Guangdong Province, Guangdong Province Engineering Laboratory for Transplantation Medicine, Guangzhou, 510630, China
- Guangdong Provincial Key Laboratory of Liver Disease Research, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510630, China
| | - Lei Lyu
- Department of Hepatic Surgery and Liver Transplantation Center, The Third Affiliated Hospital of Sun Yat-sen University, Organ Transplantation Institute of Sun Yat-sen University, Organ Transplantation Research Center of Guangdong Province, Guangdong Province Engineering Laboratory for Transplantation Medicine, Guangzhou, 510630, China
- Guangdong Provincial Key Laboratory of Liver Disease Research, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510630, China
| | - Mingshen Zhang
- Department of Hepatic Surgery and Liver Transplantation Center, The Third Affiliated Hospital of Sun Yat-sen University, Organ Transplantation Institute of Sun Yat-sen University, Organ Transplantation Research Center of Guangdong Province, Guangdong Province Engineering Laboratory for Transplantation Medicine, Guangzhou, 510630, China
- Guangdong Provincial Key Laboratory of Liver Disease Research, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510630, China
| | - Zeping Chen
- Department of General Surgery, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China
| | - Xiaowen Wang
- Department of Hepatic Surgery and Liver Transplantation Center, The Third Affiliated Hospital of Sun Yat-sen University, Organ Transplantation Institute of Sun Yat-sen University, Organ Transplantation Research Center of Guangdong Province, Guangdong Province Engineering Laboratory for Transplantation Medicine, Guangzhou, 510630, China.
- Guangdong Provincial Key Laboratory of Liver Disease Research, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510630, China.
| | - Yongwei Hu
- Department of Hepatic Surgery and Liver Transplantation Center, The Third Affiliated Hospital of Sun Yat-sen University, Organ Transplantation Institute of Sun Yat-sen University, Organ Transplantation Research Center of Guangdong Province, Guangdong Province Engineering Laboratory for Transplantation Medicine, Guangzhou, 510630, China.
- Guangdong Provincial Key Laboratory of Liver Disease Research, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510630, China.
- Laboratory of Biomaterials and Translational Medicine Center for Nanomedicine, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510630, China.
| | - Binsheng Fu
- Department of Hepatic Surgery and Liver Transplantation Center, The Third Affiliated Hospital of Sun Yat-sen University, Organ Transplantation Institute of Sun Yat-sen University, Organ Transplantation Research Center of Guangdong Province, Guangdong Province Engineering Laboratory for Transplantation Medicine, Guangzhou, 510630, China.
- Guangdong Provincial Key Laboratory of Liver Disease Research, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510630, China.
| |
Collapse
|
8
|
Lei YP, Wang J, Yin PL, Jia H, Ma WZ. Melatonin ameliorates heat stress-induced oxidative apoptosis in mouse spermatocytes via autophagy and ferroptosis pathways. Cell Stress Chaperones 2025; 30:100078. [PMID: 40262713 DOI: 10.1016/j.cstres.2025.100078] [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: 01/25/2025] [Revised: 03/29/2025] [Accepted: 04/14/2025] [Indexed: 04/24/2025] Open
Abstract
Testicular heat stress is a critical factor contributing to male infertility, with spermatocytes exhibiting heightened sensitivity to temperature elevation. This study systematically elucidates the protective mechanisms of melatonin against heat stress-induced spermatocyte injury. In a murine heat stress model, melatonin intervention significantly reduced testicular accumulation of malondialdehyde (MDA) induced by heat stress, enhanced the activities of catalase (CAT) and superoxide dismutase (SOD), and suppressed germ cell apoptosis by downregulating the pro-apoptotic protein Bax and upregulating GPX4 expression. Sycp3 immunohistochemistry demonstrated that melatonin significantly improved spermatocyte structural integrity. In the GC-2spd (ts) spermatocyte cell line model, melatonin treatment markedly reduced MDA levels and alleviated heat stress-induced oxidative apoptosis and proliferation inhibition by downregulating key apoptotic proteins (Bax, Caspase-3, and cleaved-Caspase-3). Mechanistic studies revealed that melatonin restores autophagic balance by modulating the expression of autophagy-related proteins LC3-I, LC3-II, and P62. Concurrently, melatonin downregulated ferroptosis markers P53 and COX2, inhibiting ferroptosis by blocking DNA damage response and inflammatory amplification pathways. Melatonin synergistically maintained cellular redox homeostasis by downregulating the NRF2/HO-1 pathway and upregulating GPX4 expression, significantly reducing Fe²⁺ accumulation and ameliorating iron metabolism dysregulation. This study unveils the molecular mechanisms by which melatonin mitigates testicular heat stress injury through a multitarget regulatory network, providing novel therapeutic strategies for clinical intervention in heat stress-associated infertility.
Collapse
Affiliation(s)
- Yi-Ping Lei
- Key Laboratory of Fertility Preservation and Maintenance of the Ministry of Education, Key Laboratory of Reproduction and Genetics of Ningxia Hui Autonomous Region, School of Basic Medical Science, Ningxia Medical University, Yinchuan 750004, China
| | - Jia Wang
- Key Laboratory of Fertility Preservation and Maintenance of the Ministry of Education, Key Laboratory of Reproduction and Genetics of Ningxia Hui Autonomous Region, School of Basic Medical Science, Ningxia Medical University, Yinchuan 750004, China
| | - Peng-Luo Yin
- Key Laboratory of Fertility Preservation and Maintenance of the Ministry of Education, Key Laboratory of Reproduction and Genetics of Ningxia Hui Autonomous Region, School of Basic Medical Science, Ningxia Medical University, Yinchuan 750004, China
| | - Hua Jia
- Key Laboratory of Fertility Preservation and Maintenance of the Ministry of Education, Key Laboratory of Reproduction and Genetics of Ningxia Hui Autonomous Region, School of Basic Medical Science, Ningxia Medical University, Yinchuan 750004, China.
| | - Wen-Zhi Ma
- Key Laboratory of Fertility Preservation and Maintenance of the Ministry of Education, Key Laboratory of Reproduction and Genetics of Ningxia Hui Autonomous Region, School of Basic Medical Science, Ningxia Medical University, Yinchuan 750004, China.
| |
Collapse
|
9
|
Xie G, Yang C, Pang X, Wu TC, Gu X. Cancer Cell-Intrinsic Type I Interferon Signaling Promotes Antitumor Immunity in Head and Neck Squamous Cell Carcinoma. Cancers (Basel) 2025; 17:1279. [PMID: 40282455 PMCID: PMC12025670 DOI: 10.3390/cancers17081279] [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: 02/20/2025] [Revised: 04/06/2025] [Accepted: 04/08/2025] [Indexed: 04/29/2025] Open
Abstract
Background: The cyclic GMP-AMP synthase (cGAS)-type I interferon (IFN-I) pathway detects cytoplasmic DNA and triggers immune responses. Cancer cells often suppress this pathway to evade immune surveillance; however, its therapeutic potential remains unclear. Methods: Mouse oral squamous cell carcinoma models, representing a prominent subtype of head and neck squamous cell carcinoma (HNSCC), were employed in this study. Flow cytometry, Western blot, ELISA, and PCR were used for analysis. Results: We found that immune-unresponsive MOC2 tumors exhibited a deficiency of antigen-presenting cells and cytotoxic T lymphocytes, along with a significant suppression of the cGAS-IFN-I pathway, compared to immune-responsive MOC1 tumors. An MOC2-conditioned medium impaired the differentiation of bone marrow-derived cells into dendritic cells (DCs), reducing the expression of DC markers as well as class I and II major histocompatibility complex (MHC) molecules. The activation of the cGAS-IFN-I pathway in MOC2 cells, either through exogenous DNA or direct IFN-I expression, enhanced class I MHC expression and antigen presentation on MOC2 cells. Furthermore, IFNB1 expression in MOC2 cells induced apoptosis and upregulated chemokines, such as CXCL9 and CXCL10, which recruit immune cells. In immunocompetent mice, IFNB1 expression suppressed MOC2 tumor growth by attracting DCs and T cells, an effect amplified by co-expressing the granulocyte-macrophage colony-stimulating factor. Conclusions: These findings highlight the potential of enhancing cancer cell-intrinsic cGAS-IFN-I signaling to improve tumor immune surveillance and control the progression of immune-cold HNSCC tumors.
Collapse
Affiliation(s)
- Guiqin Xie
- Department of Oral Pathology, Howard University, 600 W Street NW, Washington, DC 20059, USA; (C.Y.); (X.P.)
- Cancer Center, Howard University, 2041 Georgia Avenue NW, Washington, DC 20059, USA
| | - Cuicui Yang
- Department of Oral Pathology, Howard University, 600 W Street NW, Washington, DC 20059, USA; (C.Y.); (X.P.)
- Cancer Center, Howard University, 2041 Georgia Avenue NW, Washington, DC 20059, USA
| | - Xiaowu Pang
- Department of Oral Pathology, Howard University, 600 W Street NW, Washington, DC 20059, USA; (C.Y.); (X.P.)
| | - Tzyy-Choou Wu
- Pathology, Oncology, Obstetrics & Gynecology, and Molecular Microbiology & Immunology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA;
| | - Xinbin Gu
- Department of Oral Pathology, Howard University, 600 W Street NW, Washington, DC 20059, USA; (C.Y.); (X.P.)
- Cancer Center, Howard University, 2041 Georgia Avenue NW, Washington, DC 20059, USA
| |
Collapse
|
10
|
Xue YX, Chen YJ, Qin MZ, Shang FF, Lu YT, Sun YH, Bian LG, Zhang A, Yu Y, Ding CY. Microglial STING activation promotes neuroinflammation and pathological changes in experimental mice with intracerebral haemorrhage. Acta Pharmacol Sin 2025:10.1038/s41401-025-01540-8. [PMID: 40200123 DOI: 10.1038/s41401-025-01540-8] [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: 12/02/2024] [Accepted: 03/11/2025] [Indexed: 04/10/2025]
Abstract
Neuroinflammation, a significant contributor to secondary brain injury, plays a critical role in the pathological process and prognosis of intracerebral haemorrhage (ICH). Thus, developing interventions to mitigate secondary neuroimmune deterioration is of paramount importance. Currently, no effective immunomodulatory drugs are available for ICH. The cyclic GMP-AMP synthase (cGAS)-stimulator of interferon genes (STING) pathway is a recently identified innate immune-sensing pathway primarily expressed in microglia within the central nervous system (CNS) that has been implicated in the pathophysiology of various neurological diseases. In this study we investigated the role of cGAS-STING pathway in ICH. A collagenase model of ICH was established in mice. Brain tissues were collected on D1 or D3 post-ICH. We observed a significant increase in double-stranded (dsDNA) levels and activation of the cGAS-STING pathway in the perihaematomal region of ICH mice. Administration of a blood brain barrier-permeable STING antagonist H151 (10 mg/kg, i.p.) significantly decreased cell apoptosis, alleviated hematoma growth, and improved motor impairments in ICH mice, accompanied by inhibiting the STING pathway in microglia, reducing production/release of the cGAS-STING pathway downstream inflammatory factors, NLRP3 inflammasome activation and gasdermin D (GSDMD)-induced microglial pyroptosis. Microglial Sting conditional knockout significantly mitigated ICH-induced neuroinflammatory responses, pathological damage and motor dysfunction. These results suggest that the microglial STING pathway promotes brain pathological damage and behavioural defects in ICH mice by activating the NLRP3 inflammasome and microglial pyroptosis. The STING pathway may serve as a potential therapeutic target for ICH-induced secondary brain injury.
Collapse
Affiliation(s)
- Yu-Xiao Xue
- Department of Neurosurgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200020, China
- Shanghai Frontiers Science Center of Drug Target Identification and Delivery, National Key Laboratory of Innovative Immunotherapy, School of Pharmaceutical Sciences, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Yi-Jun Chen
- Shanghai Frontiers Science Center of Drug Target Identification and Delivery, National Key Laboratory of Innovative Immunotherapy, School of Pharmaceutical Sciences, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Mei-Zhen Qin
- Shanghai Frontiers Science Center of Drug Target Identification and Delivery, National Key Laboratory of Innovative Immunotherapy, School of Pharmaceutical Sciences, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Fan-Fan Shang
- Shanghai Frontiers Science Center of Drug Target Identification and Delivery, National Key Laboratory of Innovative Immunotherapy, School of Pharmaceutical Sciences, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Yi-Ting Lu
- Shanghai Frontiers Science Center of Drug Target Identification and Delivery, National Key Laboratory of Innovative Immunotherapy, School of Pharmaceutical Sciences, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Yu-Hao Sun
- Department of Neurosurgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200020, China
| | - Liu-Guan Bian
- Department of Neurosurgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200020, China.
| | - Ao Zhang
- Shanghai Frontiers Science Center of Drug Target Identification and Delivery, National Key Laboratory of Innovative Immunotherapy, School of Pharmaceutical Sciences, Shanghai Jiao Tong University, Shanghai, 200240, China.
| | - Yang Yu
- Shanghai Frontiers Science Center of Drug Target Identification and Delivery, National Key Laboratory of Innovative Immunotherapy, School of Pharmaceutical Sciences, Shanghai Jiao Tong University, Shanghai, 200240, China.
| | - Chun-Yong Ding
- Shanghai Frontiers Science Center of Drug Target Identification and Delivery, National Key Laboratory of Innovative Immunotherapy, School of Pharmaceutical Sciences, Shanghai Jiao Tong University, Shanghai, 200240, China.
| |
Collapse
|
11
|
Noruzi S, Mohammadi R, Jamialahmadi K. CRISPR/Cas9 system: a novel approach to overcome chemotherapy and radiotherapy resistance in cancer. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025; 398:3373-3408. [PMID: 39560750 DOI: 10.1007/s00210-024-03480-2] [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: 07/24/2024] [Accepted: 09/21/2024] [Indexed: 11/20/2024]
Abstract
Cancer presents a global health challenge with rising incidence and mortality. Despite treatment advances in cancer therapy, radiotherapy and chemotherapy remained the most common treatments for all types of cancers. However, resistance phenotype in cancer cells leads to unsatisfactory results in the efficiency of therapeutic strategies. Therefore, researchers strive to propose effective solutions to overcome treatment failure, which requires a deep knowledge of treatment-resistant mechanisms. The progression and occurrence of tumors can be attributed to gene mutation. Over the past decade, the emergence of clustered regularly interspaced short palindromic repeats and CRISPR-associated protein 9 (CRISPR/Cas9) genome editing has revolutionized cancer research. This versatile technology enables cancer modeling, manipulation of specific DNA sequences, and genome-wide screening. CRISPR/Cas9 is an effective tool for identifying radio- and chemoresistance genes and offering potential adjunctive treatments to overcome tumor recurrence after chemo- and radiotherapy. This article aims to explain the potential of the CRISPR/Cas9 system in improving the effectiveness of chemo- and radiotherapy and ultimately overcoming treatment failure.
Collapse
Affiliation(s)
- Somaye Noruzi
- Department of Medical Biotechnology and Nanotechnology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Rezvan Mohammadi
- Student Research Committee, Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Cellular and Molecular Biology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Khadijeh Jamialahmadi
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
12
|
Chiang JC, Shang Z, Rosales T, Cai L, Chen WM, Cai F, Vu H, Minna JD, Ni M, Davis AJ, Timmerman RD, DeBerardinis RJ, Zhang Y. Lipoylation inhibition enhances radiation control of lung cancer by suppressing homologous recombination DNA damage repair. SCIENCE ADVANCES 2025; 11:eadt1241. [PMID: 40073141 PMCID: PMC11900879 DOI: 10.1126/sciadv.adt1241] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/12/2024] [Accepted: 02/05/2025] [Indexed: 03/14/2025]
Abstract
Lung cancer exhibits altered metabolism, influencing its response to radiation. To investigate the metabolic regulation of radiation response, we conducted a comprehensive, metabolic-wide CRISPR-Cas9 loss-of-function screen using radiation as selection pressure in human non-small cell lung cancer. Lipoylation emerged as a key metabolic target for radiosensitization, with lipoyltransferase 1 (LIPT1) identified as a top hit. LIPT1 covalently conjugates mitochondrial 2-ketoacid dehydrogenases with lipoic acid, facilitating enzymatic functions involved in the tricarboxylic acid cycle. Inhibiting lipoylation, either through genetic LIPT1 knockout or a lipoylation inhibitor (CPI-613), enhanced tumor control by radiation. Mechanistically, lipoylation inhibition increased 2-hydroxyglutarate, leading to H3K9 trimethylation, disrupting TIP60 recruitment and ataxia telangiectasia mutated (ATM)-mediated DNA damage repair signaling, impairing homologous recombination repair. In summary, our findings reveal a critical role of LIPT1 in regulating DNA damage and chromosome stability and may suggest a means to enhance therapeutic outcomes with DNA-damaging agents.
