1
|
Li C, Ji H, Zhuang S, Xie X, Cui D, Zhang C. Update on the correlation between mitochondrial function and osteonecrosis of the femoral head osteocytes. Redox Rep 2025; 30:2491846. [PMID: 40249372 PMCID: PMC12010656 DOI: 10.1080/13510002.2025.2491846] [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: 04/19/2025] Open
Abstract
Mitochondrial health is maintained in a steady state through mitochondrial dynamics and autophagy processes. Recent studies have identified healthy mitochondria as crucial regulators of cellular function and survival. This process involves adenosine triphosphate (ATP) synthesis by mitochondrial oxidative phosphorylation (OXPHOS), regulation of calcium metabolism and inflammatory responses, and intracellular oxidative stress management. In the skeletal system, they participate in the regulation of cellular behaviors and the responses of osteoblasts, osteoclasts, chondrocytes, and osteocytes to external stimuli. Indeed, mitochondrial damage or dysfunction occurs in the development of a few bone diseases. For example, mitochondrial damage may lead to an imbalance in osteoblasts and osteoclasts, resulting in osteoporosis, osteomalacia, or poor bone production, and chondrocyte death and inflammatory infiltration in osteoarthritis are the main causes of cartilage degeneration due to mitochondrial damage. However, the opposite exists for osteosarcoma, where overactive mitochondrial metabolism is able to accelerate the proliferation and migration of osteosarcoma cells, which is a major disease feature. Bone is a dynamic organ and osteocytes play a fundamental role in all regions of bone tissue and are involved in regulating bone integrity. This review examines the impact of mitochondrial physiological function on osteocyte health and summarizes the microscopic molecular mechanisms underlying its effects. It highlights that targeted therapies focusing on osteocyte mitochondria may be beneficial for osteocyte survival, providing a new insight for the diagnosis, prevention, and treatment of diseases associated with osteocyte death.
Collapse
Affiliation(s)
- Chengming Li
- Department of Orthopedics, Zhongda Hospital Southeast University, Nanjing, People’s Republic of China
| | - Hangyu Ji
- Department of Orthopedics, Zhongda Hospital Southeast University, Nanjing, People’s Republic of China
| | - Suyang Zhuang
- Department of Orthopedics, Zhongda Hospital Southeast University, Nanjing, People’s Republic of China
| | - Xinhui Xie
- Department of Orthopedics, Zhongda Hospital Southeast University, Nanjing, People’s Republic of China
| | - Daping Cui
- Department of Orthopedics, Shenzhen Bao’an District Central Hospital, Shenzhen, People’s Republic of China
| | - Cong Zhang
- Department of Orthopedics, Zhongda Hospital Southeast University, Nanjing, People’s Republic of China
| |
Collapse
|
2
|
Xu X, Fei X, Wang H, Wu X, Zhan Y, Li X, Zhou Y, Shu C, He C, Hu Y, Liu J, Lv N, Li N, Zhu Y. Helicobacter pylori infection induces DNA double-strand breaks through the ACVR1/IRF3/POLD1 signaling axis to drive gastric tumorigenesis. Gut Microbes 2025; 17:2463581. [PMID: 39924917 PMCID: PMC11812335 DOI: 10.1080/19490976.2025.2463581] [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: 08/15/2024] [Revised: 01/06/2025] [Accepted: 02/02/2025] [Indexed: 02/11/2025] Open
Abstract
Helicobacter pylori (H. pylori) infection plays a pivotal role in gastric carcinogenesis through inflammation-related mechanisms. Activin A receptor type I (ACVR1), known for encoding the type I receptor for bone morphogenetic proteins (BMPs), has been identified as a cancer diver gene across various tumors. However, the specific role of AVCR1 in H. pylori-induced gastric tumorigenesis remains incompletely understood. We conducted a comprehensive analysis of the clinical relevance of ACVR1 by integrating data from public databases and our local collection of human gastric tissues. In vitro cell cultures, patient-derived gastric organoids, and transgenic INS-GAS mouse models were used for Western blot, qRT-PCR, immunofluorescence, immunohistochemistry, luciferase assays, ChIP, and comet assays. Furthermore, to investigate the therapeutic potential, we utilized the ACVR1 inhibitor DM3189 in our in vivo studies. H. pylori infection led to increased expression of ACVR1 in gastric epithelial cells, gastric organoid and gastric mucosa of INS-GAS mice. ACVR1 activation led to DNA double-strand break (DSB) accumulation by inhibiting POLD1, a crucial DNA repair enzyme. The activation of POLD1 was facilitated by the transcription factor IRF3, with identified binding sites. Additionally, treatment with the ACVR1 inhibitor DM3189 significantly ameliorated H. pylori-induced gastric pathology and reduced DNA damage in INS-GAS mice. Immunohistochemistry analysis showed elevated levels of ACVR1 in H. pylori-positive gastritis tissues, showing a negative correlation with POLD1 expression. This study uncovers a novel signaling axis of AVCR1/IRF3/POLD1 in the pathogenesis of H. pylori infection. The upregulation of ACVR1 and the suppression of POLD1 upon H. pylori infection establish a connection between the infection, genomic instability, and the development of gastric carcinogenesis.
Collapse
Affiliation(s)
- Xinbo Xu
- Department of Gastroenterology, Jiangxi Provincial Key Laboratory of Digestive Diseases, Jiangxi Clinical Research Center for Gastroenterology, Digestive Disease Hospital, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Xiao Fei
- Department of Gastroenterology, Jiangxi Provincial Key Laboratory of Digestive Diseases, Jiangxi Clinical Research Center for Gastroenterology, Digestive Disease Hospital, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Huan Wang
- Department of Gastroenterology, Jiangxi Provincial Key Laboratory of Digestive Diseases, Jiangxi Clinical Research Center for Gastroenterology, Digestive Disease Hospital, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Xidong Wu
- Department of Drug Safety Evaluation, Jiangxi Testing Center of Medical Instruments, Nanchang, China
| | - Yuan Zhan
- Department of Pathology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Xin Li
- Department of Gastroenterology, Jiangxi Provincial Key Laboratory of Digestive Diseases, Jiangxi Clinical Research Center for Gastroenterology, Digestive Disease Hospital, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Yan’an Zhou
- Department of Gastroenterology, Jiangxi Provincial Key Laboratory of Digestive Diseases, Jiangxi Clinical Research Center for Gastroenterology, Digestive Disease Hospital, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Chunxi Shu
- Department of Gastroenterology, Jiangxi Provincial Key Laboratory of Digestive Diseases, Jiangxi Clinical Research Center for Gastroenterology, Digestive Disease Hospital, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Cong He
- Department of Gastroenterology, Jiangxi Provincial Key Laboratory of Digestive Diseases, Jiangxi Clinical Research Center for Gastroenterology, Digestive Disease Hospital, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Yi Hu
- Department of Gastroenterology, Jiangxi Provincial Key Laboratory of Digestive Diseases, Jiangxi Clinical Research Center for Gastroenterology, Digestive Disease Hospital, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Jianping Liu
- Department of Gastroenterology, Jiangxi Provincial Key Laboratory of Digestive Diseases, Jiangxi Clinical Research Center for Gastroenterology, Digestive Disease Hospital, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Nonghua Lv
- Department of Gastroenterology, Jiangxi Provincial Key Laboratory of Digestive Diseases, Jiangxi Clinical Research Center for Gastroenterology, Digestive Disease Hospital, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Nianshuang Li
- Department of Gastroenterology, Jiangxi Provincial Key Laboratory of Digestive Diseases, Jiangxi Clinical Research Center for Gastroenterology, Digestive Disease Hospital, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Yin Zhu
- Department of Gastroenterology, Jiangxi Provincial Key Laboratory of Digestive Diseases, Jiangxi Clinical Research Center for Gastroenterology, Digestive Disease Hospital, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| |
Collapse
|
3
|
Yang H, Xia Y, Ma Y, Gao M, Hou S, Xu S, Wang Y. Inhibition of the cGAS-STING pathway: contributing to the treatment of cerebral ischemia-reperfusion injury. Neural Regen Res 2025; 20:1900-1918. [PMID: 38993125 PMCID: PMC11691458 DOI: 10.4103/nrr.nrr-d-24-00015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2024] [Revised: 03/05/2024] [Accepted: 05/02/2024] [Indexed: 07/13/2024] Open
Abstract
The cGAS-STING pathway plays an important role in ischemia-reperfusion injury in the heart, liver, brain, and kidney, but its role and mechanisms in cerebral ischemia-reperfusion injury have not been systematically reviewed. Here, we outline the components of the cGAS-STING pathway and then analyze its role in autophagy, ferroptosis, cellular pyroptosis, disequilibrium of calcium homeostasis, inflammatory responses, disruption of the blood-brain barrier, microglia transformation, and complement system activation following cerebral ischemia-reperfusion injury. We further analyze the value of cGAS-STING pathway inhibitors in the treatment of cerebral ischemia-reperfusion injury and conclude that the pathway can regulate cerebral ischemia-reperfusion injury through multiple mechanisms. Inhibition of the cGAS-STING pathway may be helpful in the treatment of cerebral ischemia-reperfusion injury.
Collapse
Affiliation(s)
- Hang Yang
- School of Clinical Medicine, Shandong Second Medical University, Weifang, Shandong Province, China
| | - Yulei Xia
- School of Clinical Medicine, Shandong Second Medical University, Weifang, Shandong Province, China
| | - Yue Ma
- School of Clinical Medicine, Shandong Second Medical University, Weifang, Shandong Province, China
| | - Mingtong Gao
- Department of Emergency, The Affiliated Hospital of Weifang Medical University, Weifang, Shandong Province, China
| | - Shuai Hou
- School of Clinical Medicine, Shandong Second Medical University, Weifang, Shandong Province, China
| | - Shanshan Xu
- Department of Emergency, The Affiliated Hospital of Weifang Medical University, Weifang, Shandong Province, China
| | - Yanqiang Wang
- Department of Neurology II, The Affiliated Hospital of Weifang Medical University, Weifang, Shandong Province, China
| |
Collapse
|
4
|
Feng D, Kang X, Wang H, He Z, Xu H, Li Y, Fan A, Xu H, Zhang Y, Song J, Hou J, Qi J, Zhang W. Photochemical bomb: Precision nuclear targeting to activate cGAS-STING pathway for enhanced bladder cancer immunotherapy. Biomaterials 2025; 318:123126. [PMID: 39884129 DOI: 10.1016/j.biomaterials.2025.123126] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Revised: 12/31/2024] [Accepted: 01/23/2025] [Indexed: 02/01/2025]
Abstract
Activating the cGAS-STING pathway presents a promising strategy to enhance the innate immunity and combat the immunosuppressive tumor microenvironment. One key mechanism for triggering this pathway involves the release of damaged DNA fragments caused by nuclear DNA damage. However, conventional cGAS-STING agonists often suffer from limited nucleus-targeting efficiency and potential biotoxicity. In this study, we develop a novel nucleus-targeting theranostic nanoplatform designed to synergistically activate the cGAS-STING pathway through the combination of photodynamic therapy (PDT) and cisplatin chemotherapy for orthotopic bladder cancer treatment. The nanoplatform integrates a new high-performance type-I photosensitizer with near-infrared-II emission, a TATSA peptide for enhanced nuclear targeting, and a biosafe platinum (IV) cisplatin prodrug. Upon NIR laser irradiation, the nanoagent delivers synergistic nucleus-targeted PDT and chemotherapy, causing substantial DNA damage and the release of double-stranded DNA, which subsequently activates the cGAS-STING pathway and triggers potent immunomodulation. This activation promotes dendritic cells maturation, enhances cytotoxic T infiltration, and facilitates the formation of memory T cells, leading to immune microenvironment remodeling, and long-lasting immune memory, thus effectively inhibiting orthotopic bladder tumors and reducing the risk of metastasis. These findings highlight the substantial potential of this strategy to overcome the limitations of current immunotherapies by leveraging nucleus-targeted PDT to activate the cGAS-STING pathway for cancer treatment.