Collapse
Affiliation(s)
- Jui-Chung Chiang
- Department of Radiation Oncology, Harold C. Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Zengfu Shang
- Department of Radiation Oncology, Harold C. Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Tracy Rosales
- Howard Hughes Medical Institute, Eugene McDermott Center for Human Growth and Development, and Children’s Medical Center Research Institute, University of Texas Southwestern Medical Center, Dallas, TX 75235, USA
| | - Ling Cai
- Peter O’Donnell, Jr. School of Public Health, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Wei-Min Chen
- Department of Radiation Oncology, Harold C. Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Feng Cai
- Howard Hughes Medical Institute, Eugene McDermott Center for Human Growth and Development, and Children’s Medical Center Research Institute, University of Texas Southwestern Medical Center, Dallas, TX 75235, USA
| | - Hieu Vu
- Howard Hughes Medical Institute, Eugene McDermott Center for Human Growth and Development, and Children’s Medical Center Research Institute, University of Texas Southwestern Medical Center, Dallas, TX 75235, USA
| | - John D. Minna
- Hamon Center for Therapeutic Oncology Research, Departments of Internal Medicine and Pharmacology, Harold C. Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Min Ni
- Howard Hughes Medical Institute, Eugene McDermott Center for Human Growth and Development, and Children’s Medical Center Research Institute, University of Texas Southwestern Medical Center, Dallas, TX 75235, USA
| | - Anthony J. Davis
- Department of Radiation Oncology, Harold C. Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Robert D. Timmerman
- Department of Radiation Oncology, Harold C. Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Ralph J. DeBerardinis
- Howard Hughes Medical Institute, Eugene McDermott Center for Human Growth and Development, and Children’s Medical Center Research Institute, University of Texas Southwestern Medical Center, Dallas, TX 75235, USA
| | - Yuanyuan Zhang
- Department of Radiation Oncology, Harold C. Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| |
Collapse
|
13
|
Li B, Ming H, Qin S, Nice EC, Dong J, Du Z, Huang C. Redox regulation: mechanisms, biology and therapeutic targets in diseases. Signal Transduct Target Ther 2025; 10:72. [PMID: 40050273 PMCID: PMC11885647 DOI: 10.1038/s41392-024-02095-6] [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: 06/20/2024] [Revised: 10/09/2024] [Accepted: 11/21/2024] [Indexed: 03/09/2025] Open
Abstract
Redox signaling acts as a critical mediator in the dynamic interactions between organisms and their external environment, profoundly influencing both the onset and progression of various diseases. Under physiological conditions, oxidative free radicals generated by the mitochondrial oxidative respiratory chain, endoplasmic reticulum, and NADPH oxidases can be effectively neutralized by NRF2-mediated antioxidant responses. These responses elevate the synthesis of superoxide dismutase (SOD), catalase, as well as key molecules like nicotinamide adenine dinucleotide phosphate (NADPH) and glutathione (GSH), thereby maintaining cellular redox homeostasis. Disruption of this finely tuned equilibrium is closely linked to the pathogenesis of a wide range of diseases. Recent advances have broadened our understanding of the molecular mechanisms underpinning this dysregulation, highlighting the pivotal roles of genomic instability, epigenetic modifications, protein degradation, and metabolic reprogramming. These findings provide a foundation for exploring redox regulation as a mechanistic basis for improving therapeutic strategies. While antioxidant-based therapies have shown early promise in conditions where oxidative stress plays a primary pathological role, their efficacy in diseases characterized by complex, multifactorial etiologies remains controversial. A deeper, context-specific understanding of redox signaling, particularly the roles of redox-sensitive proteins, is critical for designing targeted therapies aimed at re-establishing redox balance. Emerging small molecule inhibitors that target specific cysteine residues in redox-sensitive proteins have demonstrated promising preclinical outcomes, setting the stage for forthcoming clinical trials. In this review, we summarize our current understanding of the intricate relationship between oxidative stress and disease pathogenesis and also discuss how these insights can be leveraged to optimize therapeutic strategies in clinical practice.
Collapse
Affiliation(s)
- Bowen Li
- Department of Biotherapy, Institute of Oxidative Stress Medicine, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital and West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu, PR China
| | - Hui Ming
- Department of Biotherapy, Institute of Oxidative Stress Medicine, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital and West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu, PR China
| | - Siyuan Qin
- Department of Biotherapy, Institute of Oxidative Stress Medicine, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital and West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu, PR China
- Frontiers Medical Center, Tianfu Jincheng Laboratory, Chengdu, PR China
| | - Edouard C Nice
- Department of Biochemistry and Molecular Biology, Monash University, Clayton, VIC, Australia
| | - Jingsi Dong
- Department of Thoracic Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan, China.
- Lung Cancer Center/Lung Cancer Institute, West China Hospital, Sichuan University, Chengdu, Sichuan, China.
| | - Zhongyan Du
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, China.
- Key Laboratory of Blood-stasis-toxin Syndrome of Zhejiang Province, Hangzhou, China.
| | - Canhua Huang
- Department of Biotherapy, Institute of Oxidative Stress Medicine, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital and West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu, PR China.
- Frontiers Medical Center, Tianfu Jincheng Laboratory, Chengdu, PR China.
| |
Collapse
|
14
|
Sun S, Qian S, Wang R, Zhao M, Li R, Gu W, Zhao M, Qian C, Liu L, Tang X, Li Y, Shi H, Pan Y, Xiao H, Yang K, Hu C, Huang Y, Wei L, Zhang Y, Ji J, Chen Y, Liu H. Targeting GOLPH3L improves glioblastoma radiotherapy by regulating STING-NLRP3-mediated tumor immune microenvironment reprogramming. Sci Transl Med 2025; 17:eado0020. [PMID: 40043140 DOI: 10.1126/scitranslmed.ado0020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Revised: 08/04/2024] [Accepted: 02/12/2025] [Indexed: 04/18/2025]
Abstract
Radiotherapy (RT) has been the standard-of-care treatment for patients with glioblastoma (GBM); however, the clinical effectiveness is hindered by therapeutic resistance. Here, we demonstrated that the tumor immune microenvironment (TIME) exhibited immunosuppressive properties and high expression of Golgi phosphoprotein 3 like (GOLPH3L) in RT-resistant GBM. Our study showed that GOLPH3L interacted with stimulator of interferon genes (STING) at the aspartic acid residue 184 in Golgi after RT, leading to coat protein complex II-mediated retrograde transport of STING from Golgi to endoplasmic reticulum. This suppressed the STING-NOD-like receptor thermal protein domain associated protein 3 (NLRP3)-mediated pyroptosis, resulting in suppressive TIME, driving GBM resistance to RT. Genetic GOLPH3L ablation in RT-resistant GBM cells augmented antitumor immunity and overcame tumor resistance to RT. Moreover, we have identified a small molecular inhibitor of GOLPH3L, vitamin B5 calcium (VB5), which improved the therapeutic efficacy of RT and immune checkpoint blockade by inducing a robust antitumor immune response in mouse models. Clinically, patients with GBM treated with VB5 exhibited improved responses to RT. Thus, reprogramming the TIME by targeting GOLPH3L may offer a potential opportunity to improve RT in GBM.
Collapse
Affiliation(s)
- Shuo Sun
- Department of Neurosurgery, First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Shiyu Qian
- Department of Pharmacy, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing 210008, China
| | - Ran Wang
- Department of Neurosurgery, Affiliated Brain Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Mengya Zhao
- Department of Immunology, Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi People's Hospital, Wuxi Medical Center, Nanjing Medical University, Nanjing 211166, China
- Research Center of Surgery, Nanjing BenQ Medical Center, Affiliated BenQ Hospital of Nanjing Medical University, Department of Immunology, Nanjing Medical University, Nanjing 211166, China
| | - Ran Li
- Department of Immunology, Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi People's Hospital, Wuxi Medical Center, Nanjing Medical University, Nanjing 211166, China
| | - Wei Gu
- Department of Neurosurgery, First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Mengjie Zhao
- Department of Neurosurgery, Affiliated Brain Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Chunfa Qian
- Department of Neurosurgery, Affiliated Brain Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Liang Liu
- Department of Neurosurgery, Affiliated Brain Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Xianglong Tang
- Department of Neurosurgery, Affiliated Brain Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Yangyang Li
- Department of Neurosurgery, Affiliated Brain Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Hui Shi
- Department of Neurosurgery, First Hospital of Lianyungang, Lianyungang 222000, China
| | - Yunsong Pan
- Department of Neurosurgery, First Hospital of Lianyungang, Lianyungang 222000, China
| | - Hong Xiao
- Department of Neurosurgery, Affiliated Brain Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Kun Yang
- Department of Neurosurgery, Affiliated Brain Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Chupeng Hu
- Department of Immunology, Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi People's Hospital, Wuxi Medical Center, Nanjing Medical University, Nanjing 211166, China
| | - Yedi Huang
- Department of Immunology, Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi People's Hospital, Wuxi Medical Center, Nanjing Medical University, Nanjing 211166, China
| | - Liangnian Wei
- Department of Immunology, Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi People's Hospital, Wuxi Medical Center, Nanjing Medical University, Nanjing 211166, China
| | - Yuhan Zhang
- Department of Immunology, Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi People's Hospital, Wuxi Medical Center, Nanjing Medical University, Nanjing 211166, China
| | - Jing Ji
- Department of Neurosurgery, First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
- Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing 211166, China
| | - Yun Chen
- Department of Immunology, Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi People's Hospital, Wuxi Medical Center, Nanjing Medical University, Nanjing 211166, China
- Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing 211166, China
| | - Hongyi Liu
- Department of Neurosurgery, Affiliated Brain Hospital of Nanjing Medical University, Nanjing 210029, China
| |
Collapse
|
15
|
Wootton LM, Morgan EL. Ubiquitin and ubiquitin-like proteins in HPV-driven carcinogenesis. Oncogene 2025; 44:713-723. [PMID: 40011575 PMCID: PMC11888991 DOI: 10.1038/s41388-025-03310-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2024] [Revised: 01/20/2025] [Accepted: 02/12/2025] [Indexed: 02/28/2025]
Abstract
Persistent infection with high-risk (HR) human papillomaviruses (HPVs) is responsible for approximately 5% of cancer cases worldwide, including a growing number of oropharyngeal and anogenital cancers. The major HPV oncoproteins, E6 and E7, act together to manipulate cellular pathways involved in the regulation of proliferation, the cell cycle and cell survival, ultimately driving malignant transformation. Protein ubiquitination and the ubiquitin proteasome system (UPS) is often deregulated upon viral infection and in oncogenesis. HPV E6 and E7 interact with and disrupt multiple components of the ubiquitination machinery to promote viral persistence, which can also result in cellular transformation and the formation of tumours. This review highlights the ways in which HPV manipulates protein ubiquitination and the ubiquitin-like protein pathways and how this contributes to tumour development. Furthermore, we discuss how understanding the interactions between HPV and the protein ubiquitination could lead to novel therapeutic targets that are of urgent need in HPV+ carcinomas.
Collapse
Affiliation(s)
| | - Ethan L Morgan
- School of Life Sciences, University of Sussex, Brighton, UK.
| |
Collapse
|
16
|
Kelley V, Baro M, Gasperi W, Ader N, Lea H, Lee H, Phoomak C, Kabeche L, King M, Contessa J. Loss of JAK1 Function Causes G2/M Cell Cycle Defects Vulnerable to Kif18a Inhibition. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.19.638911. [PMID: 40060568 PMCID: PMC11888196 DOI: 10.1101/2025.02.19.638911] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 03/16/2025]
Abstract
To gain insight into biological mechanisms that cause resistance to DNA damage, we performed parallel pooled genetic CRISPR-Cas9 screening for survival in high risk HNSCC subtypes. Surprisingly, and in addition to ATM, DNAPK, and NFKB signaling, JAK1 was identified as a driver of tumor cell radiosensitivity. Knockout of JAK1 in HNSCC increases cell survival by enhancing the DNA damage-induced G2 arrest, and both knockout and JAK1 inhibition with abrocitinib prevent subsequent formation of radiation-induced micronuclei. Loss of JAK1 function does not affect canonical CDK1 signaling but does reduce activation of PLK1 and AURKA, kinases that regulate both G2 and M phase progression. Correspondingly, JAK1 KO was found to cause mitotic defects using both EdU labeling and live cell imaging techniques. Given this insight, we evaluated Kif18a inhibition as an approach to exacerbate mitotic stress and enhance the efficacy of radiation. These studies establish Kif18a inhibition as a novel strategy to counteract therapeutic resistance to DNA damage mediated by G2 cell cycle arrest.
Collapse
Affiliation(s)
- Vanessa Kelley
- Department of Therapeutic Radiology, Yale University School of Medicine, New Haven, CT 06510 USA
- Department of Pharmacology, Yale University School of Medicine, New Haven, CT 06510 USA
| | - Marta Baro
- Department of Therapeutic Radiology, Yale University School of Medicine, New Haven, CT 06510 USA
| | - William Gasperi
- Department of Therapeutic Radiology, Yale University School of Medicine, New Haven, CT 06510 USA
| | - Nicholas Ader
- Department of Biology, University of North Carolina Greensboro, Greensboro, NC 27412 USA
- Department of Cell Biology, Yale University School of Medicine, New Haven, CT 06510 USA
| | - Hannah Lea
- Department of Therapeutic Radiology, Yale University School of Medicine, New Haven, CT 06510 USA
- Program in Translational Biomedicine, Yale University School of Medicine, New Haven, CT 06510 USA
| | - Hojin Lee
- Department of Therapeutic Radiology, Yale University School of Medicine, New Haven, CT 06510 USA
| | - Chatchai Phoomak
- Department of Therapeutic Radiology, Yale University School of Medicine, New Haven, CT 06510 USA
- Department of Biology, Chulalongkorn University, Bangkok 10330, Thailand
| | - Lilian Kabeche
- Department Molecular Biophysics and Biomedicine, Yale University School of Medicine, New Haven, CT 06510 USA
| | - Megan King
- Department of Cell Biology, Yale University School of Medicine, New Haven, CT 06510 USA
| | - Joseph Contessa
- Department of Therapeutic Radiology, Yale University School of Medicine, New Haven, CT 06510 USA
- Department of Pharmacology, Yale University School of Medicine, New Haven, CT 06510 USA
| |
Collapse
|
17
|
Dominguez-Vigil IG, Banik K, Baro M, Contessa JN, Hayman TJ. PLK4 inhibition as a strategy to enhance non-small cell lung cancer radiosensitivity. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.19.638860. [PMID: 40027806 PMCID: PMC11870518 DOI: 10.1101/2025.02.19.638860] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/05/2025]
Abstract
Lung cancer is the leading cause of cancer-related mortality worldwide. Non-small cell lung cancer (NSCLC) is the most common subtype of lung cancer and comprises 85% of cases. Despite treatment advances, local control after curative-intent chemoradiation for NSCLC remains suboptimal. Polo-like kinase 4 (PLK4) is a serine-threonine kinase that plays a critical role in the regulation of centrosome duplication and cell cycle progression and is overexpressed in NSCLC, thus, making it a potential therapeutic target. CFI-400945 is an orally available PLK4 inhibitor currently undergoing clinical trial evaluation. As radiation causes cell death primarily by mitotic catastrophe, a process enhanced by alterations in centrosome amplification, we hypothesized that disruption of the mitotic machinery by inhibition of PLK4 would enhance the effects of radiation in NSCLC. PLK4 inhibition by CFI-400945 resulted in radiosensitization of NSCLC cell lines. In contrast, CFI-400945 had no effect on the radiosensitivity of normal lung fibroblasts. PLK4 inhibition did not affect cell-cycle phase distribution prior to radiation, but rather the combination of CFI-400945 and radiation resulted in increased G2/M cell cycle arrest, increased centrosome amplification, and a concomitant increase in cell death through mitotic catastrophe. Lastly, CFI-400945 treatment enhanced the radiation-induced tumor growth delay of NSCLC tumor xenografts. These data indicate that targeting PLK4 is a novel approach to enhance the radiation sensitivity of NSCLC in vitro and in vivo through potentiation of centrosome amplification and cell death through mitotic catastrophe.
Collapse
|
18
|
Sun Y, Aliyari SR, Parvatiyar K, Wang L, Zhen A, Sun W, Han X, Zhang A, Kato E, Shi H, De Schutter E, McBride WH, French SW, Cheng G. STING directly interacts with PAR to promote apoptosis upon acute ionizing radiation-mediated DNA damage. Cell Death Differ 2025:10.1038/s41418-025-01457-z. [PMID: 39939798 DOI: 10.1038/s41418-025-01457-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Revised: 01/13/2025] [Accepted: 01/29/2025] [Indexed: 02/14/2025] Open
Abstract
Acute ionizing radiation (IR) causes severe DNA damage, leading to cell cycle arrest, cell death, and activation of the innate immune system. The role and signaling pathway of stimulator of interferon genes (STING) in IR-induced tissue damage and cell death are not well understood. This study revealed that STING is crucial for promoting apoptosis in response to DNA damage caused by acute IR both in vitro and in vivo. STING binds to poly (ADP‒ribose) (PAR) produced by activated poly (ADP‒ribose) polymerase-1 (PARP1) upon IR. Compared with that in WT cells, apoptosis was suppressed in Stinggt-/gt- cells. Excessive PAR production by PARP1 due to DNA damage enhances STING phosphorylation, and inhibiting PARP1 reduces cell apoptosis after IR. In vivo, IR-induced crypt cell death was significantly lower in Stinggt-/gt- mice or with low-dose PARP1 inhibitor, PJ34, resulting in substantial resistance to abdominal irradiation. STING deficiency or inhibition of PARP1 function can reduce the expression of the proapoptotic gene PUMA, decrease the localization of Bax on the mitochondrial membrane, and thus reduce cell apoptosis. Our findings highlight crucial roles for STING and PAR in the IR-mediated induction of apoptosis, which may have therapeutic implications for controlling radiation-induced apoptosis or acute radiation symptoms. STING responds to acute ionizing radiation-mediated DNA damage by directly binding to poly (ADP-ribose) (PAR) produced by activated poly (ADP-ribose) polymerase-1 (PARP1), and mainly induces cell apoptosis through Puma-Bax interaction. STING deficiency or reduced production of PAR protected mice against Acute Radiation Syndrome.
Collapse
Affiliation(s)
- Yirong Sun
- CAS Key Laboratory of Regenerative Biology, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China.
- Department of Microbiology, Immunology and Molecular Genetics, University of California-Los Angeles, Los Angeles, CA, USA.
| | - Saba R Aliyari
- Department of Microbiology, Immunology and Molecular Genetics, University of California-Los Angeles, Los Angeles, CA, USA
| | - Kislay Parvatiyar
- Department of Microbiology, Immunology and Molecular Genetics, University of California-Los Angeles, Los Angeles, CA, USA
- Department of Microbiology & Immunology, Tulane University School of Medicine, New Orleans, LA, USA
| | - Lulan Wang
- Department of Microbiology, Immunology and Molecular Genetics, University of California-Los Angeles, Los Angeles, CA, USA
| | - Anjie Zhen
- Department of Microbiology, Immunology and Molecular Genetics, University of California-Los Angeles, Los Angeles, CA, USA
| | - Wei Sun
- CAS Key Laboratory of Regenerative Biology, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Xiaobo Han
- CAS Key Laboratory of Regenerative Biology, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Adele Zhang
- Department of Microbiology, Immunology and Molecular Genetics, University of California-Los Angeles, Los Angeles, CA, USA
| | - Ethan Kato
- Department of Microbiology, Immunology and Molecular Genetics, University of California-Los Angeles, Los Angeles, CA, USA
| | - Helen Shi
- Department of Microbiology, Immunology and Molecular Genetics, University of California-Los Angeles, Los Angeles, CA, USA
| | - Elena De Schutter
- Department of Microbiology, Immunology and Molecular Genetics, University of California-Los Angeles, Los Angeles, CA, USA
| | - William H McBride
- Department of Radiation Oncology, University of California-Los Angeles, Los Angeles, CA, USA
| | - Samuel W French
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California-Los Angeles, Los Angeles, CA, USA
| | - Genhong Cheng
- Department of Microbiology, Immunology and Molecular Genetics, University of California-Los Angeles, Los Angeles, CA, USA.
| |
Collapse
|
19
|
Wang Y, Han J, Zhu Y, Huang N, Qu N. New advances in the therapeutic strategy of head and neck squamous cell carcinoma: A review of latest therapies and cutting-edge research. Biochim Biophys Acta Rev Cancer 2025; 1880:189230. [PMID: 39608621 DOI: 10.1016/j.bbcan.2024.189230] [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: 09/17/2024] [Revised: 11/22/2024] [Accepted: 11/24/2024] [Indexed: 11/30/2024]
Abstract
Head and neck squamous cell carcinoma (HNSCC) is a common and aggressive malignancy with a poor prognosis, particularly when diagnosed at advanced stages. Despite progress in surgical, chemotherapeutic, and radiotherapeutic interventions, the five-year survival rate remains low due to high rates of recurrence and therapeutic resistance. This review explores recent advances in therapeutic strategies for HNSCC, focusing on targeted therapies, immunotherapy, and innovative drug delivery systems. Targeted therapies, such as EGFR inhibitors and PI3K/AKT/mTOR pathway inhibitors, offer promising options for overcoming HNSCC, though resistance challenges persist. Emerging treatments, including dual-target inhibitors and personalized therapeutic approaches, show potential in addressing these limitations. Immunotherapy, particularly PD-1/PD-L1 blockade, has achieved positive outcomes in a subset of patients, though overall response rates remain modest. Strategies aimed at enhancing immune responses, such as combination therapies and nanotechnology-based drug delivery systems, are actively being investigated to improve efficacy. This review also underscores the critical role of the tumor microenvironment and epithelial-mesenchymal transition (EMT) in HNSCC progression and therapeutic resistance. Novel approaches, including smart drug delivery systems utilizing nanotechnology and immune modulation, are opening new avenues for more personalized and effective treatments. Ongoing interdisciplinary research into molecular targets and advanced drug delivery techniques holds great promise for significantly improving patient outcomes in HNSCC.
Collapse
Affiliation(s)
- Yuting Wang
- Department of Head and Neck Surgery, Fudan University Shanghai Cancer Center, Shanghai, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Jing Han
- Department of Oral and Maxillofacial Head and Neck Oncology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, 639 Zhizaoju Rd., Huangpu District, Shanghai 200011, China
| | - Yongxue Zhu
- Department of Head and Neck Surgery, Fudan University Shanghai Cancer Center, Shanghai, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Naisi Huang
- Department of Head and Neck Surgery, Fudan University Shanghai Cancer Center, Shanghai, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Ning Qu
- Department of Head and Neck Surgery, Fudan University Shanghai Cancer Center, Shanghai, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China.
| |
Collapse
|
20
|
Chen Q, Yang Y, Ying X, Huang C, Chen J, Wang J, Wu Z, Zeng W, Miao C, Shi X, Nan Y, Huang Q, Ai K. Hierarchical Targeting Nanodrug with Holistic DNA Protection for Effective Treatment of Acute Kidney Injury. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2411254. [PMID: 39703158 PMCID: PMC11809360 DOI: 10.1002/advs.202411254] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/13/2024] [Revised: 11/08/2024] [Indexed: 12/21/2024]
Abstract
Acute kidney injury (AKI) manifests a hallmark pathological feature of extensive and severe DNA damage in renal tubules, primarily induced by the excessive of toxic reactive oxygen species (ROS) from the mitochondrial electron transport chain. The kidney's complex intricate physiological architecture and the heterogeneous intracellular environment pose significant challenges for effective sequential and high-resolution drug delivery-an urgent issue that remains unresolved. To address this, a hierarchical-targeting antioxidant nanodrug has been developed with a folic acid moiety (HAND) designed for high-resolution drug delivery in AKI treatment. For the first time, HAND enables sequential targeting from the kidney to the most severely damaged proximal tubular epithelial cells (PTECs), ultimately concentrating in the DNA-rich mitochondria and nucleus. As a result, HAND effectively scavenges ROS in situ, protecting both mitochondria and nuclei along with their vital genetic material. This action restores mitochondrial function, mitigates DNA oxidation and fragmentation, reduces apoptosis, and inhibits cGAS/STING-mediated sterile inflammation. Consequently, HAND demonstrates remarkable efficacy in safeguarding injured kidneys during AKI. Overall, this work pioneers a hierarchical, high-resolution antioxidant strategy, providing innovative guidance for the development of AKI therapies.
Collapse
Affiliation(s)
- Qiaohui Chen
- Department of PharmacyXiangya HospitalCentral South UniversityChangsha410008China
- Xiangya School of Pharmaceutical SciencesCentral South UniversityChangsha410013China
| | - Yongqi Yang
- Xiangya School of Pharmaceutical SciencesCentral South UniversityChangsha410013China
- Hunan Provincial Key Laboratory of Cardiovascular ResearchXiangya School of Pharmaceutical SciencesCentral South UniversityChangsha410013China
| | - Xiaohong Ying
- Xiangya School of Pharmaceutical SciencesCentral South UniversityChangsha410013China
- Hunan Provincial Key Laboratory of Cardiovascular ResearchXiangya School of Pharmaceutical SciencesCentral South UniversityChangsha410013China
| | - Changkun Huang
- Department of UrologyThe Second Xiangya HospitalCentral South UniversityChangshaHunan410011China
| | - Jianlin Chen
- Department of Pancreatic SurgeryXiangya HospitalCentral South UniversityChangsha410008China
- Department of General SurgeryXiangya HospitalCentral South UniversityChangsha410008China
| | - Jue Wang
- Xiangya School of Pharmaceutical SciencesCentral South UniversityChangsha410013China
- Hunan Provincial Key Laboratory of Cardiovascular ResearchXiangya School of Pharmaceutical SciencesCentral South UniversityChangsha410013China
| | - Ziyu Wu
- Xiangya School of Pharmaceutical SciencesCentral South UniversityChangsha410013China
- Hunan Provincial Key Laboratory of Cardiovascular ResearchXiangya School of Pharmaceutical SciencesCentral South UniversityChangsha410013China
| | - Wan Zeng
- Xiangya School of Pharmaceutical SciencesCentral South UniversityChangsha410013China
- Hunan Provincial Key Laboratory of Cardiovascular ResearchXiangya School of Pharmaceutical SciencesCentral South UniversityChangsha410013China
| | - Chenxi Miao
- Xiangya School of Pharmaceutical SciencesCentral South UniversityChangsha410013China
- Hunan Provincial Key Laboratory of Cardiovascular ResearchXiangya School of Pharmaceutical SciencesCentral South UniversityChangsha410013China
| | - Xiaojing Shi
- Xiangya School of Pharmaceutical SciencesCentral South UniversityChangsha410013China
- Hunan Provincial Key Laboratory of Cardiovascular ResearchXiangya School of Pharmaceutical SciencesCentral South UniversityChangsha410013China
| | - Yayun Nan
- Geriatric Medical CenterPeople's Hospital of Ningxia Hui Autonomous RegionYinchuanNingxia750002China
| | - Qiong Huang
- Department of PharmacyXiangya HospitalCentral South UniversityChangsha410008China
- National Clinical Research Center for Geriatric DisordersXiangya HospitalCentral South UniversityChangsha410008China
| | - Kelong Ai
- Xiangya School of Pharmaceutical SciencesCentral South UniversityChangsha410013China
- Hunan Provincial Key Laboratory of Cardiovascular ResearchXiangya School of Pharmaceutical SciencesCentral South UniversityChangsha410013China
- Key Laboratory of Aging‐related Bone and Joint Diseases Prevention and TreatmentMinistry of EducationXiangya HospitalCentral South UniversityChangsha410008China
| |
Collapse
|
21
|
Zhang J, Wang Z, Lin X, Gao X, Wang Q, Huang R, Ruan Y, Xu H, Tian L, Ling C, Shi R, Xu S, Chen K, Wu Y. Mn-Ce Symbiosis: Nanozymes with Multiple Active Sites Facilitate Scavenging of Reactive Oxygen Species (ROS) Based on Electron Transfer and Confinement Anchoring. Angew Chem Int Ed Engl 2025; 64:e202416686. [PMID: 39327805 DOI: 10.1002/anie.202416686] [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: 08/30/2024] [Revised: 09/19/2024] [Accepted: 09/23/2024] [Indexed: 09/28/2024]
Abstract
Regulating appropriate valence states of metal active centers, such as Ce3+/Ce4+ and Mn3+/Mn2+, as well as surface vacancy defects, is crucial for enhancing the catalytic activity of cerium-based and manganese-based nanozymes. Drawing inspiration from the efficient substance exchange in rhizobia-colonized root cells of legumes, we developed a symbiosis nanozyme system with rhizobia-like CeOx nanoclusters robustly anchored onto root-like Mn3O4 nanosupports (CeOx/Mn3O4). The process of "substance exchange" between Ce and Mn atoms-reminiscent of electron transfer-not only fine-tunes the metal active sites to achieve optimal Ce3+/Ce4+ and Mn3+/Mn2+ ratios but also enhances the vacancy ratio through interface defect engineering. Additionally, the confinement anchoring of CeOx on Mn3O4 ensures efficient electron transfer in catalytic reactions. The final CeOx/Mn3O4 nanozyme demonstrates potent catalase-like (CAT-like) and superoxide dismutase-like (SOD-like) activities, excelling in both chemical settings and cellular environments with high reactive oxygen species (ROS) levels. This research not only unveils a novel material adept at effectively eliminating ROS but also presents an innovative approach for amplifying the efficacy of nanozymes.
Collapse
Affiliation(s)
- Juan Zhang
- Department of Endocrinology, Institute of Endocrine and Metabolic Diseases, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230026, China
- Deep Space Exploration Laboratory/, School of Chemistry and Materials Science, University of Science and Technology of China, Hefei, 230026, China
- Key Laboratory of Precision and Intelligent Chemistry/, School of Chemistry and Materials Science, University of Science and Technology of China, Hefei, 230026, China
| | - Zhihua Wang
- Department of Endocrinology, Institute of Endocrine and Metabolic Diseases, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230026, China
| | - Xingen Lin
- Department of Endocrinology, Institute of Endocrine and Metabolic Diseases, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230026, China
- Deep Space Exploration Laboratory/, School of Chemistry and Materials Science, University of Science and Technology of China, Hefei, 230026, China
- Key Laboratory of Precision and Intelligent Chemistry/, School of Chemistry and Materials Science, University of Science and Technology of China, Hefei, 230026, China
| | - Xiaoping Gao
- Department of Endocrinology, Institute of Endocrine and Metabolic Diseases, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230026, China
- Deep Space Exploration Laboratory/, School of Chemistry and Materials Science, University of Science and Technology of China, Hefei, 230026, China
- Key Laboratory of Precision and Intelligent Chemistry/, School of Chemistry and Materials Science, University of Science and Technology of China, Hefei, 230026, China
| | - Qiuping Wang
- School of Food and Biological Engineering, Hefei University of Technology, Hefei, 230009, China
| | - Rui Huang
- Department of Endocrinology, Institute of Endocrine and Metabolic Diseases, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230026, China
- Deep Space Exploration Laboratory/, School of Chemistry and Materials Science, University of Science and Technology of China, Hefei, 230026, China
- Key Laboratory of Precision and Intelligent Chemistry/, School of Chemistry and Materials Science, University of Science and Technology of China, Hefei, 230026, China
| | - Yaner Ruan
- Department of Endocrinology, Institute of Endocrine and Metabolic Diseases, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230026, China
- Deep Space Exploration Laboratory/, School of Chemistry and Materials Science, University of Science and Technology of China, Hefei, 230026, China
- Key Laboratory of Precision and Intelligent Chemistry/, School of Chemistry and Materials Science, University of Science and Technology of China, Hefei, 230026, China
| | - Haonan Xu
- National Synchrotron Radiation Laboratory, University of Science and Technology of China, Hefei, 230009, China
| | - Lin Tian
- Department of Endocrinology, Institute of Endocrine and Metabolic Diseases, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230026, China
- Deep Space Exploration Laboratory/, School of Chemistry and Materials Science, University of Science and Technology of China, Hefei, 230026, China
- Key Laboratory of Precision and Intelligent Chemistry/, School of Chemistry and Materials Science, University of Science and Technology of China, Hefei, 230026, China
| | - Chen Ling
- Department of Endocrinology, Institute of Endocrine and Metabolic Diseases, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230026, China
- Deep Space Exploration Laboratory/, School of Chemistry and Materials Science, University of Science and Technology of China, Hefei, 230026, China
- Key Laboratory of Precision and Intelligent Chemistry/, School of Chemistry and Materials Science, University of Science and Technology of China, Hefei, 230026, China
| | - Ran Shi
- Department of Endocrinology, Institute of Endocrine and Metabolic Diseases, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230026, China
- Deep Space Exploration Laboratory/, School of Chemistry and Materials Science, University of Science and Technology of China, Hefei, 230026, China
- Key Laboratory of Precision and Intelligent Chemistry/, School of Chemistry and Materials Science, University of Science and Technology of China, Hefei, 230026, China
| | - Suowen Xu
- Department of Endocrinology, Institute of Endocrine and Metabolic Diseases, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230026, China
- Anhui Provincial Key Laboratory of Metabolic Health and Panvascular Diseases, Hefei, 230009, China
| | - Kong Chen
- Department of Endocrinology, Institute of Endocrine and Metabolic Diseases, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230026, China
- Deep Space Exploration Laboratory/, School of Chemistry and Materials Science, University of Science and Technology of China, Hefei, 230026, China
- Key Laboratory of Precision and Intelligent Chemistry/, School of Chemistry and Materials Science, University of Science and Technology of China, Hefei, 230026, China
| | - Yuen Wu
- Department of Endocrinology, Institute of Endocrine and Metabolic Diseases, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230026, China
- Deep Space Exploration Laboratory/, School of Chemistry and Materials Science, University of Science and Technology of China, Hefei, 230026, China
- Anhui Provincial Key Laboratory of Metabolic Health and Panvascular Diseases, Hefei, 230009, China
- Key Laboratory of Precision and Intelligent Chemistry/, School of Chemistry and Materials Science, University of Science and Technology of China, Hefei, 230026, China
| |
Collapse
|
22
|
Wang T, Bai M, Geng W, Adeli M, Ye L, Cheng C. Bioinspired artificial antioxidases for efficient redox homeostasis and maxillofacial bone regeneration. Nat Commun 2025; 16:856. [PMID: 39833195 PMCID: PMC11746915 DOI: 10.1038/s41467-025-56179-0] [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: 06/19/2024] [Accepted: 01/10/2025] [Indexed: 01/22/2025] Open
Abstract
Reconstructing large, inflammatory maxillofacial defects using stem cell-based therapy faces challenges from adverse microenvironments, including high levels of reactive oxygen species (ROS), inadequate oxygen, and intensive inflammation. Here, inspired by the reaction mechanisms of intracellular antioxidant defense systems, we propose the de novo design of an artificial antioxidase using Ru-doped layered double hydroxide (Ru-hydroxide) for efficient redox homeostasis and maxillofacial bone regeneration. Our studies demonstrate that Ru-hydroxide consists hydroxyls-synergistic monoatomic Ru centers, which efficiently react with oxygen species and collaborate with hydroxyls for rapid proton and electron transfer, thus exhibiting efficient, broad-spectrum, and robust ROS scavenging performance. Moreover, Ru-hydroxide can effectively sustain stem cell viability and osteogenic differentiation in elevated ROS environments, modulating the inflammatory microenvironment during bone tissue regeneration in male mice. We believe this Ru-hydroxide development offers a promising avenue for designing antioxidase-like materials to treat various inflammation-associated disorders, including arthritis, diabetic wounds, enteritis, and bone fractures.
Collapse
Affiliation(s)
- Ting Wang
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu, China
| | - Mingru Bai
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- Department of Cariology and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Wei Geng
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu, China
| | - Mohsen Adeli
- Institute of Chemistry and Biochemistry, Free University of Berlin, Berlin, Germany
| | - Ling Ye
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- Department of Cariology and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Chong Cheng
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu, China.
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China.
| |
Collapse
|
23
|
Dong R, Wang J, Guan R, Sun J, Jin P, Shen J. Role of Oxidative Stress in the Occurrence, Development, and Treatment of Breast Cancer. Antioxidants (Basel) 2025; 14:104. [PMID: 39857438 PMCID: PMC11760893 DOI: 10.3390/antiox14010104] [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: 12/13/2024] [Revised: 01/11/2025] [Accepted: 01/14/2025] [Indexed: 01/27/2025] Open
Abstract
Breast cancer is one of the most prevalent cancers worldwide. Recent studies have increasingly emphasized the role of oxidative stress in the initiation and progression of breast cancer. This article reviews how oxidative stress imbalance influences the occurrence and advancement of breast cancer, elucidating the intricate mechanisms through which reactive oxygen species (ROS) operate in this context and their potential therapeutic applications. By highlighting these critical insights, this review aims to enhance our understanding of oxidative stress as a potential target for innovative therapeutic strategies in the management of breast cancer.