Collapse
Affiliation(s)
- Dexiang Feng
- Department of Urology, The Fourth Affiliated Hospital of Soochow University, Suzhou Dushu Lake Hospital, Medical Center of Soochow University, Suzhou, 215000, China; Department of Urology, First Affiliated Hospital of Soochow University, Suzhou, 215006, China
| | - Xiaoying Kang
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials, Ministry of Education, Frontiers Science Center for Cell Responses, and College of Life Sciences, Nankai University, Tianjin, 300071, China
| | - He Wang
- Department of Urology, First Affiliated Hospital of Soochow University, Suzhou, 215006, China.
| | - Zhangxin He
- Department of Urology, The Fourth Affiliated Hospital of Soochow University, Suzhou Dushu Lake Hospital, Medical Center of Soochow University, Suzhou, 215000, China; Department of Urology, First Affiliated Hospital of Soochow University, Suzhou, 215006, China
| | - Haodong Xu
- Department of Urology, First Affiliated Hospital of Soochow University, Suzhou, 215006, China
| | - Yue Li
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials, Ministry of Education, Frontiers Science Center for Cell Responses, and College of Life Sciences, Nankai University, Tianjin, 300071, China
| | - Aohua Fan
- School of Chemistry and Life Science, Changchun University of Technology, Changchun, 130012, China
| | - Hongbo Xu
- Department of Urology, The Fourth Affiliated Hospital of Soochow University, Suzhou Dushu Lake Hospital, Medical Center of Soochow University, Suzhou, 215000, China
| | - Yuan Zhang
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials, Ministry of Education, Frontiers Science Center for Cell Responses, and College of Life Sciences, Nankai University, Tianjin, 300071, China
| | - Jianwen Song
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials, Ministry of Education, Frontiers Science Center for Cell Responses, and College of Life Sciences, Nankai University, Tianjin, 300071, China
| | - Jianquan Hou
- Department of Urology, The Fourth Affiliated Hospital of Soochow University, Suzhou Dushu Lake Hospital, Medical Center of Soochow University, Suzhou, 215000, China; Department of Urology, First Affiliated Hospital of Soochow University, Suzhou, 215006, China.
| | - Ji Qi
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials, Ministry of Education, Frontiers Science Center for Cell Responses, and College of Life Sciences, Nankai University, Tianjin, 300071, China.
| | - Weijie Zhang
- Department of Urology, The Fourth Affiliated Hospital of Soochow University, Suzhou Dushu Lake Hospital, Medical Center of Soochow University, Suzhou, 215000, China; Department of Urology, First Affiliated Hospital of Soochow University, Suzhou, 215006, China.
| |
Collapse
|
5
|
Fernandez-Alarcon J, Cladera MA, Rodriguez-Camenforte N, Sitia G, Guerra-Rebollo M, Borros S, Fornaguera C. Regulation of mitochondrial apoptosis via siRNA-loaded metallo-alginate hydrogels: A localized and synergistic antitumor therapy. Biomaterials 2025; 318:123164. [PMID: 39923537 DOI: 10.1016/j.biomaterials.2025.123164] [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: 09/19/2024] [Revised: 01/13/2025] [Accepted: 02/01/2025] [Indexed: 02/11/2025]
Abstract
Preventing relapse after resection of a primary tumor continues to be an unmet clinical need. Development of adjuvant biomaterials with the capacity to kill residual cancer cells after tumor resection is of clinical importance. Here we developed a library of metallo-alginate hydrogels containing high concentrations of metallic ions such as Ca2+ in combination with Zn2+, Li+, or Mg2+ to disrupt Ca2+ homeostasis in the mitochondria of cancer cells by local hyperthermia. To synergistically kill tumor cells and suppress the growth of rechallenged tumors, we embedded oncogene-silencing nucleic acids (mTOR siRNA) loaded into polymerc nanoparticles (NPs) composed of poly (β-amino esters) in the metallo-alginate hydrogels, targeting cancer cells that activate multi-drug resistance pathways such PI3K/AKT/mTOR. Metabolomic studies showed alterations in the Warburg effect, mitochondrial transport, and the TCA cycle, confirming cancer cell damage. In vivo studies of this targeted therapy in mice demonstrated a sex-dependent effect. Male B16F10-tumor-bearing mice treated with the synergistic therapy showed restrained tumor growth. In contrast, no therapeutic effect was observed in female counterparts. Our results demonstrate that in situ-formed NP-loaded metallo-alginate hydrogels can modulate two distinct immune signaling networks that are relevant for enhancing cancer cell death. On the basis of our findings, this combination therapy emerges as a promising sex-dependent strategy for clinical translation.
Collapse
Affiliation(s)
- Jennifer Fernandez-Alarcon
- Grup d'Enginyeria de Materials (GEMAT), Institut Químic de Sarrià (IQS), Universitat Ramon Llull (URL), 08017, Barcelona, Spain
| | - Margalida Artigues Cladera
- Grup d'Electroquímica i Bioanàlisi (EQBA), Institut Químic de Sarrià (IQS), Universitat Ramon Llull (URL), 08017, Barcelona, Spain
| | - Natalia Rodriguez-Camenforte
- Grup d'Enginyeria de Materials (GEMAT), Institut Químic de Sarrià (IQS), Universitat Ramon Llull (URL), 08017, Barcelona, Spain
| | - Giovanni Sitia
- Experimental Hepatology Unit, Division of Immunology, Transplantation and Infectious Diseases, IRCCS San Raffaele Scientific Institute, 20132, Milan, Italy
| | - Marta Guerra-Rebollo
- Grup d'Enginyeria de Materials (GEMAT), Institut Químic de Sarrià (IQS), Universitat Ramon Llull (URL), 08017, Barcelona, Spain
| | - Salvador Borros
- Grup d'Enginyeria de Materials (GEMAT), Institut Químic de Sarrià (IQS), Universitat Ramon Llull (URL), 08017, Barcelona, Spain
| | - Cristina Fornaguera
- Grup d'Enginyeria de Materials (GEMAT), Institut Químic de Sarrià (IQS), Universitat Ramon Llull (URL), 08017, Barcelona, Spain; Institució Catalana de Recerca i Estudis Avançats (ICREA), Passeig de Lluís Companys 23, 08010, Barcelona, Spain.
| |
Collapse
|
6
|
Cui P, Song B, Xia Z, Xu Y. Type I Interferon Signalling and Ischemic Stroke: Mechanisms and Therapeutic Potentials. Transl Stroke Res 2025; 16:962-974. [PMID: 38466560 DOI: 10.1007/s12975-024-01236-x] [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: 11/30/2023] [Revised: 01/27/2024] [Accepted: 02/06/2024] [Indexed: 03/13/2024]
Abstract
Type I interferon (IFN-I) signalling is intricately involved in the pathogenesis of multiple infectious diseases, autoimmune diseases, and neurological diseases. Acute ischemic stroke provokes overactivation of IFN-I signalling within the injured brain, particularly in microglia. Following cerebral ischemia, damage-associated molecular patterns (DAMPs) released from injured neural cells elicit marked proinflammatory episodes within minutes. Among these, self-nucleic acids, including nuclear DNA and mitochondrial DNA (mtDNA), have been recognized as a critical alarm signal to fan the flames of neuroinflammation, predominantly via inducing IFN-I signalling activation in microglia. The concept of interferon-responsive microglia (IRM), marked by upregulation of a plethora of IFN-stimulated genes, has been emergingly elucidated in ischemic mouse brains, particularly in aged ones. Among the pattern recognition receptors responsible for IFN-I induction, cyclic GMP-AMP synthase (cGAS)-stimulator of interferon genes (STING) plays integral roles in potentiating microglia-driven neuroinflammation and secondary brain injury after cerebral ischemia. Here, we aim to provide an up-to-date review on the multifaceted roles of IFN-I signalling, the detailed molecular and cellular mechanisms leading to and resulting from aberrant IFN-I signalling activation after cerebral ischemia, and the therapeutic potentials. A thorough exploration of these above points will inform our quest for IFN-based therapies as effective immunomodulatory therapeutics to complement the limited repertoire of thrombolytic agents, thereby facilitating the translation from bench to bedside.
Collapse
Affiliation(s)
- Pan Cui
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, Henan, China
- NHC Key Laboratory of Prevention and Treatment of Cerebrovascular Diseases, Zhengzhou, Henan, China
- Clinical Systems Biology Laboratories, Translation Medicine Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, Henan, China
| | - Bo Song
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, Henan, China
- NHC Key Laboratory of Prevention and Treatment of Cerebrovascular Diseases, Zhengzhou, Henan, China
| | - Zongping Xia
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, Henan, China.
- NHC Key Laboratory of Prevention and Treatment of Cerebrovascular Diseases, Zhengzhou, Henan, China.
- Clinical Systems Biology Laboratories, Translation Medicine Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, Henan, China.
| | - Yuming Xu
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, Henan, China.
- NHC Key Laboratory of Prevention and Treatment of Cerebrovascular Diseases, Zhengzhou, Henan, China.
- Henan Key Laboratory of Cerebrovascular Diseases, Zhengzhou University, Zhengzhou, Henan, China.
| |
Collapse
|
7
|
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
|
8
|
Shen X, Feng T, Li S, Wang X, Zhang W, Wang S, Zhang X, Yang J, Liu Y. Leucine enhances the cGAS-STING-NLRP3 pathway in autoimmune thyroiditis. J Transl Autoimmun 2025; 10:100284. [PMID: 40226573 PMCID: PMC11986970 DOI: 10.1016/j.jtauto.2025.100284] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2025] [Revised: 03/12/2025] [Accepted: 03/15/2025] [Indexed: 04/15/2025] Open
Abstract
Background Branched-chain amino acids (BCAAs), including isoleucine (Ile), leucine (Leu), and valine (Val), are substrates for synthesising nitrogenous compounds and signalling molecules involved in regulating immunity. To date, data on the role of BCAAs in autoimmune thyroiditis (AIT) are lacking; therefore, this study aimed to determine the causality using two-sample Mendelian randomisation (MR) and explored the role of BCAAs in the cGAS-STING-NLRP3 pathway in vitro. Methods The causal relationship between BCAAs and the pathogenesis of AIT were identified using a two-sample MR study. The anti-inflammatory effects of BCAAs and their role in the cGAS-STING-NLRP3 pathway were investigated in lipopolysaccharide (LPS)- induced thyroid follicular cells (TFCs). Results Our findings indicate that BCAAs are a pathogenic factor for AIT (IVW OR = 4.960; 95 % CI = (1.54,15.940); P = 0.007). Leu significantly exacerbated the inflammatory response of thyroid cells, as evidenced by the up-regulation of tumour necrosis factor-alpha (TNF-α) and interleukin (IL)-6 and down-regulation of TGF-β1; simultaneously aggravated cellular injury and oxidative stress; significantly increased the expression of Sestrin2/p-mTOR and cGAS/STING/NLRP3 in AIT cells. Furthermore, the expression of IL-18 and IL-1β was significantly increased. Conversely, Leu deprivation induced cell injury, decreased oxidative stress, and inhibited Sestrin2/p-mTOR and cGAS/STING/NLRP3 pathways. Conclusion Our findings suggest a potential causal effect of genetically predicted Leu on AIT; Leu significantly exacerbated the inflammatory response and cellular damage in AIT cells. The mechanism by which Leu induces inflammation involves activating the promoted Sestrin2/mTOR and cGAS-STING-NLRP3 signalling pathways. Our study proposes a novel mechanism for the contributions of Leu in AIT and potential therapeutic strategies involving Leu deprivation in treating AIT.
Collapse
Affiliation(s)
- Xin Shen
- Department of General Practice, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong Province, 250021, China
- School of Acupuncture-Moxibustion and Tuina, Shandong University of Traditional Chinese Medicine, Jinan, Shandong Province, 250355, China
| | - Tingting Feng
- Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong Province, 250021, China
| | - Shangbin Li
- Department of Health Office, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong Province, 250021, China
| | - Xingxin Wang
- School of Acupuncture-Moxibustion and Tuina, Shandong University of Traditional Chinese Medicine, Jinan, Shandong Province, 250355, China
| | - Wenhui Zhang
- School of Acupuncture-Moxibustion and Tuina, Shandong University of Traditional Chinese Medicine, Jinan, Shandong Province, 250355, China
| | - Shouyan Wang
- Heze Municipal Hospital, Heze, Shandong Province, 274000, China
| | - Xiaohan Zhang
- Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong Province, 250021, China
| | - Jiguo Yang
- School of Acupuncture-Moxibustion and Tuina, Shandong University of Traditional Chinese Medicine, Jinan, Shandong Province, 250355, China
| | - Yuanxiang Liu
- The First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, Shandong Province, 250355, China
| |
Collapse
|
9
|
Chang CK, Wu ZS, Niu GH, Chou YY, Tang SH, Zhang MM, Sung CS, Tung HT, Tsou LK, Tang CC, Sung PJ, Lo YH, Wen ZH. Marine-derived STING inhibitors, excavatolide B promote wound repair in full-thickness-incision rats. Int Immunopharmacol 2025; 155:114593. [PMID: 40209311 DOI: 10.1016/j.intimp.2025.114593] [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: 02/07/2025] [Revised: 03/26/2025] [Accepted: 03/28/2025] [Indexed: 04/12/2025]
Abstract
The process of wound healing encompasses both inflammatory and proliferative stages. Excessive inflammation is known to impede the healing of chronic wounds. Activation of the cyclic guanosine monophosphate-adenosine monophosphate synthase (cGAS)-stimulator of interferon genes (STING) signaling pathway causes inflammation during cellular stress and tissue damage. Herein, we examined the anti-inflammatory effects of the marine-derived STING antagonist excavatolide B (EXCB) and its derivatives, EXCB-61 and EXCB-79, in full-thickness-incision rats. Wound area measurements, histopathological observations, and immunohistochemical analyses were performed to evaluate the roles of these compounds in wound healing. These three compounds were found to have low toxicity, with EXCB promoting Hs68 human dermal fibroblast migration and proliferation. EXCB and EXCB61 treatments, but not EXCB79, reduced the wound area. The histopathological results showed a significant decrease in immune cell infiltration and mast cell accumulation in all compound-treated groups. Immunohistochemical analysis revealed that EXCB and its derivatives reduced cGAS-STING pathway factors such as STING, phosphorylated TANK-binding kinase 1, nuclear factor kappa-light-chain-enhancer of activated B cells, and M1 macrophages while increasing the expression of angiogenic factors vascular endothelial growth factor and CD31, as well as M2 macrophages and collagen I/III deposition. We conclude that marine-derived STING antagonists can attenuate inflammatory responses by inhibiting the cGAS-STING pathway and promoting angiogenesis, thereby aiding wound healing.