Collapse
Affiliation(s)
- Rui Dong
- Yunnan Key Laboratory of Cell Metabolism and Diseases, Center for Life Sciences, School of Life Sciences, Yunnan University, Kunming 650091, China; (R.D.); (J.W.); (R.G.); (J.S.)
| | - Jing Wang
- Yunnan Key Laboratory of Cell Metabolism and Diseases, Center for Life Sciences, School of Life Sciences, Yunnan University, Kunming 650091, China; (R.D.); (J.W.); (R.G.); (J.S.)
| | - Ruiqi Guan
- Yunnan Key Laboratory of Cell Metabolism and Diseases, Center for Life Sciences, School of Life Sciences, Yunnan University, Kunming 650091, China; (R.D.); (J.W.); (R.G.); (J.S.)
| | - Jianwei Sun
- Yunnan Key Laboratory of Cell Metabolism and Diseases, Center for Life Sciences, School of Life Sciences, Yunnan University, Kunming 650091, China; (R.D.); (J.W.); (R.G.); (J.S.)
- State Key Laboratory for Conservation and Utilization of Bio-Resources in Yunnan, Yunnan University, Kunming 650091, China
| | - Ping Jin
- Yunnan Key Laboratory of Cell Metabolism and Diseases, Center for Life Sciences, School of Life Sciences, Yunnan University, Kunming 650091, China; (R.D.); (J.W.); (R.G.); (J.S.)
- State Key Laboratory for Conservation and Utilization of Bio-Resources in Yunnan, Yunnan University, Kunming 650091, China
| | - Junling Shen
- Yunnan Key Laboratory of Cell Metabolism and Diseases, Center for Life Sciences, School of Life Sciences, Yunnan University, Kunming 650091, China; (R.D.); (J.W.); (R.G.); (J.S.)
- Key Laboratory of Tumor Immunological Prevention and Treatment of Yunnan Province, Kunming 650051, China
| |
Collapse
|
24
|
Hong R, Han Y, Chen S. Advances in micro- and nano- delivery systems for increasing the stability, bioavailability and bioactivity of coenzyme Q 10. Crit Rev Food Sci Nutr 2025:1-18. [PMID: 39819160 DOI: 10.1080/10408398.2025.2450543] [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: 01/19/2025]
Abstract
Coenzyme Q10 acts as a liposoluble quinone compound in mitochondrial oxidative phosphorylation, serving as an electron carrier and protecting the cell membrane structure as an antioxidant. Coenzyme Q10 has notable health benefits, including anti-aging, anti-inflammatory, prevention of cardiovascular diseases, and assistance in cancer treatment. However, its poor water solubility, unstable chemical properties, and low bioavailability significantly limit its application. This article reviewed the design and development processes of various delivery systems for coenzyme Q10, discussing the advantages and disadvantages of different delivery systems and their improvement strategies, including improvements in the stability and accessibility of emulsions, achieving higher penetration rates for oleogels, and reducing the use of toxic substances in the production process of liposomes. The mechanisms behind coenzyme Q10's low stability and bioavailability were analyzed, and the bioactivity and research prospects of coenzyme Q10 were also discussed. In summary, this review offered valuable insights into the design and application of delivery systems for coenzyme Q10, which may provide a reference for its development and application in pharmaceuticals, cosmetics, health products, and other industries in the future.
Collapse
Affiliation(s)
- Ruoxuan Hong
- Hubei Key Laboratory for Processing and Transformation of Agricultural Products, College of Food Science and Engineering, Wuhan Polytechnic University, Wuhan, Hubei, China
- School of Public Health, Wuhan University, Wuhan, Hubei, China
| | - Yahong Han
- Hubei Key Laboratory for Processing and Transformation of Agricultural Products, College of Food Science and Engineering, Wuhan Polytechnic University, Wuhan, Hubei, China
| | - Shuai Chen
- School of Public Health, Wuhan University, Wuhan, Hubei, China
| |
Collapse
|
25
|
Yuan H, Qiu C, Wang X, Wang P, Yi L, Peng X, Xu X, Huang W, Bai Y, Wei J, Ma J, Wong YK, Fu C, Xiao W, Chen C, Long Y, Li Z, Wang J. Engineering Semiconducting Polymeric Nanoagonists Potentiate cGAS-STING Pathway Activation and Elicit Long Term Memory Against Recurrence in Breast Cancer. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2025; 37:e2406662. [PMID: 39629527 DOI: 10.1002/adma.202406662] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Revised: 11/10/2024] [Indexed: 01/30/2025]
Abstract
Triple-negative breast cancer has an immunologically "cold" microenvironment, which leads to resistance to current immunotherapy. The activation of stimulator of interferon genes (STING) pathway has been thought a promising strategy to enhance immunotherapy efficacy. In this study, we adopted a comprehensive strategy that integrates innate immune responses with tumor-targeting photothermal therapy (PTT) to simultaneously tackle multiple immune-suppressive mechanisms in breast cancer. This semiconducting polymeric nanoagonists (DPTT-Mn Lipo NPs) mediated PTT can effectively initiate tumor cell apoptosis and induce ICD, thereby reprogramming the immunosuppressive TME and activating STING. We confirmed the modulation of the TME through the PTT-mediated ICD effect and the transactivation of the cGAS-STING pathway in immune cells of the TME due to the released dsDNA via ICD, such as macrophages and DCs. Indeed, DPTT-Mn Lipo NPs-mediated PTT promoted M1 polarization of tumor-associated macrophages, augmented T-cell infiltration, facilitated dendritic cell (DC) maturation, and regulated type I interferon factor secretion, leading to efficient tumor suppression. Most importantly, the combination of DPTT-Mn Lipo NPs-based PTT with a checkpoint blockade therapy (anti-PD-1) can elicit long-term immune memory besides tumor eradication. Collectively, this nano-system can systemically activate antitumor immunity through STING activation and potentially establish long-term memory against tumor recurrence.
Collapse
Affiliation(s)
- Haitao Yuan
- Department of Hyperbaric Oxygen Medicine, Guangdong Provincial Clinical Research Center for Geriatrics, Shenzhen Clinical Research Center for Geriatric, Shenzhen People's Hospital, The First Affiliated Hospital, Southern University of Science and Technology, Shenzhen, Guangdong, 518020, P. R. China
- Center for Drug Research and Development Guangdong Provincial Key Laboratory of Advanced Drug Delivery System, Guangdong Pharmaceutical University, Guangzhou, 510006, P. R. China
| | - Chong Qiu
- Department of Hyperbaric Oxygen Medicine, Guangdong Provincial Clinical Research Center for Geriatrics, Shenzhen Clinical Research Center for Geriatric, Shenzhen People's Hospital, The First Affiliated Hospital, Southern University of Science and Technology, Shenzhen, Guangdong, 518020, P. R. China
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao-di Herbs, Artemisinin Research Center, and Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, P. R. China
| | - Xiaoxian Wang
- Department of Hyperbaric Oxygen Medicine, Guangdong Provincial Clinical Research Center for Geriatrics, Shenzhen Clinical Research Center for Geriatric, Shenzhen People's Hospital, The First Affiliated Hospital, Southern University of Science and Technology, Shenzhen, Guangdong, 518020, P. R. China
| | - Peili Wang
- Department of Hyperbaric Oxygen Medicine, Guangdong Provincial Clinical Research Center for Geriatrics, Shenzhen Clinical Research Center for Geriatric, Shenzhen People's Hospital, The First Affiliated Hospital, Southern University of Science and Technology, Shenzhen, Guangdong, 518020, P. R. China
- National Clinical Research Center for Chinese Medicine Cardiology, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, 100700, P. R. China
| | - Letai Yi
- Inner Mongolia Medical University, Hohhot, Inner Mongolia, 010000, P. R. China
| | - Xin Peng
- Ningbo Municipal Hospital of TCM, Affiliated Hospital of Zhejiang Chinese Medical University, Ningbo, 315010, P. R. China
| | - Xiaolong Xu
- Department of Critical Care Medicine, Shenzhen People's Hospital, The Second Clinical Medical College of Jinan University, The First Affiliated Hospital of Southern University of Science and Technology, Shenzhen, 518020, P. R. China
| | - Wei Huang
- Department of Hyperbaric Oxygen Medicine, Guangdong Provincial Clinical Research Center for Geriatrics, Shenzhen Clinical Research Center for Geriatric, Shenzhen People's Hospital, The First Affiliated Hospital, Southern University of Science and Technology, Shenzhen, Guangdong, 518020, P. R. China
| | - Yunmeng Bai
- Department of Critical Care Medicine, Shenzhen People's Hospital, The Second Clinical Medical College of Jinan University, The First Affiliated Hospital of Southern University of Science and Technology, Shenzhen, 518020, P. R. China
| | - Jinxi Wei
- Department of Hyperbaric Oxygen Medicine, Guangdong Provincial Clinical Research Center for Geriatrics, Shenzhen Clinical Research Center for Geriatric, Shenzhen People's Hospital, The First Affiliated Hospital, Southern University of Science and Technology, Shenzhen, Guangdong, 518020, P. R. China
| | - Jingbo Ma
- Department of Hyperbaric Oxygen Medicine, Guangdong Provincial Clinical Research Center for Geriatrics, Shenzhen Clinical Research Center for Geriatric, Shenzhen People's Hospital, The First Affiliated Hospital, Southern University of Science and Technology, Shenzhen, Guangdong, 518020, P. R. China
| | - Yin Kwan Wong
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117600, Singapore
| | - Chunjin Fu
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao-di Herbs, Artemisinin Research Center, and Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, P. R. China
| | - Wei Xiao
- Department of Hyperbaric Oxygen Medicine, Guangdong Provincial Clinical Research Center for Geriatrics, Shenzhen Clinical Research Center for Geriatric, Shenzhen People's Hospital, The First Affiliated Hospital, Southern University of Science and Technology, Shenzhen, Guangdong, 518020, P. R. China
- Center for Drug Research and Development Guangdong Provincial Key Laboratory of Advanced Drug Delivery System, Guangdong Pharmaceutical University, Guangzhou, 510006, P. R. China
| | - Chunbo Chen
- Department of Hyperbaric Oxygen Medicine, Guangdong Provincial Clinical Research Center for Geriatrics, Shenzhen Clinical Research Center for Geriatric, Shenzhen People's Hospital, The First Affiliated Hospital, Southern University of Science and Technology, Shenzhen, Guangdong, 518020, P. R. China
- Department of Critical Care Medicine, Shenzhen People's Hospital, The Second Clinical Medical College of Jinan University, The First Affiliated Hospital of Southern University of Science and Technology, Shenzhen, 518020, P. R. China
| | - Ying Long
- Department of Hyperbaric Oxygen Medicine, Guangdong Provincial Clinical Research Center for Geriatrics, Shenzhen Clinical Research Center for Geriatric, Shenzhen People's Hospital, The First Affiliated Hospital, Southern University of Science and Technology, Shenzhen, Guangdong, 518020, P. R. China
| | - Zhijie Li
- Department of Hyperbaric Oxygen Medicine, Guangdong Provincial Clinical Research Center for Geriatrics, Shenzhen Clinical Research Center for Geriatric, Shenzhen People's Hospital, The First Affiliated Hospital, Southern University of Science and Technology, Shenzhen, Guangdong, 518020, P. R. China
| | - Jigang Wang
- Department of Hyperbaric Oxygen Medicine, Guangdong Provincial Clinical Research Center for Geriatrics, Shenzhen Clinical Research Center for Geriatric, Shenzhen People's Hospital, The First Affiliated Hospital, Southern University of Science and Technology, Shenzhen, Guangdong, 518020, P. R. China
- Center for Drug Research and Development Guangdong Provincial Key Laboratory of Advanced Drug Delivery System, Guangdong Pharmaceutical University, Guangzhou, 510006, P. R. China
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao-di Herbs, Artemisinin Research Center, and Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, P. R. China
- State Key Laboratory of Antiviral Drugs, School of Pharmacy, Henan University, Kaifeng, 475004, P. R. China
| |
Collapse
|
26
|
Saini S, Gurung P. A comprehensive review of sensors of radiation-induced damage, radiation-induced proximal events, and cell death. Immunol Rev 2025; 329:e13409. [PMID: 39425547 PMCID: PMC11742653 DOI: 10.1111/imr.13409] [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] [Indexed: 10/21/2024]
Abstract
Radiation, a universal component of Earth's environment, is categorized into non-ionizing and ionizing forms. While non-ionizing radiation is relatively harmless, ionizing radiation possesses sufficient energy to ionize atoms and disrupt DNA, leading to cell damage, mutation, cancer, and cell death. The extensive use of radionuclides and ionizing radiation in nuclear technology and medical applications has sparked global concern for their capacity to cause acute and chronic illnesses. Ionizing radiation induces DNA damage either directly through strand breaks and base change or indirectly by generating reactive oxygen species (ROS) and reactive nitrogen species (RNS) via radiolysis of water. This damage triggers a complex cellular response involving recognition of DNA damage, cell cycle arrest, DNA repair mechanisms, release of pro-inflammatory cytokines, and cell death. This review focuses on the mechanisms of radiation-induced cellular damage, recognition of DNA damage and subsequent activation of repair processes, and the critical role of the innate immune response in resolution of the injury. Emphasis is placed on pattern recognition receptors (PRRs) and related receptors that detect damage-associated molecular patterns (DAMPs) and initiate downstream signaling pathways. Radiation-induced cell death pathways are discussed in detail. Understanding these processes is crucial for developing strategies to mitigate the harmful effects of radiation and improve therapeutic outcomes.
Collapse
Affiliation(s)
- Saurabh Saini
- Inflammation ProgramUniversity of IowaIowa CityIowaUSA
- Department of Internal MedicineUniversity of IowaIowa CityIowaUSA
- Iowa City Veterans Affairs (VA) Medical CenterIowa CityIowaUSA
| | - Prajwal Gurung
- Inflammation ProgramUniversity of IowaIowa CityIowaUSA
- Department of Internal MedicineUniversity of IowaIowa CityIowaUSA
- Iowa City Veterans Affairs (VA) Medical CenterIowa CityIowaUSA
- Interdisciplinary Graduate Program in Human ToxicologyUniversity of IowaIowa CityIowaUSA
- Immunology Graduate ProgramUniversity of IowaIowa CityIowaUSA
- Center for Immunology and Immune Based DiseaseUniversity of IowaIowa CityIowaUSA
| |
Collapse
|
27
|
Wen F, Han Y, Zhang H, Zhao Z, Wang W, Chen F, Qin W, Ju J, An L, Meng Y, Yang J, Tang Y, Zhao Y, Zhang H, Li F, Bai W, Xu Y, Zhou Z, Jiao S. Epstein-Barr virus infection upregulates extracellular OLFM4 to activate YAP signaling during gastric cancer progression. Nat Commun 2024; 15:10543. [PMID: 39627192 PMCID: PMC11615309 DOI: 10.1038/s41467-024-54850-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Accepted: 11/24/2024] [Indexed: 12/06/2024] Open
Abstract
Extracellular vesicles (EVs) are known to mediate cell communications and shape tumor microenvironment. Compared to the well-studied small EVs, the function of large microvesicles (MVs) during tumorigenesis is poorly understood. Here we show the proteome of MVs in Epstein-Barr virus (EBV)-associated gastric cancer (EBVaGC), and identify olfactomedin 4 (OLFM4) is induced by EBV infection and secreted via MVs to promote tumor progression through Hippo signaling. Specifically, OLFM4 is a target gene of the cGAS-STING pathway, and EBV infection activates cGAS-STING pathway and increases OLFM4 expression. Moreover, MV-carried OLFM4 binds with the extracellular cadherin domain of FAT1, thereby impairing its intracellular interaction with MST1 and leading to YAP activation in recipient cells. Together, our study not only reveals a regulatory mechanism though which viral infection is coupled via MVs with intercellular control of the Hippo signaling, but also highlights the OLFM4-Hippo axis as a therapeutic target for EBV-associated cancers.
Collapse
Affiliation(s)
- Fuping Wen
- Department of Stomatology, Shanghai Tenth People's Hospital, Tongji University Cancer Center, Tongji University School of Medicine, Shanghai, 200072, China
| | - Yi Han
- Department of Stomatology, Shanghai Tenth People's Hospital, Tongji University Cancer Center, Tongji University School of Medicine, Shanghai, 200072, China
| | - Hui Zhang
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Zhongshan Hospital, Fudan University, Shanghai, 200438, China
| | - Zhangting Zhao
- Department of Stomatology, Shanghai Tenth People's Hospital, Tongji University Cancer Center, Tongji University School of Medicine, Shanghai, 200072, China
| | - Wenjia Wang
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Zhongshan Hospital, Fudan University, Shanghai, 200438, China
| | - Fan Chen
- CAS Center for Excellence in Molecular Cell Science, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, 200031, China
| | - Weimin Qin
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Zhongshan Hospital, Fudan University, Shanghai, 200438, China
| | - Junyi Ju
- Department of Stomatology, Shanghai Tenth People's Hospital, Tongji University Cancer Center, Tongji University School of Medicine, Shanghai, 200072, China
| | - Liwei An
- Department of Stomatology, Shanghai Tenth People's Hospital, Tongji University Cancer Center, Tongji University School of Medicine, Shanghai, 200072, China
| | - Yan Meng
- Department of Stomatology, Shanghai Tenth People's Hospital, Tongji University Cancer Center, Tongji University School of Medicine, Shanghai, 200072, China
| | - Jie Yang
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Zhongshan Hospital, Fudan University, Shanghai, 200438, China
| | - Yang Tang
- Department of Stomatology, Shanghai Tenth People's Hospital, Tongji University Cancer Center, Tongji University School of Medicine, Shanghai, 200072, China
| | - Yun Zhao
- CAS Center for Excellence in Molecular Cell Science, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, 200031, China
| | - Huanhu Zhang
- Department of Digestive Sciences, Shanxi Cancer Hospital, Taiyuan, 030001, China
| | - Feng Li
- Department of Digestive Sciences, Shanxi Cancer Hospital, Taiyuan, 030001, China
| | - Wenqi Bai
- Department of Digestive Sciences, Shanxi Cancer Hospital, Taiyuan, 030001, China.
| | - Yuanzhi Xu
- Department of Stomatology, Shanghai Tenth People's Hospital, Tongji University Cancer Center, Tongji University School of Medicine, Shanghai, 200072, China.
| | - Zhaocai Zhou
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Zhongshan Hospital, Fudan University, Shanghai, 200438, China.