Collapse
Affiliation(s)
- Chun-Kai Chang
- Department of Marine Biotechnology and Resources, National Sun Yat-sen University, Kaohsiung 80424, Taiwan; Department of Plastic & Reconstructive Surgery, Zouying Armed Forces General Hospital, No. 553, Junxiao Rd., Zuoying Dist., Kaohsiung City 81342, Taiwan
| | - Zong-Sheng Wu
- Department of Anesthesiology, Taipei Veterans General Hospital, Taipei 112201, Taiwan
| | - Guang-Hao Niu
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Miaoli, Zhunan 35053, Taiwan
| | - Yu-Yu Chou
- Department of Plastic & Reconstructive Surgery, Zouying Armed Forces General Hospital, No. 553, Junxiao Rd., Zuoying Dist., Kaohsiung City 81342, Taiwan
| | - Shih-Hsuan Tang
- Department of Anesthesiology, Taipei Veterans General Hospital, Taipei 112201, Taiwan
| | - Mingzi M Zhang
- Institute of Molecular and Genomic Medicine, National Health Research Institutes, Miaoli, Zhunan 35053, Taiwan
| | - Chun-Sung Sung
- Department of Anesthesiology, Taipei Veterans General Hospital, Taipei 112201, Taiwan; School of Medicine, National Yang-Ming Chiao Tung University, Taipei 112304, Taiwan
| | - Hsiang-Ting Tung
- Department of Marine Biotechnology and Resources, National Sun Yat-sen University, Kaohsiung 80424, Taiwan
| | - Lun Kelvin Tsou
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Miaoli, Zhunan 35053, Taiwan
| | - Chi-Chieh Tang
- Department of Early Childhood Education, National Pingtung University, 91201, Taiwan
| | - Ping-Jyun Sung
- National Museum of Marine Biology and Aquarium, Pingtung 94450, Taiwan
| | - Yi-Hao Lo
- Department of Family Medicine, Zuoying Armed Forces General Hospital, Kaohsiung 81342, Taiwan; Department of Nursing, Meiho University, Pingtung County, 91200, Taiwan.
| | - Zhi-Hong Wen
- Department of Marine Biotechnology and Resources, National Sun Yat-sen University, Kaohsiung 80424, Taiwan; National Museum of Marine Biology and Aquarium, Pingtung 94450, Taiwan; Institute of Biotechnology and Pharmaceutical Research, National Sun Yat-sen University, Kaohsiung 80424, Taiwan.
| |
Collapse
|
10
|
Meng F, He Y, Zhao J, Yuan Z, Wang J, Parra KG, Fishel ML, Ratliff TL, Yeo Y. Timely administration of drug combination improves chemoimmunotherapy of an immune-cold tumor. J Control Release 2025; 381:113579. [PMID: 40023227 PMCID: PMC12002645 DOI: 10.1016/j.jconrel.2025.02.075] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2024] [Revised: 02/22/2025] [Accepted: 02/25/2025] [Indexed: 03/04/2025]
Abstract
An immunoactive complex consisting of a polyethyleneimine derivative (2E'), paclitaxel (PTX), and cyclic dinucleotide (CDN) was developed for chemoimmunotherapy of solid tumors. Each component uniquely contributes to stimulating innate immune response to tumors: 2E' carries PTX and CDN while stimulating antigen-presenting cells, PTX induces immunogenic cell death, and CDN activates the STING pathway. A single intratumoral injection of 2E'/PTX/CDN inhibited the growth of MOC1 oral squamous cell carcinoma and KPCY (2838c3) pancreatic tumors, achieving complete tumor regression in 80-100 % of mice. However, 2E'/PTX/CDN showed limited therapeutic efficacy with immune-cold B16F10 melanoma, accompanied by the increase of innate immune cells in the tumor draining lymph nodes peaking on day 5 post-administration and subsiding thereafter. The addition of a complex of 2E' and siRNA targeting PD-L1 (siPD-L1) at an optimal 5-d interval improved the response in B16F10 melanoma, resulting in tumor-free survival in 50 % of mice and rejection of live tumor rechallenge in 67 % of surviving animals. Consistent with the function of each component, the timed combination of 2E'/PTX/CDN and 2E'/siPD-L1 increased the fractions of mature dendritic cells and M1 macrophages, prevented the increase of regulatory T cells in tumor-draining lymph nodes, and increased melanoma antigen-specific CD8+ T cells in the spleen. These results demonstrate the effectiveness of the 2E'/PTX/CDN complex in the chemoimmunotherapy of solid tumors and highlight the significance of timely intervention to sustain the immunoactive phenotype in its application to immune-cold tumors.
Collapse
Affiliation(s)
- Fanfei Meng
- Department of Industrial and Molecular Pharmaceutics, Purdue University, 575 Stadium Mall Drive, West Lafayette, IN 47907, USA; Department of Biomedical and Nutritional Sciences, University of Massachusetts Lowell, 3 Solomont Way, Lowell, MA 01854, USA
| | - Yanying He
- Department of Industrial and Molecular Pharmaceutics, Purdue University, 575 Stadium Mall Drive, West Lafayette, IN 47907, USA
| | - Jiaqi Zhao
- Department of Industrial and Molecular Pharmaceutics, Purdue University, 575 Stadium Mall Drive, West Lafayette, IN 47907, USA
| | - Zhongyue Yuan
- Department of Industrial and Molecular Pharmaceutics, Purdue University, 575 Stadium Mall Drive, West Lafayette, IN 47907, USA
| | - Jianping Wang
- Department of Industrial and Molecular Pharmaceutics, Purdue University, 575 Stadium Mall Drive, West Lafayette, IN 47907, USA
| | - Karen Gutierrez Parra
- Department of Industrial and Molecular Pharmaceutics, Purdue University, 575 Stadium Mall Drive, West Lafayette, IN 47907, USA
| | - Melissa L Fishel
- Departments of Pediatrics and of Pharmacology and Toxicology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Timothy L Ratliff
- Purdue University Institute for Cancer Research, 201 South University Street, West Lafayette, IN 47907, USA; Department of Comparative Pathobiology, Purdue University, 625 Harrison Street, West Lafayette, Indiana 47907, USA
| | - Yoon Yeo
- Department of Industrial and Molecular Pharmaceutics, Purdue University, 575 Stadium Mall Drive, West Lafayette, IN 47907, USA; Purdue University Institute for Cancer Research, 201 South University Street, West Lafayette, IN 47907, USA; Weldon School of Biomedical Engineering, Purdue University, 206 S Martin Jischke Drive, West Lafayette, IN 47907, USA.
| |
Collapse
|
11
|
Kaur A, Aran KR. Unraveling the cGAS-STING pathway in Alzheimer's disease: A new Frontier in neuroinflammation and therapeutic strategies. Neuroscience 2025; 573:430-441. [PMID: 40185388 DOI: 10.1016/j.neuroscience.2025.04.001] [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: 01/07/2025] [Revised: 03/31/2025] [Accepted: 04/01/2025] [Indexed: 04/07/2025]
Abstract
Alzheimer's disease (AD) is the most prevalent type of neurological disorder characterized by cognitive decline and memory loss, marked by the accumulation of amyloid beta (Aβ) plaques and hyperphosphorylated tau protein, causing extensive neuronal death and neuroinflammation. There is growing evidence that AD development extends beyond the neuronal compartment and has a major impact on the immunological functions of the brain. The cyclic GMP-AMP synthase (cGAS) detects cytosolic DNA, including pathogenic foreign DNA and self-DNA from cellular injury, triggering a type I interferon (IFN-I) response through activation of the stimulator of interferon genes (STING). The activation of the cGAS-STING pathway in response to mitochondrial dysfunction drives neuroinflammation in AD, which is mediated by the release of IFN-I cytokines. Furthermore, the release of oxidized mtDNA is necessary for the stimulation of the nucleotide-binding domain, leucine-rich-containing family, pyrin domain-containing-3 (NLRP3) inflammasome, which is a family of protein complexes that macrophages can produce to induce inflammation. AD becomes severe by the stimulation of the cGAS-STING pathway, which results in sterile inflammation and microglial dysfunction. This review aims to explore the potential impact of the cGAS-STING signaling pathway in the pathogenesis and progression of AD. Additionally; after overviewing recent findings, this article highlights the molecular mechanism involved in the onset of disease and its modulation regarding the therapeutic approach of AD. Finally, deliberated a deep insight, the cGAS-STING axis could provide novel therapeutic avenues for slowing or halting the progression of AD, thereby offering new prospects for treatment development.
Collapse
Affiliation(s)
- Arshdeep Kaur
- Department of Pharmacy Practice, ISF College of Pharmacy, Moga, Punjab, India
| | - Khadga Raj Aran
- Neuropharmacology Division, Department of Pharmacology, ISF College of Pharmacy, Moga, Punjab 142001, India.
| |
Collapse
|
12
|
Kou B, Zhang Y, Zhang W, Zhang J, Jiang R. STING regulates porphyromonas gingivalis lipopolysaccharide-induced pyroptosis and inflammatory response through the NF-κB/NLRP3 signaling pathway in human gingival fibroblasts. Arch Oral Biol 2025; 173:106197. [PMID: 40022901 DOI: 10.1016/j.archoralbio.2025.106197] [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: 08/29/2024] [Revised: 01/07/2025] [Accepted: 02/12/2025] [Indexed: 03/04/2025]
Abstract
OBJECTIVE The production of reactive oxygen species caused by antimicrobial response during periodontitis leads to the activation of NOD-like receptor protein 3 (NLRP3) inflammasome and pyroptosis. Stimulator of interferon genes (STING) has been found to be involved in regulating pyroptosis and inflammation in a variety of diseases. The present study aimed to investigate whether STING is involved in Porphyromonas gingivalis lipopolysaccharide (P.g LPS)-stimulated human gingival fibroblasts (HGFs) by regulating pyroptosis and inflammation. DESIGN After culturing and identifying HGFs, HGFs were treated with P.g LPS. Constructs of si-STING were transfected into HGFs, which were then stimulated with P.g LPS for 24 h. Subsequently, cell viability, pyroptosis, inflammation, oxidative stress and alterations in the STING/TANK-binding kinase 1 (TBK1)/interferon regulatory factor 3 (IRF3)/nuclear factor-kappaB (NF-κB)/NLRP3 signalling pathway were detected. RESULTS P.g LPS significantly enhanced STING expression in HGFs. Downregulation of STING rescued P.g LPS-enhanced pyroptosis, oxidative stress and inflammation in HGFs. Moreover, STING was found to bind directly to TBK1 to increase IRF3 phosphorylation and nuclear translocation of NF-κB, thus promoting NLRP3 inflammasome activation. Downregulation of STING rescued P.g LPS-enhanced TBK1/IRF3/NF-κB/NLRP3 pathway activation. CONCLUSION STING/TBK1/IRF3/NF-κB/NLRP3 is a key pathway governing pyroptosis, oxidative stress and inflammation of HGFs induced by LPS.
Collapse
Affiliation(s)
- Bo Kou
- Department of Stomatology, the 964th Hospital of Joint Logistic Support Force of PLA, Changchun, Jilin 130000, China
| | - Yuna Zhang
- Department of Stomatology, the 964th Hospital of Joint Logistic Support Force of PLA, Changchun, Jilin 130000, China
| | - Wei Zhang
- Department of Stomatology, the 964th Hospital of Joint Logistic Support Force of PLA, Changchun, Jilin 130000, China
| | - Jifang Zhang
- Department of Stomatology, the 964th Hospital of Joint Logistic Support Force of PLA, Changchun, Jilin 130000, China.
| | - Riwen Jiang
- Department of Stomatology, the 964th Hospital of Joint Logistic Support Force of PLA, Changchun, Jilin 130000, China.
| |
Collapse
|
13
|
D’Alessandro A. Red blood cell metabolism: a window on systems health towards clinical metabolomics. Curr Opin Hematol 2025; 32:111-119. [PMID: 40085132 PMCID: PMC11949704 DOI: 10.1097/moh.0000000000000863] [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: 03/16/2025]
Abstract
PURPOSE OF REVIEW This review focuses on recent advances in the understanding of red blood cell (RBC) metabolism as a function of hypoxia and oxidant stress. In particular, we will focus on RBC metabolic alterations during storage in the blood bank, a medically relevant model of erythrocyte responses to energy and redox stress. RECENT FINDINGS Recent studies on over 13 000 healthy blood donors, as part of the Recipient Epidemiology and Donor Evaluation Study (REDS) III and IV-P RBC omics, and 525 diversity outbred mice have highlighted the impact on RBC metabolism of biological factors (age, BMI), genetics (sex, polymorphisms) and exposure (dietary, professional or recreational habits, drugs that are not grounds for blood donor deferral). SUMMARY We review RBC metabolism from basic biochemistry to storage biology, briefly discussing the impact of inborn errors of metabolism and genetic factors on RBC metabolism, as a window on systems metabolic health. Expanding on the concept of clinical chemistry towards clinical metabolomics, monitoring metabolism at scale in large populations (e.g., millions of blood donors) may thus provide insights into population health as a complementary tool to genetic screening and standard clinical measurements.
Collapse
Affiliation(s)
- Angelo D’Alessandro
- Department of Biochemistry and Molecular Genetics, University of Colorado Denver – Anschutz Medical Campus, Aurora, CO, USA
| |
Collapse
|
14
|
Wang C, Lin X, Guan S, Wu Q, Liang S. Dihydroartemisinin Attenuates Radiation-Induced Lung Injury by Inhibiting the cGAS/STING/NF-κB Signaling Pathway. Drug Dev Res 2025; 86:e70090. [PMID: 40285509 PMCID: PMC12032573 DOI: 10.1002/ddr.70090] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2025] [Revised: 03/26/2025] [Accepted: 04/12/2025] [Indexed: 04/29/2025]
Abstract
Dihydroartemisinin (DHA) is a derivative of artemisinin, which affects inflammation, oxidative stress, and immune regulation. However, the mechanism underlying its effects remains largely unknown. This study aims to explore the mechanism by which DHA affects radiation-induced lung injury (RILI), providing new insights for lung radiotherapy. To elucidate its mechanism of action, C57BL/6 J mice were irradiated with 15 Gy whole chest. RILI was evaluated by qRT-PCR, ELISA, histology, Western blot analysis, immunohistochemistry, and RILI signaling cascade studies. In addition, small interfering RNAs were employed to knockdown cGAS proteins in the cGAS-STING signaling pathway in the human bronchial epithelium cell line (BEAS-2B). Both In Vivo and Vitro experiments were conducted to investigate the specific mechanism by which DHA alleviated RILI. We observed the activation of the cGAS-STING pathway, along with the phosphorylation of the downstream target NF-κB and an increase in inflammatory factor levels in the mouse model following radiation exposure. In the cell model, irradiation also triggered the activation of the cGAS-STING signaling pathway and its downstream targets, leading to elevated levels of inflammatory factors. Notably, knocking down the cGAS using small interfering RNA in the BEAS-2B cells significantly alleviated RILI in the cell model. Our study elucidated the mechanism of DHA reducing RILI through the cGAS/STING/NF-κB signaling pathway, and revealed that the GAS/STING/NF-κB axis may be a potential therapeutic target for RILI.
Collapse
Affiliation(s)
- Cailan Wang
- Department of Radiation OncologyGuangxi Medical University Cancer HospitalNanningChina
| | - Xinyi Lin
- Department of Radiation OncologyGuangxi Medical University Cancer HospitalNanningChina
| | - Shichun Guan
- Department of Radiation OncologyGuangxi Medical University Cancer HospitalNanningChina
| | - Qiaoyuan Wu
- Department of Radiation OncologyGuangxi Medical University Cancer HospitalNanningChina
| | - Shixiong Liang
- Department of Radiation OncologyGuangxi Medical University Cancer HospitalNanningChina
| |
Collapse
|
15
|
Zheng J, Ma Z, Liu P, Wei J, Min S, Shan Y, Zhang J, Li Y, Xue L, Tan Z, Wang D. EZH2 inhibits senescence-associated inflammation and attenuates intervertebral disc degeneration by regulating the cGAS/STING pathway via H3K27me3. Osteoarthritis Cartilage 2025; 33:548-559. [PMID: 39938633 DOI: 10.1016/j.joca.2025.02.771] [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: 06/15/2024] [Revised: 01/28/2025] [Accepted: 02/06/2025] [Indexed: 02/14/2025]
Abstract
OBJECTIVE Senescent nucleus pulposus mesenchymal stem cells (NPMSCs) are key instigators of local chronic inflammation and disruptions in nucleus pulposus tissue repair in intervertebral disc degeneration (IVDD). This study aimed to investigate the interplay between EZH2 and NPMSCs senescence-associated inflammation. METHODS Nucleus pulposus samples were collected from IVDD patients (n = 15, F/M = 7/8, average age 47.9 (21-72) year-old). Multiplex immunohistochemistry was conducted to detect the expression of EZH2 and the cGAS/STING pathway. Subsequently, NPMSCs were isolated from 7 patients (n = 7, F/M = 4/3, average age 49.4 (36-68) year-old). After treatment with tert-butyl hydroperoxide and lentivirus-overexpression-EZH2 (Lv-OE-EZH2), real time fluorescent quantitative PCR, immunofluorescence, western blot, and ChIP were used to detect the expression of EZH2 and the cGAS/STING pathway. Micro-CT, magnetic resonance imaging, and histological staining were performed to assess the therapeutic effects of Lv-OE-EZH2 and a STING inhibitor on rat IVDD. All experiment designs were independent. RESULTS In both human nucleus pulposus tissues and an in vitro cell model, EZH2 expression decreased while the cGAS/STING pathway became activated in senescent NPMSCs. ChIP assays and Lv-OE-EZH2 experiments validated that EZH2 epigenetically inhibited STING expression via H3K27me3, thereby impairing the cGAS/STING pathway and attenuating senescence-associated inflammation. Moreover, overexpression of EZH2 (Pfirrmann grade means difference -1.375, p = 0.0089) and inhibition of STING effectively attenuated rat IVDD. CONCLUSION The decreased expression of EZH2 in senescent NPMSCs promotes senescence-associated inflammation and the progression of IVDD, possibly by relieving the transcriptional inhibition of STING and enabling the activation of the cGAS/STING pathway.
Collapse
Affiliation(s)
- Jianrui Zheng
- Department of Bone and Joint Surgery, Peking University Shenzhen Hospital, Shenzhen 518000, PR China.
| | - Zetao Ma
- Department of Bone and Joint Surgery, Peking University Shenzhen Hospital, Shenzhen 518000, PR China.
| | - Pei Liu
- Department of Bone and Joint Surgery, Peking University Shenzhen Hospital, Shenzhen 518000, PR China.
| | - Jiewen Wei
- Department of Bone and Joint Surgery, Peking University Shenzhen Hospital, Shenzhen 518000, PR China; Shantou University Medical College, Shantou 515000, PR China.
| | - Shaoxiong Min
- Department of Spine Surgery, Peking University Shenzhen Hospital, Shenzhen 518000, PR China.
| | - Ying Shan
- Clinical Research Academy, Peking University Shenzhen Hospital, Shenzhen 518000, PR China.
| | - Jianlin Zhang
- Center of Basic Medical Research, Institute of Medical Innovation and Research, Peking University Third Hospital, Beijing 100000, PR China.
| | - Ye Li
- Department of Rehabilitation Sciences, The Hong Kong Polytechnic University, 999077, Hong Kong.
| | - Lixiang Xue
- Center of Basic Medical Research, Institute of Medical Innovation and Research, Peking University Third Hospital, Beijing 100000, PR China.
| | - Zhen Tan
- Department of Bone and Joint Surgery, Peking University Shenzhen Hospital, Shenzhen 518000, PR China.
| | - Deli Wang
- Department of Bone and Joint Surgery, Peking University Shenzhen Hospital, Shenzhen 518000, PR China.
| |
Collapse
|
16
|
Zhang H, Ya J, Sun M, Du X, Ren J, Qu X. Inhibition of the cGAS-STING pathway via an endogenous copper ion-responsive covalent organic framework nanozyme for Alzheimer's disease treatment. Chem Sci 2025; 16:7215-7226. [PMID: 40144496 PMCID: PMC11934151 DOI: 10.1039/d4sc07963a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2024] [Accepted: 03/06/2025] [Indexed: 03/28/2025] Open
Abstract
Inhibition of cGAS-STING overactivation has recently emerged as a promising strategy to counteract Alzheimer's disease (AD). However, current cGAS-STING inhibitors as immunosuppressants suffer from instability, non-specific targeting, and innate immune disruption. Here, an endogenous AD brain copper ion-responsive covalent organic framework (COF)-based nanozyme (denoted as TP@PB-COF@NADH) has been designed for targeted inhibition of the cGAS-STING pathway for AD treatment. The effective trapping of excess brain endogenous copper ions by TP@PB-COF@NADH not only inhibits the Cu2+-induced harmful reactive oxygen species (ROS) production which is one of the mediators of cGAS-STING activation, but also activates the nanozyme activity of TP@PB-COF@NADH. Furthermore, the well-prepared nanozyme catalytically generates NAD+ and consumes hydrogen peroxide (H2O2) through second near-infrared (NIR-II) enhanced nicotinamide adenine dinucleotide (NADH) peroxidase (NPX)-like activity, realizing the efficient inhibition of the cGAS-STING pathway and associated neuroinflammation. Moreover, replenishing NAD+ levels efficiently restores mitochondrial function and ATP supply. In vivo studies demonstrate that TP@PB-COF@NADH with NIR-II irradiation significantly improves cognitive function in 3× Tg-AD mice, with a reduction in amyloid-β (Aβ) plaque, neuroinflammation and neuronal damage. Collectively, this work presents a promising approach for AD treatment by using an AD brain harmful excess endogenous copper ion-responsive and efficient nanozyme.
Collapse
Affiliation(s)
- Haochen Zhang
- Laboratory of Chemical Biology and State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences Changchun Jilin 130022 China
- School of Applied Chemistry and Engineering, University of Science and Technology of China Hefei Anhui 230029 China
| | - Junlin Ya
- Laboratory of Chemical Biology and State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences Changchun Jilin 130022 China
- School of Applied Chemistry and Engineering, University of Science and Technology of China Hefei Anhui 230029 China
| | - Mengyu Sun
- Laboratory of Chemical Biology and State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences Changchun Jilin 130022 China
- School of Applied Chemistry and Engineering, University of Science and Technology of China Hefei Anhui 230029 China
| | - Xiubo Du
- College of Life Sciences and Oceanography, Shenzhen Key Laboratory of Microbial Genetic Engineering, Shenzhen University Shenzhen 518060 China
| | - Jinsong Ren
- Laboratory of Chemical Biology and State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences Changchun Jilin 130022 China
- School of Applied Chemistry and Engineering, University of Science and Technology of China Hefei Anhui 230029 China
| | - Xiaogang Qu
- Laboratory of Chemical Biology and State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences Changchun Jilin 130022 China
- School of Applied Chemistry and Engineering, University of Science and Technology of China Hefei Anhui 230029 China
| |
Collapse
|
17
|
Zhang S, Shang K, Gong L, Xie Q, Sun J, Xu M, Wei X, Xie Z, Liu X, Tang H, Xu Z, Wang W, Xiao H, Lin Z, Han H. Smart Organic-Inorganic Copolymer Nanoparticles Distinguish Between Microglia and Cancer Cells for Synergistic Immunotherapy in Glioma. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025:e2500882. [PMID: 40298877 DOI: 10.1002/advs.202500882] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/05/2025] [Revised: 04/10/2025] [Indexed: 04/30/2025]
Abstract
The stimulator of interferon genes (STING) pathway has emerged as a new immunotherapy strategy with potent local stimulation specificity, showing promising potential to counteract the immunosuppression in glioma. Herein, a tumor microenvironment (TME) responsive nanoagonists are developed based on an organic-inorganic copolymer composed of the polymer PC6AB coupled with manganous phosphate ionic oligomers (MnP). The degradation of nanoagonists into PC6AB and MnP in the acidic TME enables spatiotemporal control of their delivery to tumor cells and immune cells, respectively. PC6AB with membranolytic activity selectively interacts with tumor cell membranes to induce immunogenic cell death, while manganese metal can activate the STING pathway in immune cells and trigger downstream immunostimulatory signals. Nanoagonists can stimulate robust antitumor immunity after local injection into the brain extracellular space (ECS), showing significant therapeutic efficacy in mouse glioma. Nanoagonists can achieve spatiotemporal orchestration of STING activation in response to TME and enhance immune response against "cold" solid tumors, providing a promising approach for clinical immunotherapy.