- Collaborative Innovation Center for Cancer Personalized Medicine, School of Public Health, Nanjing Medical University, Nanjing, 211166, China.
| | - Shi Jiao
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Zhongshan Hospital, Fudan University, Shanghai, 200438, China.
| |
Collapse
|
28
|
Wei M, Li Q, Li S, Wang D, Wang Y. Multifaceted roles of cGAS-STING pathway in the lung cancer: from mechanisms to translation. PeerJ 2024; 12:e18559. [PMID: 39588006 PMCID: PMC11587877 DOI: 10.7717/peerj.18559] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2024] [Accepted: 10/31/2024] [Indexed: 11/27/2024] Open
Abstract
Lung cancer (LC) remains one of the most prevalent and lethal malignancies globally, with a 5-year survival rate for advanced cases persistently below 10%. Despite the significant advancements in immunotherapy, a substantial proportion of patients with advanced LC fail to respond effectively to these treatments, highlighting an urgent need for novel immunotherapeutic targets. The cyclic GMP-AMP synthase (cGAS)-stimulator of interferon genes (STING) pathway has gained prominence as a potential target for improving LC immunotherapy due to its pivotal role in enhancing anti-tumor immune responses, augmenting tumor antigen presentation, and promoting T cell infiltration. However, emerging evidence also suggests that the cGAS-STING pathway may have pro-tumorigenic effects in the context of LC. This review aims to provide a comprehensive analysis of the cGAS-STING pathway, including its biological composition, activation mechanisms, and physiological functions, as well as its dual roles in LC and the current and emerging LC treatment strategies that target the pathway. By addressing these aspects, we intend to highlight the potential of the cGAS-STING pathway as a novel immunotherapeutic target, while also considering the challenges and future directions for its clinical application.
Collapse
Affiliation(s)
- Mingming Wei
- School of Basic Medical Sciences, State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Qingzhou Li
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Shengrong Li
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Dong Wang
- School of Basic Medical Sciences, State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Yumei Wang
- School of Basic Medical Sciences, State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| |
Collapse
|
29
|
Xiao R, Zhao W, Lin W, Xiao Y, Ren J, Zhou Y, Meng W, Bi E, Jiang L. Bendamustine-rituximab elicits dual tumoricidal and immunomodulatory responses via cGAS-STING activation in diffuse large B-cell lymphoma. J Immunother Cancer 2024; 12:e009212. [PMID: 39521616 PMCID: PMC11551994 DOI: 10.1136/jitc-2024-009212] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/13/2024] [Indexed: 11/16/2024] Open
Abstract
BACKGROUND Bendamustine-rituximab (BR) therapy stands out as a promising alternative for elderly patients with diffuse large B-cell lymphoma (DLBCL), demonstrating notable efficacy when conventional regimens pose challenges. Despite its clinical success, the intricate mechanisms underlying BR therapy have remained elusive. METHODS DLBCL cell lines were used to investigate the mechanism of BR therapy in vitro. RNA-seq and Western blot were used to explore the target pathways of BR therapy. STING was knocked out using Crispr-cas9 and inhibited using H-151 to investigate its role in BR therapy. Bulk RNA-seq and single-cell RNA-seq data from patients were analyzed to investigate the association between STING and pyroptosis pathways, validated using STING downregulated cells. Flow cytometry, transwell experiments and co-culture experiments were performed to investigate the inflammatory phenotype of DLBCL cells after BR treatment and its effect on T-cell recruitment and activation. RESULTS This study elucidates that BR elicits direct tumoricidal effects by promoting apoptosis and inducing cell cycle arrest. The synergistic impact with rituximab is further potentiated by complement addition, demonstrating the pivotal role of in vivo antibody-dependent cellular cytotoxicity. Moreover, our investigation reveals that, through a cGAS-STING-dependent pathway, prolonged exposure to BR induces pyroptosis in DLBCL cells. Activation of the cGAS-STING pathway by BR therapy triggers the release of inflammatory factors and upregulates major histocompatibility complex molecules, shaping an immunologically hot tumor microenvironment. CONCLUSIONS This unique dual influence not only directly targets DLBCL cells but also engages the patient's immune system, paving the way for innovative combination therapies. The study provides comprehensive insights into the multifaceted actions of BR in DLBCL, offering a foundation for refined and personalized treatment strategies in elderly patients.
Collapse
Affiliation(s)
- Ruipei Xiao
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Guangdong Provincial Key Laboratory of Single Cell Technology and Application, Southern Medical University, Guangzhou, Guangdong, China
- Department of General Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Wenli Zhao
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Guangdong Provincial Key Laboratory of Single Cell Technology and Application, Southern Medical University, Guangzhou, Guangdong, China
| | - Wei Lin
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Guangdong Provincial Key Laboratory of Single Cell Technology and Application, Southern Medical University, Guangzhou, Guangdong, China
| | - Yudian Xiao
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Guangdong Provincial Key Laboratory of Single Cell Technology and Application, Southern Medical University, Guangzhou, Guangdong, China
| | - Jie Ren
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Guangdong Provincial Key Laboratory of Single Cell Technology and Application, Southern Medical University, Guangzhou, Guangdong, China
| | - Yang Zhou
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Guangdong Provincial Key Laboratory of Single Cell Technology and Application, Southern Medical University, Guangzhou, Guangdong, China
| | - Wei Meng
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Guangdong Provincial Key Laboratory of Single Cell Technology and Application, Southern Medical University, Guangzhou, Guangdong, China
| | - Enguang Bi
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Guangdong Provincial Key Laboratory of Single Cell Technology and Application, Southern Medical University, Guangzhou, Guangdong, China
- Department of Urology, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Ling Jiang
- Department of Hematology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| |
Collapse
|
30
|
Zhao Y, Qin C, Lin C, Li Z, Zhao B, Li T, Zhang X, Wang W. Pancreatic ductal adenocarcinoma cells reshape the immune microenvironment: Molecular mechanisms and therapeutic targets. Biochim Biophys Acta Rev Cancer 2024; 1879:189183. [PMID: 39303859 DOI: 10.1016/j.bbcan.2024.189183] [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: 06/09/2024] [Revised: 08/23/2024] [Accepted: 09/13/2024] [Indexed: 09/22/2024]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a digestive system malignancy characterized by challenging early detection, limited treatment alternatives, and generally poor prognosis. Although there have been significant advancements in immunotherapy for hematological malignancies and various solid tumors in recent decades, with impressive outcomes in recent preclinical and clinical trials, the effectiveness of these therapies in treating PDAC continues to be modest. The unique immunological microenvironment of PDAC, especially the abnormal distribution, complex composition, and variable activation states of tumor-infiltrating immune cells, greatly restricts the effectiveness of immunotherapy. Undoubtedly, integrating data from both preclinical models and human studies helps accelerate the identification of reliable molecules and pathways responsive to targeted biological therapies and immunotherapies, thereby continuously optimizing therapeutic combinations. In this review, we delve deeply into how PDAC cells regulate the immune microenvironment through complex signaling networks, affecting the quantity and functional status of immune cells to promote immune escape and tumor progression. Furthermore, we explore the multi-modal immunotherapeutic strategies currently under development, emphasizing the transformation of the immunosuppressive environment into an anti-tumor milieu by targeting specific molecular and cellular pathways, providing insights for the development of novel treatment strategies.
Collapse
Affiliation(s)
- Yutong Zhao
- Department of General Surgery, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing 100023, PR China; Key Laboratory of Research in Pancreatic Tumor, Chinese Academy of Medical Sciences, Beijing 100023, PR China; National Science and Technology Key Infrastructure on Translational Medicine in Peking Union Medical College Hospital, Beijing 100023, PR China
| | - Cheng Qin
- Department of General Surgery, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing 100023, PR China; Key Laboratory of Research in Pancreatic Tumor, Chinese Academy of Medical Sciences, Beijing 100023, PR China; National Science and Technology Key Infrastructure on Translational Medicine in Peking Union Medical College Hospital, Beijing 100023, PR China
| | - Chen Lin
- Department of General Surgery, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing 100023, PR China; Key Laboratory of Research in Pancreatic Tumor, Chinese Academy of Medical Sciences, Beijing 100023, PR China; National Science and Technology Key Infrastructure on Translational Medicine in Peking Union Medical College Hospital, Beijing 100023, PR China
| | - Zeru Li
- Department of General Surgery, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing 100023, PR China; Key Laboratory of Research in Pancreatic Tumor, Chinese Academy of Medical Sciences, Beijing 100023, PR China; National Science and Technology Key Infrastructure on Translational Medicine in Peking Union Medical College Hospital, Beijing 100023, PR China
| | - Bangbo Zhao
- Department of General Surgery, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing 100023, PR China; Key Laboratory of Research in Pancreatic Tumor, Chinese Academy of Medical Sciences, Beijing 100023, PR China; National Science and Technology Key Infrastructure on Translational Medicine in Peking Union Medical College Hospital, Beijing 100023, PR China
| | - Tianyu Li
- Department of General Surgery, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing 100023, PR China; Key Laboratory of Research in Pancreatic Tumor, Chinese Academy of Medical Sciences, Beijing 100023, PR China; National Science and Technology Key Infrastructure on Translational Medicine in Peking Union Medical College Hospital, Beijing 100023, PR China
| | - Xiangyu Zhang
- Department of General Surgery, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing 100023, PR China; Key Laboratory of Research in Pancreatic Tumor, Chinese Academy of Medical Sciences, Beijing 100023, PR China; National Science and Technology Key Infrastructure on Translational Medicine in Peking Union Medical College Hospital, Beijing 100023, PR China
| | - Weibin Wang
- Department of General Surgery, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing 100023, PR China; Key Laboratory of Research in Pancreatic Tumor, Chinese Academy of Medical Sciences, Beijing 100023, PR China; National Science and Technology Key Infrastructure on Translational Medicine in Peking Union Medical College Hospital, Beijing 100023, PR China.
| |
Collapse
|
31
|
Bhat AA, Gupta G, Afzal M, Thapa R, Ali H, Alqahtani SM, almalki WH, Kazmi I, Alzarea SI, Saleem S, Subramaniyan V. Polyphenol-Loaded Nano-carriers for Breast Cancer Therapy: A Comprehensive Review. BIONANOSCIENCE 2024; 14:4219-4237. [DOI: 10.1007/s12668-023-01288-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/20/2023] [Indexed: 11/22/2024]
|
32
|
Islam S, Islam MM, Akhand MRN, Park BY, Akanda MR. Recent advancements in cGAS-STING activation, tumor immune evasion, and therapeutic implications. Med Oncol 2024; 41:291. [PMID: 39419913 DOI: 10.1007/s12032-024-02539-7] [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: 08/15/2024] [Accepted: 10/07/2024] [Indexed: 10/19/2024]
Abstract
The cGAS-STING signaling pathway is indeed a pivotal component of the immune system and serve as a crucial link between innate and adaptive immune responses. STING is involved in the cellular response to pathogen invasion and DNA damage, and which has important consequences for host defense mechanisms and cancer regulation. Ongoing research aiming to modulate the cGAS-STING pathway for improved clinical outcomes in cancer and autoimmune diseases is underway. Indeed, the interaction between the cGAS-STING pathway and immune evasion mechanisms is a complex and critical aspect of cancer biology. Pathogens and various host factors can exploit this pathway to reduce the effectiveness of cancer therapies, particularly immunotherapies. Thus, immunotherapies or combination therapies may assist in overcoming the immune suppression and improving clinical outcomes. This review explores recent advancements in understanding the cGAS-STING signaling pathway, with particular emphasis on its activation mechanisms and role in tumor immune evasion. The dual role of the pathway in boosting immune responses while simultaneously enabling tumors to evade the immune system makes it a crucial target for innovative cancer treatment approaches.Please confirm if the author names are presented accurately and in the correct sequence (given name, middle name/initial, family name). Author 2 Given name: [Md Mazedul] Last name [Islam], Author 3 Given name: [Mst Rubaiat Nazneen] Last name [Akhand] and Author 5 Given name: [Md Rashedunnabi] Last name [Akanda]. Also, kindly confirm the details in the metadata are correct.AQ1: Here Author 4 given name: [Byung-Yong] Last name [Park] is missing. Metadata are correct.
Collapse
Affiliation(s)
- Saiful Islam
- Department of Physiology, Sylhet Agricultural University, Sylhet, 3100, Bangladesh
| | - Md Mazedul Islam
- Faculty of Veterinary, Animal and Biomedical Sciences, Sylhet Agricultural University, Sylhet, 3100, Bangladesh
| | | | - Byung-Yong Park
- Institute of Animal Transplantation, College of Veterinary Medicine, Jeonbuk National University, Iksan, 54596, South Korea
| | - Md Rashedunnabi Akanda
- Department of Pharmacology and Toxicology, Sylhet Agricultural University, Sylhet, 3100, Bangladesh.
| |
Collapse
|
33
|
Lu X, Li X, Li L, Han C, Li S. Advances in the prerequisite and consequence of STING downstream signalosomes. MEDICAL REVIEW (2021) 2024; 4:435-451. [PMID: 39444795 PMCID: PMC11495525 DOI: 10.1515/mr-2024-0016] [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] [Received: 02/20/2024] [Accepted: 04/11/2024] [Indexed: 10/25/2024]
Abstract
The cyclic GMP-AMP synthase (cGAS)-stimulator of interferon genes (STING) pathway is an evolving DNA-sensing mechanism involved in innate immunity and pathogen defense that has been optimized while remaining conserved. Aside from recognizing pathogens through conserved motifs, these receptors also detect aberrant or misplaced self-molecules as possible signs of perturbed homeostasis. Upon binding external or self-derived DNA, a mobile secondary messenger 2'3'-cyclic GMP-AMP (cGAMP) is produced by cGAS and in turn activates its adapter STING in the endoplasmic reticulum (ER). Resting-state or activated STING protein is finely restricted by multiple degradation machineries. The post-translational changes of the STING protein, along with the regulatory machinery of the secret routes, limit the onset, strength and sustention of STING signal. STING experiences a conformational shift and relocates with TBK1 from the ER to perinuclear vesicles containing transcription factors, provoking the transcription activity of IRF3/IFN-I and NF-κB pathways, as well as to initiate a number of cellular processes that have been shown to alter the immune landscape in cancer, such as autophagy, NLRP3 inflammasome, ER stress, and cell death. STING signal thus serves as a potent activator for immune mobilization yet also triggers immune-mediated pathology in tissues. Recent advances have established the vital role of STING in immune surveillance as well as tumorigenic process. This review provides an overview of the disparate outcomes of cancer attributed to the actions of pleiotropic and coordinated STING downstream signalosomes, along with the underlying mechanisms of STING function in pathologies, providing therapeutic implications for new approaches in hunt for the next generation of cancer immunotherapy base on STING.
Collapse
Affiliation(s)
- Xinliang Lu
- Institute of Immunology and Bone Marrow Transplantation Center of the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Xiaobing Li
- InvivoGen Ltd., Hong Kong Science and Technology Parks, Hong Kong, China
| | - Lili Li
- Center for Systems Medicine, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- Suzhou Institute of Systems Medicine, Suzhou, Jiangsu, China
| | - Chuanhui Han
- Peking University International Cancer Institute, Peking University Cancer Hospital and Institute, Health Science Center, Peking University, Beijing, China
| | - Sirui Li
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| |
Collapse
|
34
|
Heintzman DR, Sinard RC, Fisher EL, Ye X, Patterson AR, Elasy JH, Voss K, Chi C, Sugiura A, Rodriguez-Garcia GJ, Chowdhury NU, Arner EN, Krystoviak ES, Mason FM, Toudji YT, Steiner KK, Khan W, Olson LM, Jones AL, Hong HS, Bass L, Beier KL, Deng W, Lyssiotis CA, Newcomb DC, Bick AG, Rathmell WK, Wilson JT, Rathmell JC. Subset-specific mitochondrial stress and DNA damage shape T cell responses to fever and inflammation. Sci Immunol 2024; 9:eadp3475. [PMID: 39303018 PMCID: PMC11607909 DOI: 10.1126/sciimmunol.adp3475] [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: 03/20/2024] [Revised: 07/05/2024] [Accepted: 08/23/2024] [Indexed: 09/22/2024]
Abstract
Heat is a cardinal feature of inflammation, yet its impacts on immune cells remain uncertain. We show that moderate-grade fever temperatures (39°C) increased murine CD4 T cell metabolism, proliferation, and inflammatory effector activity while decreasing regulatory T cell suppressive capacity. However, heat-exposed T helper 1 (TH1) cells selectively developed mitochondrial stress and DNA damage that activated Trp53 and stimulator of interferon genes pathways. Although many TH1 cells subjected to such temperatures died, surviving TH1 cells exhibited increased mitochondrial mass and enhanced activity. Electron transport chain complex 1 (ETC1) was rapidly impaired under fever-range temperatures, a phenomenon that was specifically detrimental to TH1 cells. TH1 cells with elevated DNA damage and ETC1 signatures were also detected in human chronic inflammation. Thus, fever-relevant temperatures disrupt ETC1 to selectively drive apoptosis or adaptation of TH1 cells to maintain genomic integrity and enhance effector functions.