Collapse
Affiliation(s)
- Shiming Zhang
- Institute of Medical Technology, Peking University Health Science Center, Beijing, 100190, P. R. China
| | - Kun Shang
- Department of Nuclear Medicine, Peking University People's Hospital, Beijing, 100190, P. R. China
| | - Lidong Gong
- Institute of Systems Biomedicine, Department of Pathology, Department of Biophysics School of Basic Medical Sciences, Peking University Health Science Center, Beijing, 100191, P. R. China
| | - Qian Xie
- Division of Nephrology, Peking University Third Hospital, Beijing, 100191, P. R. China
| | - Jianfei Sun
- Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096, P. R. China
| | - Meng Xu
- Institute of Medical Technology, Peking University Health Science Center, Beijing, 100190, P. R. China
| | - Xunbin Wei
- Institute of Medical Technology, Peking University Health Science Center, Beijing, 100190, P. R. China
| | - Zhaoheng Xie
- Institute of Medical Technology, Peking University Health Science Center, Beijing, 100190, P. R. China
| | - Xinyu Liu
- Institute of Medical Technology, Peking University Health Science Center, Beijing, 100190, P. R. China
| | - Hao Tang
- Department of Computer Science, Peking University, Beijing, 100191, P. R. China
| | - Zhengren Xu
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, P. R. China
| | - Wei Wang
- Department of Rehabilitation Radiology, Beijing Rehabilitation Hospital, Capital Medical University, Beijing, 100144, P. R. China
| | - Haihua Xiao
- Beijing National Laboratory for Molecular Science State Key, Laboratory of Polymer Physics and Chemistry, Institute of Chemistry, Chinese Academy of Science, Beijing, 100190, P. R. China
| | - Zhiqiang Lin
- Institute of Systems Biomedicine, Department of Pathology, Department of Biophysics School of Basic Medical Sciences, Peking University Health Science Center, Beijing, 100191, P. R. China
| | - Hongbin Han
- Department of Radiology, Peking University Third Hospital, Institute of Medical Technology, Peking University Health Science Center, Beijing, 100190, P. R. China
| |
Collapse
|
18
|
Huang Y, Mao J, Li Z, Wang W, Ni Z, Cai F, Tang J, Wang W, Zhang L, Zhou L, Jiang X, Wu J, Guo Q, Rui M, Huang Z, Jiang H, Wang L, Xi K, Gu Y, Chen L. Signal Converter-Based Therapy Platform Promoting Aging Bone Healing by Improving Permeability of the Mitochondrial Membrane. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2025:e2500156. [PMID: 40289881 DOI: 10.1002/adma.202500156] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/03/2025] [Revised: 03/19/2025] [Indexed: 04/30/2025]
Abstract
The aging microenvironment promotes persistent inflammation and loss of intrinsic regenerative capacity. These are major obstacles to effective bone tissue repair in older adults. This study aims to explore how physical thermal stimulation can effectively delay the bone marrow mesenchymal stem cells (BMSCs) aging process. Based on this, an implantable physical signal-converter platform is designed as a therapeutic system that enables stable heat signals at the bone injury site under ultrasound stimulation (US). It is found that the therapeutic platform controllably reduces the mitochondrial outer membrane permeabilization of aging BMSCs, bidirectionally inhibiting mitochondrial reactive oxygen species and mitochondrial DNA (mtDNA) leakage. The leakage ratio of mtDNA decreases by 22.7%. This effectively mitigates the activation of the cGAS-STING pathway and its downstream NF-κB signaling induced by oxidative stress in aging BMSCs, thereby attenuating the pathological advancement of chronic inflammation. Thus, it effectively restores the metabolism and osteogenic differentiation of aging BMSCs in vitro, which is further confirmed in a rat model. In the GMPG/US group, the bone mineral density increases 2-3 times at 4 weeks in the rats femoral defect model. Therefore, this ultrasound-based signal-conversion platform provides a promising strategy for aging bone defect repair.
Collapse
Affiliation(s)
- Yiyang Huang
- Department of Orthopedics, First Affiliated Hospital of Soochow University, Orthopedic Institute, Soochow University, 188 Shizi Road, Suzhou, Jiangsu, 215006, P. R. China
| | - Jiannan Mao
- Department of Orthopedics, First Affiliated Hospital of Soochow University, Orthopedic Institute, Soochow University, 188 Shizi Road, Suzhou, Jiangsu, 215006, P. R. China
- Department of Orthopedics, Wuxi Key Laboratory of Biomaterials for Clinical Application, Department of Central Laboratory, Jiangyin Clinical College of Xuzhou Medical University, 163 Shoushan Road, Jiang Yin, 214400, P. R. China
| | - Ziang Li
- Department of Orthopedics, First Affiliated Hospital of Soochow University, Orthopedic Institute, Soochow University, 188 Shizi Road, Suzhou, Jiangsu, 215006, P. R. China
| | - Wenbo Wang
- Department of Orthopedics, First Affiliated Hospital of Soochow University, Orthopedic Institute, Soochow University, 188 Shizi Road, Suzhou, Jiangsu, 215006, P. R. China
| | - Zhengxia Ni
- Department of Orthopedics, First Affiliated Hospital of Soochow University, Orthopedic Institute, Soochow University, 188 Shizi Road, Suzhou, Jiangsu, 215006, P. R. China
| | - Feng Cai
- Department of Orthopedics, First Affiliated Hospital of Soochow University, Orthopedic Institute, Soochow University, 188 Shizi Road, Suzhou, Jiangsu, 215006, P. R. China
| | - Jincheng Tang
- Department of Orthopedics, First Affiliated Hospital of Soochow University, Orthopedic Institute, Soochow University, 188 Shizi Road, Suzhou, Jiangsu, 215006, P. R. China
| | - Wei Wang
- Department of Orthopedics, First Affiliated Hospital of Soochow University, Orthopedic Institute, Soochow University, 188 Shizi Road, Suzhou, Jiangsu, 215006, P. R. China
| | - Lichen Zhang
- Department of Orthopedics, First Affiliated Hospital of Soochow University, Orthopedic Institute, Soochow University, 188 Shizi Road, Suzhou, Jiangsu, 215006, P. R. China
| | - Liang Zhou
- Department of Orthopedics, First Affiliated Hospital of Soochow University, Orthopedic Institute, Soochow University, 188 Shizi Road, Suzhou, Jiangsu, 215006, P. R. China
| | - Xinzhao Jiang
- Department of Orthopedics, First Affiliated Hospital of Soochow University, Orthopedic Institute, Soochow University, 188 Shizi Road, Suzhou, Jiangsu, 215006, P. R. China
| | - Jie Wu
- Department of Orthopedics, First Affiliated Hospital of Soochow University, Orthopedic Institute, Soochow University, 188 Shizi Road, Suzhou, Jiangsu, 215006, P. R. China
| | - Qiangqiang Guo
- Department of Orthopedics, First Affiliated Hospital of Soochow University, Orthopedic Institute, Soochow University, 188 Shizi Road, Suzhou, Jiangsu, 215006, P. R. China
| | - Min Rui
- Department of Orthopedics, First Affiliated Hospital of Soochow University, Orthopedic Institute, Soochow University, 188 Shizi Road, Suzhou, Jiangsu, 215006, P. R. China
- Department of Orthopedics, Wuxi Key Laboratory of Biomaterials for Clinical Application, Department of Central Laboratory, Jiangyin Clinical College of Xuzhou Medical University, 163 Shoushan Road, Jiang Yin, 214400, P. R. China
| | - Ziyan Huang
- Department of Orthopedics, First Affiliated Hospital of Soochow University, Orthopedic Institute, Soochow University, 188 Shizi Road, Suzhou, Jiangsu, 215006, P. R. China
| | - Haochen Jiang
- Department of Orthopedics, First Affiliated Hospital of Soochow University, Orthopedic Institute, Soochow University, 188 Shizi Road, Suzhou, Jiangsu, 215006, P. R. China
| | - Lingjun Wang
- Department of Orthopedics, First Affiliated Hospital of Soochow University, Orthopedic Institute, Soochow University, 188 Shizi Road, Suzhou, Jiangsu, 215006, P. R. China
| | - Kun Xi
- Department of Orthopedics, First Affiliated Hospital of Soochow University, Orthopedic Institute, Soochow University, 188 Shizi Road, Suzhou, Jiangsu, 215006, P. R. China
| | - Yong Gu
- Department of Orthopedics, First Affiliated Hospital of Soochow University, Orthopedic Institute, Soochow University, 188 Shizi Road, Suzhou, Jiangsu, 215006, P. R. China
| | - Liang Chen
- Department of Orthopedics, First Affiliated Hospital of Soochow University, Orthopedic Institute, Soochow University, 188 Shizi Road, Suzhou, Jiangsu, 215006, P. R. China
| |
Collapse
|
19
|
Wei SG, Chen HH, Xie LR, Qin Y, Mai YY, Huang LH, Liao HB. RNA interference-mediated osteoprotegerin silencing increases the receptor activator of nuclear factor-kappa B ligand/osteoprotegerin ratio and promotes osteoclastogenesis. World J Stem Cells 2025; 17:101290. [DOI: 10.4252/wjsc.v17.i4.101290] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Revised: 12/13/2024] [Accepted: 03/21/2025] [Indexed: 04/23/2025] Open
Abstract
BACKGROUND In vivo degradation of bone scaffolds is significantly influenced by osteoclast (OC) activity, which is orchestrated by the interplay between receptor activator of nuclear factor-kappa B ligand (RANKL) and osteoprotegerin (OPG). The ratio of RANKL/OPG is a crucial determinant of OC-mediated bone resorption, which plays an integral role in bone remodeling and scaffold degradation. Elevated levels of RANKL relative to OPG enhance osteoclastogenesis, thereby accelerating the degradation process essential for integrating bone scaffolds into the host tissue.
AIM To elucidate the effects of OPG gene silencing on osteoclastogenesis within rat bone marrow-derived mesenchymal stem cells (BMSCs). By investigating these effects, the study aimed to provide deeper insights into the regulatory mechanisms that influence bone scaffold degradation, potentially leading to improved bone repair and regeneration strategies.
METHODS We employed recombinant lentiviral plasmids to silence the OPG gene in rat BMSCs to achieve the aims. The efficacy of gene silencing was assessed using quantitative reverse transcription polymerase chain reaction and western blot analysis to measure the expression levels of OPG and RANKL. Tartrate-resistant acid phosphatase staining was utilized to evaluate the formation of OCs. Additionally, co-immunoprecipitation assays were conducted to explore the interactions between RANKL and OPG proteins, further assessing the biochemical pathways involved in osteoclastogenesis.
RESULTS The silencing of the OPG gene in BMSCs resulted in a significant increase in the RANKL/OPG ratio, evidenced by decreased expression levels of OPG and increased levels of RANKL. Enhanced osteoclastogenesis was observed through tartrate-resistant acid phosphatase staining, which indicated a substantial rise in OC formation in response to the altered RANKL/OPG balance. The co-immunoprecipitation assays provided concrete evidence of the direct interaction between RANKL and OPG proteins, substantiating their pivotal roles in regulating OC activity.
CONCLUSION The findings from this study underscore the critical role of the RANKL/OPG axis in osteoclastogenesis. Silencing of the OPG gene in BMSCs effectively increases the RANKL/OPG ratio, promoting OC activity and potentially enhancing bone scaffold degradation. This regulatory mechanism offers a promising avenue for modulating bone remodeling processes, which is essential for effective bone repair and the successful integration of bone scaffolds into damaged sites. Future research might focus on optimizing the control of this axis to better facilitate bone tissue engineering and regenerative therapies.
Collapse
Affiliation(s)
- Song-Guan Wei
- Guangxi Key Laboratory of Oral and Maxillofacial Rehabilitation and Reconstruction, College & Hospital of Stomatology, Guangxi Medical University, Nanning 530021, Guangxi Zhuang Autonomous Region, China
- Department of Stomatology, The Fourth Affiliated Hospital of Guangxi Medical University, Liuzhou 545005, Guangxi Zhuang Autonomous Region, China
| | - Hui-Hong Chen
- Guangxi Key Laboratory of Oral and Maxillofacial Rehabilitation and Reconstruction, College & Hospital of Stomatology, Guangxi Medical University, Nanning 530021, Guangxi Zhuang Autonomous Region, China
| | - Liu-Rong Xie
- Guangxi Key Laboratory of Oral and Maxillofacial Rehabilitation and Reconstruction, College & Hospital of Stomatology, Guangxi Medical University, Nanning 530021, Guangxi Zhuang Autonomous Region, China
| | - Yuan Qin
- Guangxi Key Laboratory of Oral and Maxillofacial Rehabilitation and Reconstruction, College & Hospital of Stomatology, Guangxi Medical University, Nanning 530021, Guangxi Zhuang Autonomous Region, China
| | - Yu-Ying Mai
- Guangxi Key Laboratory of Oral and Maxillofacial Rehabilitation and Reconstruction, College & Hospital of Stomatology, Guangxi Medical University, Nanning 530021, Guangxi Zhuang Autonomous Region, China
| | - Lin-Hui Huang
- Guangxi Key Laboratory of Oral and Maxillofacial Rehabilitation and Reconstruction, College & Hospital of Stomatology, Guangxi Medical University, Nanning 530021, Guangxi Zhuang Autonomous Region, China
| | - Hong-Bing Liao
- Guangxi Key Laboratory of Oral and Maxillofacial Rehabilitation and Reconstruction, College & Hospital of Stomatology, Guangxi Medical University, Nanning 530021, Guangxi Zhuang Autonomous Region, China
| |
Collapse
|
20
|
Zhong N, Zu Z, Lu Y, Sha X, Li Y, Liu Y, Lu S, Luo X, Zhou Y, Tao J, Wu F, Teng Z, Tang Y, Wang S. Mitochondria-targeted manganese-based mesoporous silica nanoplatforms trigger cGAS-STING activation and sensitize anti PD-L1 therapy in triple-negative breast cancer. Acta Biomater 2025:S1742-7061(25)00293-4. [PMID: 40294811 DOI: 10.1016/j.actbio.2025.04.040] [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: 01/23/2025] [Revised: 04/11/2025] [Accepted: 04/21/2025] [Indexed: 04/30/2025]
Abstract
Activation of the cyclic GMP-AMP synthase-stimulator of interferon genes (cGAS-STING) pathway could effectively initiate antitumor immunity in triple-negative breast cancer. However, current nuclear DNA-mediated activation of STING pathway remains constrained by the tight protection of nuclear membrane and histones, highlighting the need for new strategies to enhance its efficacy. Mitochondrial DNA (mtDNA), in contrast, is more vulnerable to damage. Herein, our nanoplatforms exploited the high glutathione (GSH) environment characteristic of tumors to release abundant Mn2+, which induced mitochondrial dysfunction and the release of endogenous mtDNA. The released mtDNA, in conjunction with Mn2+ itself functioning as a strong cGAS agonist, effectively activated cGAS-STING pathway. Consequently, the cGAS-STING-dependent secretion of type I interferon successively enhanced the maturation of dendritic cells and cross-priming of CD8+ T cells. In a poorly immunogenic 4T1 tumor model, TPP-MMONs efficiently primed systemic antitumor immunity and significantly enhanced the therapeutic efficacy of αPD-L1 therapy, suppressing tumor growth in both localized and metastatic tumor models. These findings provided an innovative and straightforward strategy to enhance TNBC immunogenicity by targeting mitochondrial damage to induce mtDNA-mediated cGAS-STING activation, thereby sensitizing tumors to immune checkpoint inhibitor therapy. STATEMENT OF SIGNIFICANCE: The cGAS-STING pathway is a promising target for overcoming immunoresistance in TNBC. However, current nuclear DNA-based activation strategies are limited by the tight protection of nuclear membrane and histones. Herein, we reported novel manganese-rich, mitochondria-targeting nanoplatforms (TPP-MMONs), which can release abundant Mn²⁺ and significantly induce mitochondrial dysfunction, leading to the release of mtDNA. As a result, the nanoplatforms can effectively stimulate the cGAS-STING pathway, thereby enhancing immune responses and improving the therapeutic efficacy of αPD-L1 therapy, offering new insights into TNBC treatments.
Collapse
Affiliation(s)
- Nan Zhong
- Laboratory of Molecular Imaging, Department of Radiology, The First Affiliated Hospital with Nanjing Medical University, Nanjing, China
| | - Ziyue Zu
- Laboratory of Molecular Imaging, Department of Radiology, The First Affiliated Hospital with Nanjing Medical University, Nanjing, China
| | - Yishi Lu
- Laboratory of Molecular Imaging, Department of Radiology, The First Affiliated Hospital with Nanjing Medical University, Nanjing, China
| | - Xuan Sha
- Laboratory of Molecular Imaging, Department of Radiology, The First Affiliated Hospital with Nanjing Medical University, Nanjing, China
| | - Yang Li
- Laboratory of Molecular Imaging, Department of Radiology, The First Affiliated Hospital with Nanjing Medical University, Nanjing, China
| | - Yang Liu
- Laboratory of Molecular Imaging, Department of Radiology, The First Affiliated Hospital with Nanjing Medical University, Nanjing, China
| | - Shangyu Lu
- Department of Interventional Radiology, The First Affiliated Hospital with Nanjing Medical University, Nanjing, China
| | - Xi Luo
- Department of Interventional Radiology, The First Affiliated Hospital with Nanjing Medical University, Nanjing, China
| | - Yan Zhou
- Laboratory of Molecular Imaging, Department of Radiology, The First Affiliated Hospital with Nanjing Medical University, Nanjing, China
| | - Jun Tao
- Key Laboratory for Organic Electronics and Information Displays and Jiangsu Key Laboratory for Biosensors, Institute of Advanced Materials, Jiangsu National Synergetic Innovation Centre for Advanced Materials, Nanjing University of Posts and Telecommunications, Nanjing, China
| | - Feiyun Wu
- Laboratory of Molecular Imaging, Department of Radiology, The First Affiliated Hospital with Nanjing Medical University, Nanjing, China
| | - Zhaogang Teng
- Key Laboratory for Organic Electronics and Information Displays and Jiangsu Key Laboratory for Biosensors, Institute of Advanced Materials, Jiangsu National Synergetic Innovation Centre for Advanced Materials, Nanjing University of Posts and Telecommunications, Nanjing, China
| | - Yuxia Tang
- Laboratory of Molecular Imaging, Department of Radiology, The First Affiliated Hospital with Nanjing Medical University, Nanjing, China
| | - Shouju Wang
- Laboratory of Molecular Imaging, Department of Radiology, The First Affiliated Hospital with Nanjing Medical University, Nanjing, China.
| |
Collapse
|
21
|
Premadasa LS, McDew-White M, Romero L, Gondo B, Drawec JA, Ling B, Okeoma CM, Mohan M. Epigenetic modulation of the NLRP6 inflammasome sensor as a therapeutic modality to reduce necroptosis-driven gastrointestinal mucosal dysfunction in HIV/SIV infection. Cell Commun Signal 2025; 23:199. [PMID: 40281523 PMCID: PMC12023470 DOI: 10.1186/s12964-025-02193-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2024] [Accepted: 04/08/2025] [Indexed: 04/29/2025] Open
Abstract
BACKGROUND Gastrointestinal (GI) disease/dysfunction persists in people living with HIV (PLWH) receiving suppressive combination anti-retroviral therapy (ART) leading to epithelial barrier breakdown, microbial translocation and systemic inflammation that can drive non-AIDS associated comorbidities. Although epigenetic mechanisms are predicted to drive GI dysfunction, they remain unknown and unaddressed in HIV/SIV infection. The present study investigated genome-wide changes in DNA methylation, and gene expression exclusively in colon epithelial cells (CE) in response to simian immunodeficiency virus infection (SIV) and long-term low-dose delta-9-tetrahydrocannabinol (THC). METHODS Using reduced-representation bisulfite sequencing, we characterized DNA methylation changes in colonic epithelium (CE) of uninfected controls (n=5) and SIV-infected rhesus macaques (RMs) administered vehicle (VEH/SIV; n=7) or THC (THC/SIV; n=6). Intact jejunum resection segments (~5cm) were collected from sixteen ART treated SIV-infected RMs [(VEH/SIV/ART; n=8) and (THC/SIV/ART; n=8)] to confirm protein expression data identified in the colon of ART-naïve SIV-infected RMs. Transcriptomics data was used to confirm expression of differentially methylated genes. Protein expression of differentially methylated genes and their downstream targets was assessed using Immunofluorescence followed by HALO quantification. RESULTS SIV infection in ART-naïve RMs induced marked hypomethylation throughout promoter-associated CpG islands (paCGIs) in genes related to inflammatory response (NLRP6, cGAS), cellular adhesion (PCDH17, CDH7) and proliferation (WIF1, SFRP1, TERT, and HAND2) in CEs. Moreover, low-dose THC reduced NLRP6 protein expression in CE by hypermethylating the NLRP6 paCGI and blocked polyI:C induced NLRP6 upregulation in vitro. In ART suppressed SIV-infected RMs, significant NLRP6 protein upregulation during acute infection was unaffected by long-term ART administration during chronic infection despite successful plasma and tissue viral suppression. In this group, NLRP6 protein upregulation was associated with significantly increased expression of necroptosis-driving proteins; phosphorylated-RIPK3(Ser199), phosphorylated-MLKL(Thr357/Ser358), and HMGB1. Most strikingly, adding ART to THC-treated SIV-infected RMs effectively reduced NLRP6 and necroptosis-driving protein expression to pre-infection levels. CONCLUSIONS We conclude that DNA hypomethylation-assisted NLRP6 upregulation can lead to its constitutively high expression resulting in the activation of necroptosis signaling via the RIPK3/p-MLKL pathway that can eventually drive intestinal epithelial loss/death. From a clinical standpoint, low-dose phytocannabinoids in combination with ART could safely and successfully reduce/reverse persistent GI inflammatory responses via modulating DNA methylation.
Collapse
Affiliation(s)
- Lakmini S Premadasa
- Southwest National Primate Research Center, Texas Biomedical Research Institute, San Antonio, TX, USA
| | - Marina McDew-White
- Southwest National Primate Research Center, Texas Biomedical Research Institute, San Antonio, TX, USA
| | - Luis Romero
- Southwest National Primate Research Center, Texas Biomedical Research Institute, San Antonio, TX, USA
| | - Beverly Gondo
- Southwest National Primate Research Center, Texas Biomedical Research Institute, San Antonio, TX, USA
| | - Jade A Drawec
- Southwest National Primate Research Center, Texas Biomedical Research Institute, San Antonio, TX, USA
| | - Binhua Ling
- Southwest National Primate Research Center, Texas Biomedical Research Institute, San Antonio, TX, USA
| | - Chioma M Okeoma
- Department of Pathology, Microbiology, and Immunology, New York Medical College, Valhalla, NY, 10595-1524, USA.
- Lovelace Biomedical Institute, Albuquerque, NM, 87108-5127, USA.
| | - Mahesh Mohan
- Southwest National Primate Research Center, Texas Biomedical Research Institute, San Antonio, TX, USA.
| |
Collapse
|
22
|
Li H, Wu Y, Xiang L, Zhao Q, Liu L, Zhu Z, Lin W, Li Z, Yang Y, Ze Y, Zhang L, Fu P, Guo Y, Zhang P, Shao B. A20 attenuates oxidized self-DNA-mediated inflammation in acute kidney injury. Signal Transduct Target Ther 2025; 10:154. [PMID: 40280946 PMCID: PMC12032302 DOI: 10.1038/s41392-025-02194-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Revised: 02/03/2025] [Accepted: 02/20/2025] [Indexed: 04/29/2025] Open
Abstract
The ubiquitin-editing enzyme A20 is known to regulate inflammation and maintain homeostasis, but its role in self-DNA-mediated inflammation in acute kidney injury (AKI) is not well understood. Here, our study demonstrated that oxidized self-DNA accumulates in the serum of AKI mice and patients. This oxidized self-DNA exacerbates the progression of AKI by activating the cGAS-STING pathway and NLRP3 inflammasome. While inhibition of the STING pathway only slightly attenuates AKI progression, suppression of NLRP3 inflammasome-mediated pyroptosis significantly alleviates AKI progression and improves the survival of AKI mice. Subsequently, we found that Tnfaip3 (encoding A20) is significantly upregulated following oxidized self-DNA treatment. A20 significantly alleviates AKI development by dampening STING signaling pathway and NLRP3-mediated pyroptosis. Moreover, A20-derived peptide (P-II) also significantly alleviates ox-dsDNA-induced pyroptosis and improves the survival and renal injury of AKI mice. Mechanistically, A20 competitively binds with NEK7 and thus inhibiting NLRP3 inflammasome. A20 and P-II interfere with the interaction between NEK7 and NLRP3 through Lys140 of NEK7. Mutation of Lys140 effects on the interaction of NEK7 with A20 and/or NLRP3 complex. Conditional knockout of NEK7 in macrophages or pharmacological inhibition of NEK7 both significantly rescue AKI mouse models. This study reveals a new mechanism by which A20 attenuates oxidized self-DNA-mediated inflammation and provides a new therapeutic strategy for AKI.
Collapse
Affiliation(s)
- Hanwen Li
- 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, Sichuan, PR China
| | - Yongyao Wu
- 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, Sichuan, PR China
| | - Lisha Xiang
- Division of Thoracic Tumor Multimodality Treatment and Department of Medical Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, PR China
| | - Qing Zhao
- 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, Sichuan, PR China
| | - Lu Liu
- Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, PR China
| | - Zhixiong Zhu
- Department of Biotherapy, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, PR China
| | - Weimin Lin
- 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, Sichuan, PR China
| | - Zhan Li
- Department of Stem Cell and Regenerative Medicine, State Key Laboratory of Trauma, Burn and Combined Injury, Daping Hospital, Army Medical University, Chongqing, PR China
| | - Yang Yang
- Department of Biotherapy, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, PR China
| | - Yiting Ze
- 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, Sichuan, PR China
| | - Lulu Zhang
- College of Foreign Languages and Cultures, Sichuan University. Sichuan University, Chengdu, Sichuan, PR China
| | - Ping Fu
- Kidney Research Institute, National Clinical Research Center for Geriatrics and Division of Nephrology, West China Hospital of Sichuan University, Chengdu, Sichuan, PR China
| | - Yingqiang Guo
- Department of Cardiovascular Surgery and Cardiovascular Surgery Research Laboratory, West China Hospital, Sichuan University, Chengdu, Sichuan, PR China.
| | - Ping Zhang
- 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, Sichuan, PR China.
| | - Bin Shao
- Department of Biotherapy, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, PR China.