Collapse
Affiliation(s)
- Darren R Heintzman
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Rachael C Sinard
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, TN, USA
| | - Emilie L Fisher
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Xiang Ye
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Andrew R Patterson
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Joel H Elasy
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Kelsey Voss
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Channing Chi
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Ayaka Sugiura
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Gabriel J Rodriguez-Garcia
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Nowrin U Chowdhury
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Emily N Arner
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Evan S Krystoviak
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, TN, USA
- Cell Imaging Shared Resource, Vanderbilt University, Nashville, TN, USA
| | - Frank M Mason
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Yasmine T Toudji
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - KayLee K Steiner
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Wasay Khan
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Lana M Olson
- Vanderbilt Technologies for Advanced Genomics (VANTAGE), Vanderbilt University Medical Center, Nashville, TN, USA
| | - Angela L Jones
- Vanderbilt Technologies for Advanced Genomics (VANTAGE), Vanderbilt University Medical Center, Nashville, TN, USA
| | - Hanna S Hong
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI, USA
| | - Lindsay Bass
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Katherine L Beier
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Wentao Deng
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Costas A Lyssiotis
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI, USA
| | - Dawn C Newcomb
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
- Vanderbilt Center for Immunobiology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Alexander G Bick
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
- Vanderbilt Center for Immunobiology, Vanderbilt University Medical Center, Nashville, TN, USA
- Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN, USA
| | - W Kimryn Rathmell
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
- Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN, USA
| | - John T Wilson
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, TN, USA
- Vanderbilt Center for Immunobiology, Vanderbilt University Medical Center, Nashville, TN, USA
- Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Jeffrey C Rathmell
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA
- Vanderbilt Center for Immunobiology, Vanderbilt University Medical Center, Nashville, TN, USA
- Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN, USA
| |
Collapse
|
35
|
Gao Y, Sun W, Wang J, Zhao D, Tian H, Qiu Y, Ji S, Wang S, Fu Q, Zhang F, Zhang Z, Wang F, Shao J, Zheng S, Meng J. Oxidative stress induces ferroptosis in tendon stem cells by regulating mitophagy through cGAS-STING pathway. Int Immunopharmacol 2024; 138:112652. [PMID: 38986301 DOI: 10.1016/j.intimp.2024.112652] [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: 05/14/2024] [Revised: 06/27/2024] [Accepted: 07/06/2024] [Indexed: 07/12/2024]
Abstract
Tendinopathy is one of the most prevalent sports injury diseases in orthopedics. However, there is no effective treatment or medicine. Recently, the discovery of tendon stem cells (TSCs) provides a new perspective to find new therapeutic methods for Tendinopathy. Studies have shown that oxidative stress will inevitably cause TSCs injury during tendinopathy, but the mechanism has not been fully elucidated. Here, we report the oxidative damage of TSCs induced by H2O2 via ferroptosis, as well, treatment with H2O2 raised the proportion of mitochondria engulfed by autophagosomes in TSCs. The suppression of mitophagy by Mdivi-1 significantly attenuates the H2O2-induced ferroptosis in TSCs. Mechanically, H2O2 actives the cGAS-STING pathway, which can regulate the level of mitophagy. Interfering with cGAS could impair mitophagy and the classical ferroptotic events. In the rat model of tendinopathy, interference of cGAS could relieve tendon injury by inhibiting ferroptosis. Overall, these results provided novel implications to reveal the molecular mechanism of tendinopathy, by which pointed to cGAS as a potential therapeutic target for the treatment of tendinopathy.
Collapse
Affiliation(s)
- Yuanyuan Gao
- Nanjing Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing 210002, China; Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Wenshuang Sun
- Nanjing Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing 210002, China
| | - Junrui Wang
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Danli Zhao
- NanTong Health College of Jiangsu Province, Nantong 226000, China
| | - Haoyuan Tian
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Yangling Qiu
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Shufan Ji
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Shuqi Wang
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Qiuyu Fu
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Feng Zhang
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Zili Zhang
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Feixia Wang
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Jiangjuan Shao
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing 210023, China.
| | - Shizhong Zheng
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing 210023, China.
| | - Jia Meng
- Nanjing Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing 210002, China.
| |
Collapse
|
36
|
Shao C, Chen J, Qiang B, Ye J, Yan F, Zhu Y. The role of cGAS-STING signaling in the development and therapy of head and neck squamous cell carcinoma. Front Immunol 2024; 15:1451305. [PMID: 39295867 PMCID: PMC11408205 DOI: 10.3389/fimmu.2024.1451305] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Accepted: 08/19/2024] [Indexed: 09/21/2024] Open
Abstract
The cGAS-STING signaling pathway plays a critical role in innate immunity and defense against viral infections by orchestrating intracellular and adaptive immune responses to DNA. In the context of head and neck squamous cell carcinoma (HNSCC), this pathway has garnered significant attention due to its potential relevance in disease development and progression. HNSCC is strongly associated with risk factors such as smoking, heavy alcohol consumption, and human papillomavirus (HPV) infection. The presence or absence of HPV in HNSCC patients has been shown to have a profound impact on patient survival and prognosis, possibly due to the distinct biological characteristics of HPV-associated tumors. This review aims to provide a comprehensive overview of the current therapeutic approaches and challenges in HNSCC management, as well as the involvement of cGAS-STING signaling and its potential in the therapy of HNSCC. In addition, by advancing the present understanding of the mechanisms underlying this pathway, Activation of cGAS-STING-dependent inflammatory signaling downstream of chromosomal instability can exert both anti-tumoral and pro-tumoral effects in a cell-intrinsic manner, suggesting individualized therapy is of great importance. However, further exploration of the cGAS-STING signaling pathway is imperative for the effective management of HNSCC.
Collapse
Affiliation(s)
- Chengze Shao
- Department of Otolaryngology Head and Neck Surgery, The First Affiliated Hospital of Yangtze University, Jingzhou, Hubei, China
- Department of Oncology, The First Affiliated Hospital of Yangtze University, Jingzhou, Hubei, China
| | - Jiawen Chen
- Research Center of Biostatistics and Computational Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Bi Qiang
- Department of Otolaryngology Head and Neck Surgery, The First Affiliated Hospital of Yangtze University, Jingzhou, Hubei, China
| | - Junmei Ye
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
| | - Fangrong Yan
- Research Center of Biostatistics and Computational Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Yongbo Zhu
- Department of Otolaryngology Head and Neck Surgery, The First Affiliated Hospital of Yangtze University, Jingzhou, Hubei, China
| |
Collapse
|
37
|
Chen Z, Ji W, Feng W, Cui J, Wang Y, Li F, Chen J, Guo Z, Xia L, Zhu X, Niu X, Zhang Y, Li Z, Wong AST, Lu S, Xia W. PTPRT loss enhances anti-PD-1 therapy efficacy by regulation of STING pathway in non-small cell lung cancer. Sci Transl Med 2024; 16:eadl3598. [PMID: 39231239 DOI: 10.1126/scitranslmed.adl3598] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2023] [Revised: 04/18/2024] [Accepted: 08/08/2024] [Indexed: 09/06/2024]
Abstract
With the revolutionary progress of immune checkpoint inhibitors (ICIs) in non-small cell lung cancer, identifying patients with cancer who would benefit from ICIs has become critical and urgent. Here, we report protein tyrosine phosphatase receptor type T (PTPRT) loss as a precise and convenient predictive marker independent of PD-L1 expression for anti-PD-1/PD-L1 axis therapy. Anti-PD-1/PD-L1 axis treatment markedly increased progression-free survival in patients with PTPRT-deficient tumors. PTPRT-deficient tumors displayed cumulative DNA damage, increased cytosolic DNA release, and higher tumor mutation burden. Moreover, the tyrosine residue 240 of STING was identified as a direct substrate of PTPRT. PTPRT loss elevated phosphorylation of STING at Y240 and thus inhibited its proteasome-mediated degradation. PTPRT-deficient tumors released more IFN-β, CCL5, and CXCL10 by activation of STING pathway and increased immune cell infiltration, especially of CD8 T cells and natural killer cells, ultimately enhancing the efficacy of anti-PD-1 therapy in multiple subcutaneous and orthotopic tumor mouse models. The response of PTPRT-deficient tumors to anti-PD-1 therapy depends on the tumor-intrinsic STING pathway. In summary, our findings reveal the mechanism of how PTPRT-deficient tumors become sensitive to anti-PD-1 therapy and highlight the biological function of PTPRT in innate immunity. Considering the prevalence of PTPRT mutations and negative expression, this study has great value for patient stratification and clinical decision-making.
Collapse
Affiliation(s)
- Zhuo Chen
- State Key Laboratory of Systems Medicine for Cancer, Renji Hospital, School of Medicine and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200030, China
| | - Wenxiang Ji
- Shanghai Lung Cancer Center, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200030, China
| | - Wenxin Feng
- State Key Laboratory of Systems Medicine for Cancer, Renji Hospital, School of Medicine and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200030, China
| | - Jingchuan Cui
- State Key Laboratory of Systems Medicine for Cancer, Renji Hospital, School of Medicine and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200030, China
| | - Yuchen Wang
- State Key Laboratory of Systems Medicine for Cancer, Renji Hospital, School of Medicine and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200030, China
| | - Fan Li
- State Key Laboratory of Systems Medicine for Cancer, Renji Hospital, School of Medicine and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200030, China
| | - Jiachen Chen
- State Key Laboratory of Systems Medicine for Cancer, Renji Hospital, School of Medicine and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200030, China
| | - Ziheng Guo
- State Key Laboratory of Systems Medicine for Cancer, Renji Hospital, School of Medicine and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200030, China
| | - Liliang Xia
- Shanghai Lung Cancer Center, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200030, China
| | - Xiaokuan Zhu
- Shanghai Lung Cancer Center, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200030, China
| | - Xiaomin Niu
- Shanghai Lung Cancer Center, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200030, China
| | - Yanshuang Zhang
- State Key Laboratory of Systems Medicine for Cancer, Renji Hospital, School of Medicine and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200030, China
| | - Ziming Li
- Shanghai Lung Cancer Center, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200030, China
| | - Alice S T Wong
- School of Biological Sciences, University of Hong Kong, Pokfulam Road, 999077, Hong Kong
| | - Shun Lu
- Shanghai Lung Cancer Center, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200030, China
| | - Weiliang Xia
- State Key Laboratory of Systems Medicine for Cancer, Renji Hospital, School of Medicine and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200030, China
| |
Collapse
|
38
|
Wei Z, Wang Y, Bi Z, Feng L, Sun Y, Zhang H, Song X, Zhang S. Thermoresponsive and Substrate Self-Cycling Nanoenzyme System for Efficient Tumor Therapy. ACS APPLIED BIO MATERIALS 2024; 7:5337-5344. [PMID: 38968606 DOI: 10.1021/acsabm.4c00542] [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] [Indexed: 07/07/2024]
Abstract
Cerium oxide (CeO2-x) performs well in photothermal and catalytic properties due to its abundance of oxygen vacancies. Based on this, we designed a thermosensitive therapeutic nanoplatform to achieve continuous circular drug release in tumor. It can solve the limitation caused by insufficient substrate in the process of tumor treatment. Briefly, CeO2-x and camptothecin (CPT) were wrapped in an agarose hydrogel, which could be melted by the photothermal effect of CeO2-x. At the same time, the local temperature increase provided photothermal treatment, which could induce the apoptosis of tumor cell. After that, CPT was released to damage the DNA in tumor cells to realize chemical treatment. In addition, CPT could active nicotinamide adenine dinucleotide oxidase to react with O2 to increase the intracellular H2O2. After that, the exposed CeO2-x could catalyze H2O2 to generate cytotoxic reactive oxygen species for chemodynamic therapy. More importantly, CeO2-x could catalyze H2O2 to produce O2, which could combine with the catalytic action of CPT to construct a substrate self-cycling nanoenzyme system. Overall, this self-cycling nanoplatform released hypoxia in the tumor microenvironment and built a multimode tumor treatment, which achieved an ideal antitumor affect.
Collapse
Affiliation(s)
- Zizhen Wei
- Collaborative Innovation Center of Tumor Marker Detection Technology, Equipment and Diagnosis-Therapy Integration in Universities of Shandong, Shandong Province Key Laboratory of Detection Technology for Tumor Makers, School of Chemistry and Chemical Engineering, Linyi University, Linyi 276005, China
| | - Yuqi Wang
- Collaborative Innovation Center of Tumor Marker Detection Technology, Equipment and Diagnosis-Therapy Integration in Universities of Shandong, Shandong Province Key Laboratory of Detection Technology for Tumor Makers, School of Chemistry and Chemical Engineering, Linyi University, Linyi 276005, China
| | - Zhiru Bi
- Collaborative Innovation Center of Tumor Marker Detection Technology, Equipment and Diagnosis-Therapy Integration in Universities of Shandong, Shandong Province Key Laboratory of Detection Technology for Tumor Makers, School of Chemistry and Chemical Engineering, Linyi University, Linyi 276005, China
| | - Lu Feng
- Collaborative Innovation Center of Tumor Marker Detection Technology, Equipment and Diagnosis-Therapy Integration in Universities of Shandong, Shandong Province Key Laboratory of Detection Technology for Tumor Makers, School of Chemistry and Chemical Engineering, Linyi University, Linyi 276005, China
| | - Yongbiao Sun
- Collaborative Innovation Center of Tumor Marker Detection Technology, Equipment and Diagnosis-Therapy Integration in Universities of Shandong, Shandong Province Key Laboratory of Detection Technology for Tumor Makers, School of Chemistry and Chemical Engineering, Linyi University, Linyi 276005, China
| | - Huairong Zhang
- Collaborative Innovation Center of Tumor Marker Detection Technology, Equipment and Diagnosis-Therapy Integration in Universities of Shandong, Shandong Province Key Laboratory of Detection Technology for Tumor Makers, School of Chemistry and Chemical Engineering, Linyi University, Linyi 276005, China
| | - Xinyue Song
- Collaborative Innovation Center of Tumor Marker Detection Technology, Equipment and Diagnosis-Therapy Integration in Universities of Shandong, Shandong Province Key Laboratory of Detection Technology for Tumor Makers, School of Chemistry and Chemical Engineering, Linyi University, Linyi 276005, China
| | - Shusheng Zhang
- Collaborative Innovation Center of Tumor Marker Detection Technology, Equipment and Diagnosis-Therapy Integration in Universities of Shandong, Shandong Province Key Laboratory of Detection Technology for Tumor Makers, School of Chemistry and Chemical Engineering, Linyi University, Linyi 276005, China
| |
Collapse
|
39
|
Saha S, Ghosh M, Li J, Wen A, Galluzzi L, Martinez LA, Montrose DC. Serine Depletion Promotes Antitumor Immunity by Activating Mitochondrial DNA-Mediated cGAS-STING Signaling. Cancer Res 2024; 84:2645-2659. [PMID: 38861367 PMCID: PMC11326969 DOI: 10.1158/0008-5472.can-23-1788] [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: 06/15/2023] [Revised: 03/06/2024] [Accepted: 06/04/2024] [Indexed: 06/13/2024]
Abstract
Serine is critical for supporting cancer metabolism, and depriving malignant cells of this nonessential amino acid exerts antineoplastic effects, in large part, through disrupting metabolic pathways. Given the intricate relationship between cancer metabolism and the immune system, the metabolic defects imposed by serine deprivation might impact tumor-targeting immunity. In this study, we demonstrated that restricting endogenous and exogenous sources of serine in colorectal cancer cells results in mitochondrial dysfunction, leading to mitochondrial DNA (mtDNA) accumulation in the cytosol and consequent cGAS-STING1-driven type I IFN secretion. Depleting mtDNA or blocking its release attenuated cGAS-STING1 activation during serine deprivation. In vivo studies revealed that serine deprivation limits tumor growth, accompanied by enhanced type I IFN signaling and intratumoral infiltration of immune effector cells. Notably, the tumor-suppressive and immune-enhancing effects of serine restriction were impaired by T-cell depletion and IFN receptor blockade. Moreover, disrupting cGAS-STING1 signaling in colorectal cancer cells limited the immunostimulatory and tumor-suppressive effects of serine deprivation. Lastly, serine depletion increased the sensitivity of tumors to an immune checkpoint inhibitor targeting PD-1. Taken together, these findings reveal a role for serine as a suppressor of antitumor immunity, suggesting that serine deprivation may be employed to enhance tumor immunogenicity and improve responsiveness to immune checkpoint inhibitors. Significance: Depriving cancer cells of serine provokes mitochondrial perturbations that induce cytosolic mitochondrial DNA accumulation and subsequent activation of cGAS-STING signaling, stimulating tumor-targeting immune responses that can be enhanced with PD-1 targeted therapy. See related commentary by Borges and Garg, p. 2569.
Collapse
Affiliation(s)
- Suchandrima Saha
- Department of Pathology, Renaissance School of Medicine, Stony Brook University, Stony Brook, NY, USA
| | - Monisankar Ghosh
- Department of Pathology, Renaissance School of Medicine, Stony Brook University, Stony Brook, NY, USA
| | - Jinyu Li
- Department of Pathology, Renaissance School of Medicine, Stony Brook University, Stony Brook, NY, USA
| | - Asher Wen
- Department of Pathology, Renaissance School of Medicine, Stony Brook University, Stony Brook, NY, USA
| | - Lorenzo Galluzzi
- Department of Radiation Oncology, Weill Cornell Medical College, New York, NY, USA
- Sandra and Edward Meyer Cancer Center, New York, NY, USA
- Caryl and Israel Englander Institute for Precision Medicine, New York, NY, USA
| | - Luis A. Martinez
- Department of Pathology, Renaissance School of Medicine, Stony Brook University, Stony Brook, NY, USA
- Stony Brook Cancer Center, Stony Brook, NY, USA
| | - David C. Montrose
- Department of Pathology, Renaissance School of Medicine, Stony Brook University, Stony Brook, NY, USA
- Stony Brook Cancer Center, Stony Brook, NY, USA
| |
Collapse
|
40
|
Geng F, Chen J, Song B, Tang Z, Li X, Zhang S, Yang T, Liu Y, Mo W, Zhang Y, Sun C, Tan L, Tu W, Yu D, Cao J, Zhang S. Chaperone- and PTM-mediated activation of IRF1 tames radiation-induced cell death and the inflammatory response. Cell Mol Immunol 2024; 21:856-872. [PMID: 38849539 PMCID: PMC11291999 DOI: 10.1038/s41423-024-01185-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Accepted: 05/13/2024] [Indexed: 06/09/2024] Open
Abstract
The key role of structural cells in immune modulation has been revealed with the advent of single-cell multiomics, but the underlying mechanism remains poorly understood. Here, we revealed that the transcriptional activation of interferon regulatory factor 1 (IRF1) in response to ionizing radiation, cytotoxic chemicals and SARS-CoV-2 viral infection determines the fate of structural cells and regulates communication between structural and immune cells. Radiation-induced leakage of mtDNA initiates the nuclear translocation of IRF1, enabling it to regulate the transcription of inflammation- and cell death-related genes. Novel posttranslational modification (PTM) sites in the nuclear localization sequence (NLS) of IRF1 were identified. Functional analysis revealed that mutation of the acetylation site and the phosphorylation sites in the NLS blocked the transcriptional activation of IRF1 and reduced cell death in response to ionizing radiation. Mechanistically, reciprocal regulation between the single-stranded DNA sensors SSBP1 and IRF1, which restrains radiation-induced and STING/p300-mediated PTMs of IRF1, was revealed. In addition, genetic deletion or pharmacological inhibition of IRF1 tempered radiation-induced inflammatory cell death, and radiation mitigators also suppressed SARS-CoV-2 NSP-10-mediated activation of IRF1. Thus, we revealed a novel cytoplasm-oriented mechanism of IRF1 activation in structural cells that promotes inflammation and highlighted the potential effectiveness of IRF1 inhibitors against immune disorders.