- Department of Cardiovascular Surgery and Cardiovascular Surgery Research Laboratory, West China Hospital, Sichuan University, Chengdu, Sichuan, PR China.
| |
Collapse
|
23
|
Zhao Y, Xu T, Wu Z, Li N, Liang Q. Rebalancing redox homeostasis: A pivotal regulator of the cGAS-STING pathway in autoimmune diseases. Autoimmun Rev 2025:103823. [PMID: 40286888 DOI: 10.1016/j.autrev.2025.103823] [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: 07/11/2024] [Revised: 04/22/2025] [Accepted: 04/23/2025] [Indexed: 04/29/2025]
Abstract
Autoimmune diseases (ADs) arise from the breakdown of immune tolerance to self-antigens, leading to pathological tissue damage. Proinflammatory cytokine overproduction disrupts redox homeostasis across diverse cell populations, generating oxidative stress that induces DNA damage through multiple mechanisms. Oxidative stress-induced alterations in membrane permeability and DNA damage can lead to the recognition of double-stranded DNA (dsDNA), mitochondrial DNA (mtDNA) and micronuclei-DNA(MN-DNA) by DNA sensors, thereby initiating activation of the cyclic GMP-AMP synthase-stimulator of interferon genes (cGAS-STING) pathway. While previous reviews have characterized cGAS-STING activation in autoimmunity, the reciprocal regulation between redox homeostasis and cGAS-STING activation remains insufficiently defined. This narrative review examines oxidative stress-mediated DNA damage as a critical driver of pathological cGAS-STING signaling and delineates molecular mechanisms linking redox homeostasis to autoimmune pathogenesis. Furthermore, we propose therapeutic strategies that combine redox restoration with the attenuation of aberrant cGAS-STING activation, thereby establishing a mechanistic foundation for precision interventions in autoimmune disorders. METHODS: The manuscript is formatted as a narrative review. We conducted a comprehensive search strategy using electronic databases such as PubMed, Google Scholar and Web of Science. Various keywords were used, such as "cGAS-STING," "Redox homeostasis," "Oxidative stress," "pentose phosphate pathway," "Ferroptosis," "mtDNA," "dsDNA," "DNA damage," "Micronuclei," "Reactive oxygen species," "Reactive nitrogen species," "Nanomaterial," "Autoimmune disease," "Systemic lupus erythematosus," "Type 1 diabetes," "Rheumatoid arthritis," "Multiple sclerosis," "Experimental autoimmune encephalomyelitis," "Psoriasis," etc. The titles and abstracts were reviewed for inclusion into this review. After removing duplicates and irrelevant studies, 174 articles met inclusion criteria (original research, English language).
Collapse
Affiliation(s)
- Yuchen Zhao
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, 725 Wan-Ping South Road, Shanghai 200032, China; Spine Institute, Shanghai University of Traditional Chinese Medicine, 725 Wan-Ping South Road, Shanghai 200032, China; Shanghai University of Traditional Chinese Medicine, 1200 Cailun Road, Shanghai 201203, China
| | - Tianhao Xu
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, 725 Wan-Ping South Road, Shanghai 200032, China; Spine Institute, Shanghai University of Traditional Chinese Medicine, 725 Wan-Ping South Road, Shanghai 200032, China
| | - Zhaoshun Wu
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, 725 Wan-Ping South Road, Shanghai 200032, China; Spine Institute, Shanghai University of Traditional Chinese Medicine, 725 Wan-Ping South Road, Shanghai 200032, China
| | - Ning Li
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, 725 Wan-Ping South Road, Shanghai 200032, China; Spine Institute, Shanghai University of Traditional Chinese Medicine, 725 Wan-Ping South Road, Shanghai 200032, China.
| | - Qianqian Liang
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, 725 Wan-Ping South Road, Shanghai 200032, China; Spine Institute, Shanghai University of Traditional Chinese Medicine, 725 Wan-Ping South Road, Shanghai 200032, China; Shanghai University of Traditional Chinese Medicine, 1200 Cailun Road, Shanghai 201203, China.
| |
Collapse
|
24
|
Zhang W, Zhang Y, Han L, Bo T, Qi Z, Zhong H, Xu H, Hu L, Chen S, Zhang S. Double-stranded DNA enhances platelet activation, thrombosis, and myocardial injury via cyclic GMP-AMP synthase. Cardiovasc Res 2025; 121:353-366. [PMID: 39302147 DOI: 10.1093/cvr/cvae218] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Revised: 06/19/2024] [Accepted: 08/07/2024] [Indexed: 09/22/2024] Open
Abstract
AIMS Elevated dsDNA levels in ST-elevated myocardial infarction (STEMI) patients are associated with increased infarct size and worse clinical outcomes. However, the direct effect of dsDNA on platelet activation remains unclear. This study aims to investigate the direct influence of dsDNA on platelet activation, thrombosis, and the underlying mechanisms. METHODS AND RESULTS Analysis of clinical samples revealed elevated plasma dsDNA levels in STEMI patients, which positively correlated with platelet aggregation and markers of neutrophil extracellular traps such as MPO-DNA and CitH3. Platelet assays demonstrated the activation of the cGAS-STING pathway in platelets from STEMI patients. DsDNA directly potentiated platelet activation and thrombus formation. Mechanistic studies using G150 (cGAS inhibitor), H151 (STING inhibitor), and MCC950 (NLRP3 inhibitor), as well as cGAS-/-, STING-/-, and NLRP3-/- mice, showed that dsDNA activated cGAS, a previously unreported DNA sensor in platelets, and induced activation of the STING/NLRP3/caspase-1/IL-1β axis. This cascade enhanced platelet activation and thrombus formation. Platelet cGAS depletion or Palbociclib, a cGAS-STING inhibitor, approved by the FDA for advanced breast cancer, ameliorated myocardial ischaemia-reperfusion injury in ApoE-/- mice fed with a high-fat diet for 12 weeks. CONCLUSIONS These results suggested that dsDNA is a novel driver of platelet activation and thrombus formation in STEMI patients.
Collapse
Affiliation(s)
- Wei Zhang
- NHC Key Laboratory of Glycoconjugate Research, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, 130 Dong'an Road, Shanghai 200032, China
| | - Yan Zhang
- NHC Key Laboratory of Glycoconjugate Research, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, 130 Dong'an Road, Shanghai 200032, China
| | - Liping Han
- Department of Transfusion Medicine, Huashan Hospital, Fudan University, 12 Middle Wulumuqi Road, Shanghai 200040, China
| | - Tao Bo
- NHC Key Laboratory of Glycoconjugate Research, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, 130 Dong'an Road, Shanghai 200032, China
| | - Zhiyong Qi
- Department of Cardiology, National Clinical Research Center for Interventional Medicine, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, 180 Fenglin Road, Shanghai 200032, China
| | - Haoxuan Zhong
- Department of Cardiology, National Clinical Research Center for Interventional Medicine, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, 180 Fenglin Road, Shanghai 200032, China
| | - Huajie Xu
- Department of Cardiology, National Clinical Research Center for Interventional Medicine, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, 180 Fenglin Road, Shanghai 200032, China
| | - Liang Hu
- The Research Center for Traditional Chinese Medicine, Shanghai Institute of Infectious Diseases and Biosecurity, School of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, 1200 Cailun Road, Shanghai 201203, China
| | - She Chen
- NHC Key Laboratory of Glycoconjugate Research, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, 130 Dong'an Road, Shanghai 200032, China
| | - Si Zhang
- NHC Key Laboratory of Glycoconjugate Research, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, 130 Dong'an Road, Shanghai 200032, China
| |
Collapse
|
25
|
Guilbaud E, Naulin F, Meziani L, Deutsch E, Galluzzi L. Impact of radiation therapy on the immunological tumor microenvironment. Cell Chem Biol 2025:S2451-9456(25)00099-6. [PMID: 40280118 DOI: 10.1016/j.chembiol.2025.04.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2024] [Revised: 02/22/2025] [Accepted: 04/03/2025] [Indexed: 04/29/2025]
Abstract
External beam radiation therapy (RT) is a cornerstone of modern cancer management, being utilized in both curative and palliative settings due to its safety, efficacy, and widespread availability. A primary biological effect of RT is DNA damage, which leads to significant cytostatic and cytotoxic effects. Importantly, malignant cells possess a limited capacity for DNA repair compared to normal cells, and when combined with irradiation techniques that minimize damage to healthy tissues, this creates an advantageous therapeutic window. However, the clinical effectiveness of RT also appears to involve both direct and indirect interactions between RT and non-transformed components of the tumoral ecosystem, particularly immune cells. In this review, we describe the molecular and cellular mechanisms by which irradiated cancer cells modify the immunological tumor microenvironment and how such changes ultimately impact tumor growth.
Collapse
Affiliation(s)
- Emma Guilbaud
- Cancer Signaling and Microenvironment Program, Fox Chase Cancer Center, Philadelphia, PA, USA
| | - Flavie Naulin
- Cancer Signaling and Microenvironment Program, Fox Chase Cancer Center, Philadelphia, PA, USA; Department of Radiotherapy, Gustave Roussy Cancer Campus, Villejuif, France; INSERM RAMO-IT U1030, Villejuif, France; Faculty of Medicine, University of Paris-Saclay, Le Kremlin, Bicêtre, France
| | - Lydia Meziani
- Department of Radiotherapy, Gustave Roussy Cancer Campus, Villejuif, France; INSERM RAMO-IT U1030, Villejuif, France; Faculty of Medicine, University of Paris-Saclay, Le Kremlin, Bicêtre, France
| | - Eric Deutsch
- Department of Radiotherapy, Gustave Roussy Cancer Campus, Villejuif, France; INSERM RAMO-IT U1030, Villejuif, France; Faculty of Medicine, University of Paris-Saclay, Le Kremlin, Bicêtre, France.
| | - Lorenzo Galluzzi
- Cancer Signaling and Microenvironment Program, Fox Chase Cancer Center, Philadelphia, PA, USA.
| |
Collapse
|
26
|
Du H, Cui D, Hu S, Zhou X, Lin X, Fu X, Feng S, Xu S, Jian W, Guo Y, Zhang S, Chen Q. The induction of type I interferonopathy in Trex1-P212fs mice is mediated by activation of the cGAS-STING pathway. Int J Biol Macromol 2025; 310:143414. [PMID: 40268028 DOI: 10.1016/j.ijbiomac.2025.143414] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2024] [Revised: 04/03/2025] [Accepted: 04/20/2025] [Indexed: 04/25/2025]
Abstract
The cGAS-STING pathway is crucial for immune tolerance, pathogen resistance, and tumor immunity. Knocking out the cGAS gene can reverse the type I interferonopathy seen in Trex1-/- and Trex1D18N/D18N mice. TREX1, a key DNA-specific exonuclease in mammalian cells, degrades cytoplasmic DNA to prevent excessive immune activation. Mutations in TREX1 are linked to various autoimmune diseases. In prior research, we generated a Trex1-P212fs mouse model associated with systemic lupus erythematosus (SLE) using CRISPR-Cas9 gene editing. This model displays systemic inflammation that mirrors numerous characteristics of both Aicardi-Goutières syndrome (AGS) and SLE in humans. In this study, we found that the TREX1-P212fs mutation resulted in reduced dsDNA enzyme activity. DNA accumulation was present in the cytoplasm of Trex1P212fs/P212fs MEFs. Nonetheless, the role of the cGAS-STING pathway in mediating the disease phenotype in Trex1-P212fs mice associated with SLE has yet to be elucidated. We observed that cGas knockout mitigated systemic inflammation, lymphocyte proliferation, vasculitis, renal disease, and spontaneous T cell activation in Trex1-P212fs mice. Similarly, inhibition of STING with C-176 treatment ameliorated the disease phenotype in Trex1-P212fs mice. These findings elucidate the pathogenesis of TREX1-P212fs-associated type I interferonopathy and offer potential therapeutic targets for their treatment.