Collapse
Affiliation(s)
- Fenghao Geng
- Laboratory of Radiation Medicine, Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, 610041, China
- Department of Radiation Medicine, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, 610041, China
| | - Jianhui Chen
- Department of Radiation Medicine, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, 610041, China
| | - Bin Song
- Laboratory of Radiation Medicine, Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, 610041, China
| | - Zhicheng Tang
- School of Radiation Medicine and Protection, State Key Laboratory of Radiation Medicine, Soochow University, Suzhou, 215123, China
| | - Xiaoqian Li
- Department of Radiation Medicine, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, 610041, China
| | - Shuaijun Zhang
- Laboratory of Radiation Medicine, Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, 610041, China
| | - Tingyi Yang
- Department of Radiation Medicine, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, 610041, China
| | - Yulan Liu
- The Second Affiliated Hospital of Chengdu Medical College, China National Nuclear Corporation 416 Hospital, Chengdu, 610051, China
| | - Wei Mo
- School of Radiation Medicine and Protection, State Key Laboratory of Radiation Medicine, Soochow University, Suzhou, 215123, China
| | - Yining Zhang
- Department of Radiation Medicine, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, 610041, China
| | - Chuntang Sun
- Laboratory of Radiation Medicine, Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, 610041, China
| | - Lei Tan
- Laboratory of Radiation Medicine, Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, 610041, China
| | - Wenling Tu
- The Second Affiliated Hospital of Chengdu Medical College, China National Nuclear Corporation 416 Hospital, Chengdu, 610051, China
| | - Daojiang Yu
- The Second Affiliated Hospital of Chengdu Medical College, China National Nuclear Corporation 416 Hospital, Chengdu, 610051, China
| | - Jianping Cao
- School of Radiation Medicine and Protection, State Key Laboratory of Radiation Medicine, Soochow University, Suzhou, 215123, China
| | - Shuyu Zhang
- Laboratory of Radiation Medicine, Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, 610041, China.
- Department of Radiation Medicine, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, 610041, China.
- The Second Affiliated Hospital of Chengdu Medical College, China National Nuclear Corporation 416 Hospital, Chengdu, 610051, China.
- NHC Key Laboratory of Nuclear Technology Medical Transformation, Mianyang Central Hospital, Mianyang, 621099, China.
| |
Collapse
|
41
|
Zhou H, Ye P, Xiong W, Duan X, Jing S, He Y, Zeng Z, Wei Y, Ye Q. Genome-scale CRISPR-Cas9 screening in stem cells: theories, applications and challenges. Stem Cell Res Ther 2024; 15:218. [PMID: 39026343 PMCID: PMC11264826 DOI: 10.1186/s13287-024-03831-z] [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: 03/06/2024] [Accepted: 07/02/2024] [Indexed: 07/20/2024] Open
Abstract
Due to the rapid development of stem cell technology, there have been tremendous advances in molecular biological and pathological research, cell therapy as well as organoid technologies over the past decades. Advances in genome editing technology, particularly the discovery of clustered regularly interspaced short palindromic repeats (CRISPR) and CRISPR-related protein 9 (Cas9), have further facilitated the rapid development of stem cell researches. The CRISPR-Cas9 technology now goes beyond creating single gene editing to enable the inhibition or activation of endogenous gene loci by fusing inhibitory (CRISPRi) or activating (CRISPRa) domains with deactivated Cas9 proteins (dCas9). These tools have been utilized in genome-scale CRISPRi/a screen to recognize hereditary modifiers that are synergistic or opposing to malady mutations in an orderly and fair manner, thereby identifying illness mechanisms and discovering novel restorative targets to accelerate medicinal discovery investigation. However, the application of this technique is still relatively rare in stem cell research. There are numerous specialized challenges in applying large-scale useful genomics approaches to differentiated stem cell populations. Here, we present the first comprehensive review on CRISPR-based functional genomics screening in the field of stem cells, as well as practical considerations implemented in a range of scenarios, and exploration of the insights of CRISPR-based screen into cell fates, disease mechanisms and cell treatments in stem cell models. This review will broadly benefit scientists, engineers and medical practitioners in the areas of stem cell research.
Collapse
Affiliation(s)
- Heng Zhou
- Center of Regenerative Medicine and Department of Stomatology, Renmin Hospital of Wuhan University, Wuhan, 430060, People's Republic of China
| | - Peng Ye
- Department of Pharmacy, Renmin Hospital of Wuhan University, Wuhan, 430060, People's Republic of China
| | - Wei Xiong
- Center of Regenerative Medicine and Department of Stomatology, Renmin Hospital of Wuhan University, Wuhan, 430060, People's Republic of China
| | - Xingxiang Duan
- Center of Regenerative Medicine and Department of Stomatology, Renmin Hospital of Wuhan University, Wuhan, 430060, People's Republic of China
| | - Shuili Jing
- Center of Regenerative Medicine and Department of Stomatology, Renmin Hospital of Wuhan University, Wuhan, 430060, People's Republic of China
| | - Yan He
- Institute of Regenerative and Translational Medicine, Tianyou Hospital of Wuhan University of Science and Technology, Wuhan, 430064, Hubei, People's Republic of China
- Department of Oral and Maxillofacial Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, 02114, USA
| | - Zhi Zeng
- Department of Pathology, Renmin Hospital of Wuhan University, Wuhan, 430060, People's Republic of China.
| | - Yen Wei
- The Key Laboratory of Bioorganic Phosphorus Chemistry and Chemical Biology (Ministry of Education), Department of Chemistry, Tsinghua University, Beijing, 100084, People's Republic of China.
| | - Qingsong Ye
- Center of Regenerative Medicine and Department of Stomatology, Renmin Hospital of Wuhan University, Wuhan, 430060, People's Republic of China.
| |
Collapse
|
42
|
Meng X, Wang WD, Li SR, Sun ZJ, Zhang L. Harnessing cerium-based biomaterials for the treatment of bone diseases. Acta Biomater 2024; 183:30-49. [PMID: 38849022 DOI: 10.1016/j.actbio.2024.05.046] [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: 03/11/2024] [Revised: 05/24/2024] [Accepted: 05/30/2024] [Indexed: 06/09/2024]
Abstract
Bone, an actively metabolic organ, undergoes constant remodeling throughout life. Disturbances in the bone microenvironment can be responsible for pathologically bone diseases such as periodontitis, osteoarthritis, rheumatoid arthritis and osteoporosis. Conventional bone tissue biomaterials are not adequately adapted to complex bone microenvironment. Therefore, there is an urgent clinical need to find an effective strategy to improve the status quo. In recent years, nanotechnology has caused a revolution in biomedicine. Cerium(III, IV) oxide, as an important member of metal oxide nanomaterials, has dual redox properties through reversible binding with oxygen atoms, which continuously cycle between Ce(III) and Ce(IV). Due to its special physicochemical properties, cerium(III, IV) oxide has received widespread attention as a versatile nanomaterial, especially in bone diseases. This review describes the characteristics of bone microenvironment. The enzyme-like properties and biosafety of cerium(III, IV) oxide are also emphasized. Meanwhile, we summarizes controllable synthesis of cerium(III, IV) oxide with different nanostructural morphologies. Following resolution of synthetic principles of cerium(III, IV) oxide, a variety of tailored cerium-based biomaterials have been widely developed, including bioactive glasses, scaffolds, nanomembranes, coatings, and nanocomposites. Furthermore, we highlight the latest advances in cerium-based biomaterials for inflammatory and metabolic bone diseases and bone-related tumors. Tailored cerium-based biomaterials have already demonstrated their value in disease prevention, diagnosis (imaging and biosensors) and treatment. Therefore, it is important to assist in bone disease management by clarifying tailored properties of cerium(III, IV) oxide in order to promote the use of cerium-based biomaterials in the future clinical setting. STATEMENT OF SIGNIFICANCE: In this review, we focused on the promising of cerium-based biomaterials for bone diseases. We reviewed the key role of bone microenvironment in bone diseases and the main biological activities of cerium(III, IV) oxide. By setting different synthesis conditions, cerium(III, IV) oxide nanostructures with different morphologies can be controlled. Meanwhile, tailored cerium-based biomaterials can serve as a versatile toolbox (e.g., bioactive glasses, scaffolds, nanofibrous membranes, coatings, and nanocomposites). Then, the latest research advances based on cerium-based biomaterials for the treatment of bone diseases were also highlighted. Most importantly, we analyzed the perspectives and challenges of cerium-based biomaterials. In future perspectives, this insight has given rise to a cascade of cerium-based biomaterial strategies, including disease prevention, diagnosis (imaging and biosensors) and treatment.
Collapse
Affiliation(s)
- Xiang Meng
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Frontier Science Center for Immunology and Metabolism, TaiKang Center for Life and Medical Sciences, Wuhan University, Wuhan 430079, PR China
| | - Wen-Da Wang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Frontier Science Center for Immunology and Metabolism, TaiKang Center for Life and Medical Sciences, Wuhan University, Wuhan 430079, PR China
| | - Su-Ran Li
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Frontier Science Center for Immunology and Metabolism, TaiKang Center for Life and Medical Sciences, Wuhan University, Wuhan 430079, PR China
| | - Zhi-Jun Sun
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Frontier Science Center for Immunology and Metabolism, TaiKang Center for Life and Medical Sciences, Wuhan University, Wuhan 430079, PR China.
| | - Lu Zhang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Frontier Science Center for Immunology and Metabolism, TaiKang Center for Life and Medical Sciences, Wuhan University, Wuhan 430079, PR China; Department of Endodontics, School and Hospital of Stomatology, Wuhan University, HongShan District, LuoYu Road No. 237, Wuhan, 430079, PR China.
| |
Collapse
|
43
|
Zheng Z, Sun J, Wang J, He S, Liu Z, Xie J, Yu CY, Wei H. Enhancing myocardial infarction treatment through bionic hydrogel-mediated spatial combination therapy via mtDNA-STING crosstalk modulation. J Control Release 2024; 371:570-587. [PMID: 38852624 DOI: 10.1016/j.jconrel.2024.06.015] [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: 02/22/2024] [Revised: 05/06/2024] [Accepted: 06/04/2024] [Indexed: 06/11/2024]
Abstract
Myocardial infarction (MI)-induced impaired cardiomyocyte (CM) mitochondrial function and microenvironmental inflammatory cascades severely accelerate the progression of heart failure for compromised myocardial repair. Modulation of the crosstalk between CM mitochondrial DNA (mtDNA) and STING has been recently identified as a robust strategy in enhancing MI treatment, but remains seldom explored. To develop a novel approach that can address persistent myocardial injury using this crosstalk, we report herein construction of a biomimetic hydrogel system, Rb1/PDA-hydrogel comprised of ginsenoside Rb1/polydopamine nanoparticles (Rb1/PDA NPs)-loaded carboxylated chitosan, 4-arm-PEG-phenylboronic acid (4-arm-PEG-PBA), and 4-arm-PEG-dopamine (4-arm-PEG-DA) crosslinked networks. An optimized hydrogel formulation presents not only desired adhesion properties to the surface of the myocardium, but also adaptability for deep myocardial injection, resulting in ROS scavenging, CM mitochondrial function protection, M1 macrophage polarization inhibition through the STING pathway, and angiogenesis promotion via an internal-external spatial combination. The enhanced therapeutic efficiency is supported by the histological analysis of the infarcted area, which shows that the fibrotic area of the MI rats decreases from 58.4% to 5.5%, the thickness of the left ventricular wall increases by 1-fold, and almost complete recovery of cardiac function after 28 days of treatment. Overall, this study reported the first use of a strong adhesive and injectable hydrogel with mtDNA and STING signaling characteristics for enhanced MI treatment via an internal-external spatial combination strategy.
Collapse
Affiliation(s)
- Zhi Zheng
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study & School of Pharmaceutical Science, Hengyang Medical School, University of South China, 28 W Changsheng Road, Hengyang 421001, Hunan, China
| | - Jian Sun
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study & School of Pharmaceutical Science, Hengyang Medical School, University of South China, 28 W Changsheng Road, Hengyang 421001, Hunan, China
| | - Jun Wang
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study & School of Pharmaceutical Science, Hengyang Medical School, University of South China, 28 W Changsheng Road, Hengyang 421001, Hunan, China
| | - Suisui He
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study & School of Pharmaceutical Science, Hengyang Medical School, University of South China, 28 W Changsheng Road, Hengyang 421001, Hunan, China
| | - Zhenqiu Liu
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study & School of Pharmaceutical Science, Hengyang Medical School, University of South China, 28 W Changsheng Road, Hengyang 421001, Hunan, China
| | - Jiahao Xie
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study & School of Pharmaceutical Science, Hengyang Medical School, University of South China, 28 W Changsheng Road, Hengyang 421001, Hunan, China
| | - Cui-Yun Yu
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study & School of Pharmaceutical Science, Hengyang Medical School, University of South China, 28 W Changsheng Road, Hengyang 421001, Hunan, China; Affiliated Hospital of Hunan Academy of Chinese Medicine, Hunan Academy of Chinese Medicine, Changsha 410006, China.
| | - Hua Wei
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study & School of Pharmaceutical Science, Hengyang Medical School, University of South China, 28 W Changsheng Road, Hengyang 421001, Hunan, China.
| |
Collapse
|
44
|
Geng F, Zhong L, Yang T, Chen J, Yang P, Jiang F, Yan T, Song B, Yu Z, Yu D, Zhang J, Cao J, Zhang S. A Frog Skin-Derived Peptide Targeting SCD1 Exerts Radioprotective Effects Against Skin Injury by Inhibiting STING-Mediated Inflammation. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2306253. [PMID: 38582510 PMCID: PMC11220654 DOI: 10.1002/advs.202306253] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Revised: 03/22/2024] [Indexed: 04/08/2024]
Abstract
The extensive application of nuclear technology has increased the potential of uncontrolled radiation exposure to the public. Since skin is the largest organ, radiation-induced skin injury remains a serious medical concern. Organisms evolutionally develop distinct strategies to protect against environment insults and the related research may bring novel insights into therapeutics development. Here, 26 increased peptides are identified in skin tissues of frogs (Pelophylax nigromaculatus) exposed to electron beams, among which four promoted the wound healing of irradiated skin in rats. Specifically, radiation-induced frog skin peptide-2 (RIFSP-2), from histone proteolysis exerted membrane permeability property, maintained cellular homeostasis, and reduced pyroptosis of irradiated cells with decreased TBK1 phosphorylation. Subsequently, stearyl-CoA desaturase 1 (SCD1) is identified, a critical enzyme in biogenesis of monounsaturated fatty acids (MUFAs) as a direct target of RIFSP-2 based on streptavidin-biotin system. The lipidomic analysis further assured the restrain of MUFAs biogenesis by RIFSP-2 following radiation. Moreover, the decreased MUFA limited radiation-induced and STING-mediated inflammation response. In addition, genetic depletion or pharmacological inhibition of STING counteracted the decreased pyroptosis by RIFSP-2 and retarded tissue repair process. Altogether, RIFSP-2 restrains radiation-induced activation of SCD1-MUFA-STING axis. Thus, the stress-induced amphibian peptides can be a bountiful source of novel radiation mitigators.
Collapse
Affiliation(s)
- Fenghao Geng
- Laboratory of Radiation MedicineWest China School of Basic Medical Sciences & Forensic MedicineSichuan UniversityChengdu610041China
- Laboratory of Radiation MedicineWest China Second University HospitalSichuan UniversityChengdu610041China
- Radiation Medicine Department of Institute of Preventive MedicineFourth Military Medical UniversityXi'an710032China
| | - Li Zhong
- School of Radiation Medicine and ProtectionState Key Laboratory of Radiation MedicineSoochow UniversitySuzhou215123China
| | - Tingyi Yang
- Laboratory of Radiation MedicineWest China School of Basic Medical Sciences & Forensic MedicineSichuan UniversityChengdu610041China
| | - Jianhui Chen
- Laboratory of Radiation MedicineWest China School of Basic Medical Sciences & Forensic MedicineSichuan UniversityChengdu610041China
| | - Ping Yang
- Laboratory of Radiation MedicineWest China School of Basic Medical Sciences & Forensic MedicineSichuan UniversityChengdu610041China
| | - Fengdi Jiang
- Laboratory of Radiation MedicineWest China School of Basic Medical Sciences & Forensic MedicineSichuan UniversityChengdu610041China
| | - Tao Yan
- Laboratory of Radiation MedicineWest China School of Basic Medical Sciences & Forensic MedicineSichuan UniversityChengdu610041China
| | - Bin Song
- Laboratory of Radiation MedicineWest China Second University HospitalSichuan UniversityChengdu610041China
| | - Zuxiang Yu
- Laboratory of Radiation MedicineWest China School of Basic Medical Sciences & Forensic MedicineSichuan UniversityChengdu610041China
| | - Daojiang Yu
- The Second Affiliated Hospital of Chengdu Medical CollegeChina National Nuclear Corporation 416 HospitalChengdu610051China
| | - Jie Zhang
- Radiation Medicine Department of Institute of Preventive MedicineFourth Military Medical UniversityXi'an710032China
| | - Jianping Cao
- School of Radiation Medicine and ProtectionState Key Laboratory of Radiation MedicineSoochow UniversitySuzhou215123China
| | - Shuyu Zhang
- Laboratory of Radiation MedicineWest China School of Basic Medical Sciences & Forensic MedicineSichuan UniversityChengdu610041China
- Laboratory of Radiation MedicineWest China Second University HospitalSichuan UniversityChengdu610041China
- The Second Affiliated Hospital of Chengdu Medical CollegeChina National Nuclear Corporation 416 HospitalChengdu610051China
- NHC Key Laboratory of Nuclear Technology Medical Transformation (Mianyang Central Hospital)Mianyang621099China
| |
Collapse
|
45
|
Lanng KRB, Lauridsen EL, Jakobsen MR. The balance of STING signaling orchestrates immunity in cancer. Nat Immunol 2024; 25:1144-1157. [PMID: 38918609 DOI: 10.1038/s41590-024-01872-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2024] [Accepted: 05/14/2024] [Indexed: 06/27/2024]
Abstract
Over the past decade, it has become clear that the stimulator of interferon genes (STING) pathway is critical for a variety of immune responses. This endoplasmic reticulum-anchored adaptor protein has regulatory functions in host immunity across a spectrum of conditions, including infectious diseases, autoimmunity, neurobiology and cancer. In this Review, we outline the central importance of STING in immunological processes driven by expression of type I and III interferons, as well as inflammatory cytokines, and we look at therapeutic options for targeting STING. We also examine evidence that challenges the prevailing notion that STING activation is predominantly beneficial in combating cancer. Further exploration is imperative to discern whether STING activation in the tumor microenvironment confers true benefits or has detrimental effects. Research in this field is at a crossroads, as a clearer understanding of the nuanced functions of STING activation in cancer is required for the development of next-generation therapies.
Collapse
|
46
|
Lamontagne F, Paz-Trejo C, Zamorano Cuervo N, Grandvaux N. Redox signaling in cell fate: Beyond damage. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2024; 1871:119722. [PMID: 38615720 DOI: 10.1016/j.bbamcr.2024.119722] [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: 12/18/2023] [Revised: 02/20/2024] [Accepted: 04/02/2024] [Indexed: 04/16/2024]
Abstract
This review explores the nuanced role of reactive oxygen species (ROS) in cell fate, challenging the traditional view that equates ROS with cellular damage. Through significant technological advancements in detecting localized redox states and identifying oxidized cysteines, a paradigm shift has emerged: from ROS as merely damaging agents to crucial players in redox signaling. We delve into the intricacies of redox mechanisms, which, although confined, exert profound influences on cellular physiological responses. Our analysis extends to both the positive and negative impacts of these mechanisms on cell death processes, including uncontrolled and programmed pathways. By unraveling these complex interactions, we argue against the oversimplified notion of a 'stress response', advocating for a more nuanced understanding of redox signaling. This review underscores the importance of localized redox states in determining cell fate, highlighting the sophistication and subtlety of ROS functions beyond mere damage.
Collapse
Affiliation(s)
- Felix Lamontagne
- CRCHUM - Centre de Recherche du Centre Hospitalier de l'Université de Montréal, 900 rue Saint Denis, Montréal H2X 0A9, Québec, Canada
| | - Cynthia Paz-Trejo
- CRCHUM - Centre de Recherche du Centre Hospitalier de l'Université de Montréal, 900 rue Saint Denis, Montréal H2X 0A9, Québec, Canada; Department of Biochemistry and Molecular Medicine, Faculty of Medicine, Université de Montréal, Montréal H3C 3J7, Québec, Canada
| | - Natalia Zamorano Cuervo
- CRCHUM - Centre de Recherche du Centre Hospitalier de l'Université de Montréal, 900 rue Saint Denis, Montréal H2X 0A9, Québec, Canada
| | - Nathalie Grandvaux
- CRCHUM - Centre de Recherche du Centre Hospitalier de l'Université de Montréal, 900 rue Saint Denis, Montréal H2X 0A9, Québec, Canada; Department of Biochemistry and Molecular Medicine, Faculty of Medicine, Université de Montréal, Montréal H3C 3J7, Québec, Canada.
| |
Collapse
|
47
|
Qian W, Ye J, Xia S. DNA sensing of dendritic cells in cancer immunotherapy. Front Mol Biosci 2024; 11:1391046. [PMID: 38841190 PMCID: PMC11150630 DOI: 10.3389/fmolb.2024.1391046] [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: 02/24/2024] [Accepted: 05/02/2024] [Indexed: 06/07/2024] Open
Abstract
Dendritic cells (DCs) are involved in the initiation and maintenance of immune responses against malignant cells by recognizing conserved pathogen-associated molecular patterns (PAMPs) and damage-associated molecular patterns (DAMPs) through pattern recognition receptors (PRRs). According to recent studies, tumor cell-derived DNA molecules act as DAMPs and are recognized by DNA sensors in DCs. Once identified by sensors in DCs, these DNA molecules trigger multiple signaling cascades to promote various cytokines secretion, including type I IFN, and then to induce DCs mediated antitumor immunity. As one of the potential attractive strategies for cancer therapy, various agonists targeting DNA sensors are extensively explored including the combination with other cancer immunotherapies or the direct usage as major components of cancer vaccines. Moreover, this review highlights different mechanisms through which tumor-derived DNA initiates DCs activation and the mechanisms through which the tumor microenvironment regulates DNA sensing of DCs to promote tumor immune escape. The contributions of chemotherapy, radiotherapy, and checkpoint inhibitors in tumor therapy to the DNA sensing of DCs are also discussed. Finally, recent clinical progress in tumor therapy utilizing agonist-targeted DNA sensors is summarized. Indeed, understanding more about DNA sensing in DCs will help to understand more about tumor immunotherapy and improve the efficacy of DC-targeted treatment in cancer.
Collapse
Affiliation(s)
- Wei Qian
- Department of Immunology, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu, China
| | - Jun Ye
- Department of Immunology, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu, China
- The Center for Translational Medicine, The Affiliated Taizhou People’s Hospital of Nanjing Medical University, Taizhou School of Clinical Medicine, Nanjing Medical University, Taizhou, Jiangsu, China
| | - Sheng Xia
- Department of Immunology, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu, China
| |
Collapse
|
48
|
Wu S, Wang B, Li H, Wang H, Du S, Huang X, Fan Y, Gao Y, Gu L, Huang Q, Chen J, Zhang X, Huang Y, Ma X. Targeting STING elicits GSDMD-dependent pyroptosis and boosts anti-tumor immunity in renal cell carcinoma. Oncogene 2024; 43:1534-1548. [PMID: 38548966 DOI: 10.1038/s41388-024-03013-4] [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: 11/11/2023] [Revised: 03/16/2024] [Accepted: 03/20/2024] [Indexed: 05/15/2024]
Abstract
While Stimulator-of-interferon genes (STING) is an innate immune adapter cruicial for sensing cytosolic DNA and modulating immune microenvironment, its tumor-promoting role in tumor survival and immune evasion remains largely unknown. Here we reported that renal cancer cells are exceptionally dependent on STING for survival and evading immunosurveillance via suppressing ER stress-mediated pyroptosis. We found that STING is significantly amplified and upregulated in clear cell renal cell carcinoma (ccRCC), and its elevated expression is associated with worse clinical outcomes. Mechanically, STING depletion in RCC cells specifically triggers activation of the PERK/eIF2α/ATF4/CHOP pathway and activates cleavage of Caspase-8, thereby inducing GSDMD-mediated pyroptosis, which is independent of the innate immune pathway of STING. Moreover, animal study revealed that STING depletion promoted infiltration of CD4+ and CD8+ T cells, consequently boosting robust antitumor immunity via pyroptosis-induced inflammation. From the perspective of targeted therapy, we found that Compound SP23, a PROTAC STING degrader, demonstrated comparable efficacy to STING depletion both in vitro and in vivo for treatment of ccRCC. These findings collectively unveiled an unforeseen function of STING in regulating GSDMD-dependent pyroptosis, thus regulating immune response in RCC. Consequently, pharmacological degradation of STING by SP23 may become an attractive strategy for treatment of advanced RCC.
Collapse
Affiliation(s)
- Shengpan Wu
- Department of Urology, The Third Medical Center, Chinese PLA General Hospital, 100853, Beijing, China
| | - Baojun Wang
- Department of Urology, The Third Medical Center, Chinese PLA General Hospital, 100853, Beijing, China
| | - Hongzhao Li
- Department of Urology, The Third Medical Center, Chinese PLA General Hospital, 100853, Beijing, China
| | - Hanfeng Wang
- Department of Urology, The Third Medical Center, Chinese PLA General Hospital, 100853, Beijing, China
| | - Songliang Du
- Department of Urology, The Third Medical Center, Chinese PLA General Hospital, 100853, Beijing, China
| | - Xing Huang
- Department of Urology, The Third Medical Center, Chinese PLA General Hospital, 100853, Beijing, China
| | - Yang Fan
- Department of Urology, The Third Medical Center, Chinese PLA General Hospital, 100853, Beijing, China
| | - Yu Gao
- Department of Urology, The Third Medical Center, Chinese PLA General Hospital, 100853, Beijing, China
| | - Liangyou Gu
- Department of Urology, The Third Medical Center, Chinese PLA General Hospital, 100853, Beijing, China
| | - Qingbo Huang
- Department of Urology, The Third Medical Center, Chinese PLA General Hospital, 100853, Beijing, China
| | - Jianjun Chen
- School of Pharmaceutical Sciences, Guangdong Provincial Key Laboratory of New Drug Screening, Southern Medical University, Guangzhou, 510515, China
| | - Xu Zhang
- Department of Urology, The Third Medical Center, Chinese PLA General Hospital, 100853, Beijing, China.
| | - Yan Huang
- Department of Urology, The Third Medical Center, Chinese PLA General Hospital, 100853, Beijing, China.
| | - Xin Ma
- Department of Urology, The Third Medical Center, Chinese PLA General Hospital, 100853, Beijing, China.
| |
Collapse
|
49
|
Lai Y, Yang N, Chen X, Ma X, Chen Z, Dong C, Yu G, Huang Y, Shi D, Fang P, Fu K, Jiang R, Mao C, Ding J, Gao W. Dihydrocapsaicin suppresses the STING-mediated accumulation of ROS and NLRP3 inflammasome and alleviates apoptosis after ischemia-reperfusion injury of perforator skin flap. Phytother Res 2024; 38:2539-2559. [PMID: 38459660 DOI: 10.1002/ptr.8167] [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: 07/13/2023] [Revised: 01/16/2024] [Accepted: 02/08/2024] [Indexed: 03/10/2024]
Abstract
Avascular necrosis frequently occurs as a complication following surgery involving the distal perforator flap. Dihydrocapsaicin (DHC) can protect tissue from ischemia-reperfusion (I/R) injury, but its specific role in multizone perforator flaps remains unclear. In this study, the prospective target of DHC in the context of I/R injury was predicted using network pharmacology analysis. Flap viability was determined through survival area analysis, laser Doppler blood flow, angiograms, and histological examination. The expressions of angiogenesis, apoptosis, NLR family pyrin domain containing 3 (NLRP3) inflammasome, oxidative stress, and molecules related to cyclic guanosine monophosphate (GMP)-adenosine monophosphate synthase (cGAS)-interferon gene stimulant (STING) pathway were assessed using western blotting, immunofluorescence, TUNEL staining, and dihydroethidium (DHE) staining. Our finding revealed that DHC promoted the perforator flap survival, which involves the cGAS-STING pathway, oxidative stress, NLRP3 inflammasome, apoptosis, and angiogenesis. DHC induced oxidative stress resistance and suppressed the NLRP3 inflammasome, preventing apoptosis in vascular endothelial cells. Through regulation of STING pathway, DHC controlled oxidative stress in endothelial cells and NLRP3 levels in ischemic flaps. However, activation of the cGAS-STING pathway led to the accumulation of reactive oxygen species (ROS) and NLRP3 inflammasome, thereby diminishing the protective role of DHC. DHC enhanced the survival of multidomain perforator flaps by suppressing the cGAS-STING pathway, oxidative stress, and the formation of NLRP3 inflammasome. These findings unveil a potentially novel mechanism with clinical significance for promoting the survival of multidomain perforator flaps.
Collapse
Affiliation(s)
- Yingying Lai
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
- Zhejiang Provincial Key Laboratory of Orthopaedics, Wenzhou, China
- The Second Clinical Medical College of Wenzhou Medical University, Wenzhou, China
| | - Ningning Yang
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
- Zhejiang Provincial Key Laboratory of Orthopaedics, Wenzhou, China
- The Second Clinical Medical College of Wenzhou Medical University, Wenzhou, China
| | - Xuankuai Chen
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
- Zhejiang Provincial Key Laboratory of Orthopaedics, Wenzhou, China
- The Second Clinical Medical College of Wenzhou Medical University, Wenzhou, China
| | - Xianhui Ma
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
- Zhejiang Provincial Key Laboratory of Orthopaedics, Wenzhou, China
- The Second Clinical Medical College of Wenzhou Medical University, Wenzhou, China
| | - Zhuliu Chen
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
- Zhejiang Provincial Key Laboratory of Orthopaedics, Wenzhou, China
- The Second Clinical Medical College of Wenzhou Medical University, Wenzhou, China
| | - Chengji Dong
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
- Zhejiang Provincial Key Laboratory of Orthopaedics, Wenzhou, China
- The Second Clinical Medical College of Wenzhou Medical University, Wenzhou, China
| | - Gaoxiang Yu
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
- Zhejiang Provincial Key Laboratory of Orthopaedics, Wenzhou, China
- The Second Clinical Medical College of Wenzhou Medical University, Wenzhou, China
| | - Yingying Huang
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
- Zhejiang Provincial Key Laboratory of Orthopaedics, Wenzhou, China
- The Second Clinical Medical College of Wenzhou Medical University, Wenzhou, China
| | - Donghao Shi
- The Second Clinical Medical College of Wenzhou Medical University, Wenzhou, China
| | - Pin Fang
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
- Zhejiang Provincial Key Laboratory of Orthopaedics, Wenzhou, China
- The Second Clinical Medical College of Wenzhou Medical University, Wenzhou, China
| | - Kejian Fu
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
- Zhejiang Provincial Key Laboratory of Orthopaedics, Wenzhou, China
- The Second Clinical Medical College of Wenzhou Medical University, Wenzhou, China
| | - Renhao Jiang
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
- Zhejiang Provincial Key Laboratory of Orthopaedics, Wenzhou, China
- The Second Clinical Medical College of Wenzhou Medical University, Wenzhou, China
| | - Cong Mao
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
- Zhejiang Provincial Key Laboratory of Orthopaedics, Wenzhou, China
- The Second Clinical Medical College of Wenzhou Medical University, Wenzhou, China
| | - Jian Ding
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
- Zhejiang Provincial Key Laboratory of Orthopaedics, Wenzhou, China
- The Second Clinical Medical College of Wenzhou Medical University, Wenzhou, China
| | - Weiyang Gao
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
- Zhejiang Provincial Key Laboratory of Orthopaedics, Wenzhou, China
- The Second Clinical Medical College of Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
50
|
Fu C, Tong W, Yu L, Miao Y, Wei Q, Yu Z, Chen B, Wei M. When will the immune-stimulating antibody conjugates (ISACs) be transferred from bench to bedside? Pharmacol Res 2024; 203:107160. [PMID: 38547937 DOI: 10.1016/j.phrs.2024.107160] [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: 01/21/2024] [Revised: 03/17/2024] [Accepted: 03/25/2024] [Indexed: 04/30/2024]
Abstract
Immunostimulatory antibody conjugates (ISACs) as a promising new generation of targeted therapeutic antibody-drug conjugates (ADCs), that not only activate innate immunity but also stimulate adaptive immunity, providing a dual therapeutic effect to eliminate tumor cells. However, several ISACs are still in the early stages of clinical development or have already failed. Therefore, it is crucial to design ISACs more effectively to overcome their limitations, including high toxicity, strong immunogenicity, long development time, and poor pharmacokinetics. This review aims to summarize the composition and function of ISACs, incorporating current design considerations and ongoing clinical trials. Additionally, the review delves into the current issues with ISACs and potential solutions, such as adjusting the drug-antibody ratio (DAR) to improve the bioavailability of ISACs. By leveraging the affinity and bioavailability-enhancing properties of bispecific antibodies, the utility between antibodies and immunostimulatory agents can be balanced. Commonly used immunostimulatory agents may induce systemic immune reactions, and BTK (Bruton's tyrosine kinase) inhibitors can regulate immunogenicity. Finally, the concept of grafting ADC's therapeutic principles is simple, but the combination of payload, linker, and targeted functional molecules is not a simple permutation and combination problem. The development of conjugate drugs faces more complex pharmacological and toxicological issues. Standing on the shoulders of ADC, the development and application scenarios of ISAC are endowed with broader space.
Collapse
Affiliation(s)
- Chen Fu
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang 110122, PR China; Liaoning Key Laboratory of Molecular Targeted Anti-tumor Drug Development and Evaluation, China Medical University, Shenyang 110122, PR China
| | - Weiwei Tong
- Department of Laboratory Medicine, Shengjing Hospital of China Medical University, Shenyang 110122, PR China
| | - Lifeng Yu
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang 110122, PR China; Liaoning Key Laboratory of Molecular Targeted Anti-tumor Drug Development and Evaluation, China Medical University, Shenyang 110122, PR China
| | - Yuxi Miao
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang 110122, PR China; Liaoning Key Laboratory of Molecular Targeted Anti-tumor Drug Development and Evaluation, China Medical University, Shenyang 110122, PR China
| | - Qian Wei
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang 110122, PR China; Liaoning Key Laboratory of Molecular Targeted Anti-tumor Drug Development and Evaluation, China Medical University, Shenyang 110122, PR China
| | - Zhaojin Yu
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang 110122, PR China; Liaoning Key Laboratory of Molecular Targeted Anti-tumor Drug Development and Evaluation, China Medical University, Shenyang 110122, PR China.
| | - Bo Chen
- Department of Breast Surgery, The First Hospital of China Medical University, No. 155, Nanjing North Street, Shenyang 110122, PR China.
| | - Minjie Wei
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang 110122, PR China; Liaoning Key Laboratory of Molecular Targeted Anti-tumor Drug Development and Evaluation, China Medical University, Shenyang 110122, PR China.
| |
Collapse
|