Collapse
Affiliation(s)
- Hekang Du
- Department of Pathology, The First Affiliated Hospital, Fujian Medical University, 20 Chazhong Road, Fuzhou 350005, China; Fujian Key Laboratory of Innate Immune Biology, Biomedical Research Center of South China, College of Life Sciences, Fujian Normal University Qishan Campus, College Town, Fuzhou, Fujian Province 350117, China.; Department of Pathology, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, 999 Huashan Road, Fuzhou 350212, China
| | - Dongya Cui
- Fujian Key Laboratory of Innate Immune Biology, Biomedical Research Center of South China, College of Life Sciences, Fujian Normal University Qishan Campus, College Town, Fuzhou, Fujian Province 350117, China
| | - Shun Hu
- Department of Pathology, The First Affiliated Hospital, Fujian Medical University, 20 Chazhong Road, Fuzhou 350005, China; Department of Pathology, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, 999 Huashan Road, Fuzhou 350212, China
| | - Xueyuan Zhou
- Fujian Key Laboratory of Innate Immune Biology, Biomedical Research Center of South China, College of Life Sciences, Fujian Normal University Qishan Campus, College Town, Fuzhou, Fujian Province 350117, China
| | - Xiaofang Lin
- Fujian Key Laboratory of Innate Immune Biology, Biomedical Research Center of South China, College of Life Sciences, Fujian Normal University Qishan Campus, College Town, Fuzhou, Fujian Province 350117, China
| | - Xiaodan Fu
- Department of Pathology, The First Affiliated Hospital, Fujian Medical University, 20 Chazhong Road, Fuzhou 350005, China; Department of Pathology, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, 999 Huashan Road, Fuzhou 350212, China
| | - Sisi Feng
- Department of Pathology, The First Affiliated Hospital, Fujian Medical University, 20 Chazhong Road, Fuzhou 350005, China; Department of Pathology, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, 999 Huashan Road, Fuzhou 350212, China
| | - Shan Xu
- Fujian Key Laboratory of Innate Immune Biology, Biomedical Research Center of South China, College of Life Sciences, Fujian Normal University Qishan Campus, College Town, Fuzhou, Fujian Province 350117, China
| | - Wentin Jian
- Department of Pathology, The First Affiliated Hospital, Fujian Medical University, 20 Chazhong Road, Fuzhou 350005, China; Department of Pathology, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, 999 Huashan Road, Fuzhou 350212, China
| | - Yuanli Guo
- Department of Pathology, The First Affiliated Hospital, Fujian Medical University, 20 Chazhong Road, Fuzhou 350005, China; Department of Pathology, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, 999 Huashan Road, Fuzhou 350212, China
| | - Sheng Zhang
- Department of Pathology, The First Affiliated Hospital, Fujian Medical University, 20 Chazhong Road, Fuzhou 350005, China; Department of Pathology, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, 999 Huashan Road, Fuzhou 350212, China..
| | - Qi Chen
- Fujian Key Laboratory of Innate Immune Biology, Biomedical Research Center of South China, College of Life Sciences, Fujian Normal University Qishan Campus, College Town, Fuzhou, Fujian Province 350117, China..
| |
Collapse
|
27
|
Ding K, Zhang L, Zhang Y, Jing Y, Liao H, Chen R, Meng Q. TFAM Deficiency Triggers mtDNA Leakage and cGAS-STING-Mediated Intestinal Ischemia-Reperfusion Injury. Inflammation 2025:10.1007/s10753-025-02302-8. [PMID: 40257650 DOI: 10.1007/s10753-025-02302-8] [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: 02/11/2025] [Revised: 03/21/2025] [Accepted: 04/11/2025] [Indexed: 04/22/2025]
Abstract
Intestinal Ischemia-Reperfusion (IIR) injury is a common clinical pathophysiological condition, yet the complex molecular mechanisms underlying its pathology remain incompletely understood. This study aims to explore the precise molecular mechanisms of IIR injury, with a focus on the role of the cGAS-STING signaling pathway. Using a mouse IIR model and hypoxia/reoxygenation (HR) model in HT-29 cells and small intestinal organoids, we observed that IIR significantly induces oxidative stress and activates the cGAS-STING pathway, which is associated with exacerbated small intestinal tissue damage and enhanced inflammatory responses. Further investigation revealed that mitochondrial DNA (mtDNA) leakage is a critical trigger for the activation of the cGAS-STING pathway. The introduction of exogenous mtDNA into cells activated the STING pathway and exacerbated cellular damage. In contrast, the depletion of intracellular mtDNA effectively suppressed HR-induced activation of the cGAS-STING pathway. Mechanistically, we found that IIR downregulates mitochondrial transcription factor A (TFAM), which subsequently affects mtDNA stability, promoting the release of mtDNA into the cytoplasm and triggering the cGAS-STING pathway. Overexpression of TFAM stabilized mtDNA, reduced the accumulation of cytoplasmic mtDNA, inhibited cGAS-STING pathway activation, and alleviated cellular damage. Moreover, STING-deficient mice exhibited reduced inflammation, less tissue damage, and improved survival rates following IIR, highlighting the critical role of the STING pathway in IIR-induced injury. Our findings elucidate the close association between oxidative stress, inflammation, and cGAS-STING pathway activation in IIR. mtDNA leakage and TFAM downregulation are key mechanisms driving this activation. Importantly, TFAM plays a crucial role in stabilizing mtDNA and reducing mtDNA leakage during IIR. These results not only deepen our understanding of the molecular pathogenesis of IIR injury but also provide potential therapeutic strategies targeting the cGAS-STING pathway for treating IIR-related diseases.
Collapse
Affiliation(s)
- Ke Ding
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Jiefang Road 238, Wuhan, 430060, Hubei, China
| | - Lele Zhang
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Jiefang Road 238, Wuhan, 430060, Hubei, China
| | - Yiguo Zhang
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Jiefang Road 238, Wuhan, 430060, Hubei, China
| | - Yixin Jing
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Jiefang Road 238, Wuhan, 430060, Hubei, China
| | - Huiyang Liao
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Jiefang Road 238, Wuhan, 430060, Hubei, China
| | - Rong Chen
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Jiefang Road 238, Wuhan, 430060, Hubei, China
| | - Qingtao Meng
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Jiefang Road 238, Wuhan, 430060, Hubei, China.
| |
Collapse
|
28
|
Zhang S, Tang J, Cui P, He W, Lin X, Wang S, Liu Y, Tan X, Xu S, Feng M, Lai H. Accurate and Efficient Detection of Nasopharyngeal Carcinoma Using Multi-Dimensional Features of Plasma Cell-Free DNA. Head Neck 2025. [PMID: 40256837 DOI: 10.1002/hed.28154] [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: 12/16/2024] [Revised: 03/08/2025] [Accepted: 03/23/2025] [Indexed: 04/22/2025] Open
Abstract
BACKGROUND The incidence of Nasopharyngeal carcinoma (NPC) is rising in recent years, especially in some non-developed parts of the world. Hence, cost-efficient means for sensitive detection of NPC are vital. METHODS We recruited 646 participants, including healthy individuals, patients with benign nasopharyngeal diseases, and NPC patients for plasma cell-free DNA(cfDNA), which underwent low-depth whole-genome sequencing (WGS) to extract multi-dimensional molecular features, including fragmentation pattern, end motif, copy number variation(CNV), and transcription factors(TF). Based on these features, we employed a machine learning algorithm to build prediction models for NPC detection. RESULTS We achieved a sensitivity of 95.8% and a specificity of 99.4% to discriminate NPC patients from healthy individuals. CONCLUSIONS This study can be a proof-of-concept for these multi-dimensional molecular features to be implemented as a noninvasive approach for the detection and even early detection of NPC.
Collapse
Affiliation(s)
- Song Zhang
- Department of Otolaryngology, Shenzhen Guangming District People's Hospital, Shenzhen, China
| | - Jiahui Tang
- Department of Otolaryngology, Shenzhen Guangming District People's Hospital, Shenzhen, China
| | - Pin Cui
- Shenzhen Rapha Biotechnology Incorporate, Shenzhen, China
| | - Weihuang He
- Shenzhen Rapha Biotechnology Incorporate, Shenzhen, China
| | - Xiaohui Lin
- Department of Oncology, People's Hospital of Shenzhen Baoan District, The Second Affiliated Hospital of Shenzhen University, Shenzhen, China
| | - Shubing Wang
- Department of Oncology, Shenzhen Key Laboratory of Gastrointestinal Cancer Translational Research, Cancer Institute, Peking University Shenzhen Hospital, Shenzhen-Peking University-Hong Kong University of Science and Technology Medical Center, Shenzhen, China
| | - Yuanxian Liu
- Department of Otolaryngology, Shenzhen Traditional Chinese Medicine Hospital, Shenzhen, China
| | - Xiaohua Tan
- Department of Oncology, Shenzhen Third People's Hospital, Shenzhen, China
| | - Shu Xu
- Department of Oncology, Shenzhen Guangming District People's Hospital, Shenzhen, China
| | - Mingji Feng
- Department of Oncology, Shenzhen Key Laboratory of Gastrointestinal Cancer Translational Research, Cancer Institute, Peking University Shenzhen Hospital, Shenzhen-Peking University-Hong Kong University of Science and Technology Medical Center, Shenzhen, China
| | - Hanming Lai
- Department of Oncology, Shenzhen Key Laboratory of Gastrointestinal Cancer Translational Research, Cancer Institute, Peking University Shenzhen Hospital, Shenzhen-Peking University-Hong Kong University of Science and Technology Medical Center, Shenzhen, China
| |
Collapse
|
29
|
Tapia PJ, Martina JA, Contreras PS, Prashar A, Jeong E, De Nardo D, Puertollano R. TFEB and TFE3 regulate STING1-dependent immune responses by controlling type I interferon signaling. Autophagy 2025:1-18. [PMID: 40195022 DOI: 10.1080/15548627.2025.2487036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2024] [Revised: 03/20/2025] [Accepted: 03/27/2025] [Indexed: 04/09/2025] Open
Abstract
STING1 is an essential component of the innate immune defense against a wide variety of pathogens. Whereas induction of type I interferon (IFN) responses is one of the best-defined functions of STING1, our transcriptomic analysis revealed IFN-independent activities of STING1 in macrophages, including transcriptional upregulation of numerous lysosomal and autophagic genes. This upregulation was mediated by the STING1-induced activation of the transcription factors TFEB and TFE3, and led to increased autophagy, lysosomal biogenesis, and lysosomal acidification. TFEB and TFE3 also modulated IFN-dependent STING1 signaling by controlling IRF3 activation. IFN production and cell death were increased in TFEB- and TFE3-depleted iBMDMs. Conversely, TFEB overexpression led to reduced IRF3 activation and an almost complete inhibition of IFN synthesis and secretion, resulting in decreased CASP3 activation and increased cell survival. Our study reveals a key role of TFEB and TFE3 as regulators of STING1-mediated innate antiviral immunity.Abbreviation: ACOD1/IRG1, aconitate decarboxylase 1; cGAMP, cyclic guanosine monophosphate-adenosine monophosphate; CGAS, cyclic GMP-AMP synthase; DMXAA, 5,6-dimethylxanthenone-4-acetic acid; EIF4EBP1, eukaryotic translation initiation factor 4E binding protein 1; GABARAP, GABA type A receptor-associated protein; HSV-1, herpes simplex virus type; iBMDMs, immortalized bone marrow-derived macrophages; IFN, type I interferon; IFNB, interferon beta; IKBKE, inhibitor of nuclear factor kappa B kinase subunit epsilon; IRF3, interferon regulatory factor 3; LAMP1, lysosomal associated membrane protein 1; LAMP2, lysosomal associated membrane protein 2; MTORC1, mechanistic target of rapamycin kinase complex 1; RPS6, ribosomal protein S6; STING1, stimulator of interferon response cGAMP interactor 1; TBK1, TANK binding kinase 1; TFE3, transcription factor binding to IGHM enhancer 3; TFEB, transcription factor EB.
Collapse
Affiliation(s)
- Pablo J Tapia
- Cell and Developmental Biology Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - José A Martina
- Cell and Developmental Biology Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Pablo S Contreras
- Cell and Developmental Biology Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Akriti Prashar
- Cell and Developmental Biology Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Eutteum Jeong
- Cell and Developmental Biology Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Dominic De Nardo
- Department of Biochemistry and Molecular Biology, Monash Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia
| | - Rosa Puertollano
- Cell and Developmental Biology Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
30
|
Sheng M, Zheng Y, Li K, Ye C, Cao G. Optimized electroporation buffer improves transfection and prime editing efficiency in adult bovine fibroblasts. Gene 2025; 946:149315. [PMID: 39922551 DOI: 10.1016/j.gene.2025.149315] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2024] [Revised: 01/25/2025] [Accepted: 02/03/2025] [Indexed: 02/10/2025]
Abstract
For livestock breeding, using somatic cells from adult animals for gene editing and subsequent cloning allows the preservation and enhancement of superior traits from the parent directly in the offspring, while avoiding the loss of genetic gain that can occur through crossbreeding. However, primary cells generally more difficult to transfect and perform gene editing. To date, most related studies have used more vigorous fetal fibroblasts as donor cells, while using somatic cells from adult animals requires more post-editing screening efforts due to the low yield of edited cells. Here, we performed electroporation on adult bovine ear fibroblasts (BEFs) under various conditions such as electrical pulse settings, plasmid dosage, cell density, and concentration of electroporation buffer, and evaluated the transfect efficiency using flow cytometry analysis. We confirmed that the 270 V-10-10 program (270 V, 10 ms, 10 cycles) using 1.5 million cells and 5 µg of EGFP plasmid yielded the highest number of EGFP positive cells. Additionally, we used prime editor (PE) to edit the MSTN locus in BEFs. More importantly, we discovered that lowering the osmolarity of the electroporation buffer improves both electroporation and gene editing efficiency, which relate to the repression of cGAS-STING pathway. Our finding provides valuable references for using electroporation methods in adult bovine primary cell gene editing.
Collapse
Affiliation(s)
- Mingxuan Sheng
- Henan University Kaifeng China; Henan Province Livestock Genome Editing and Biobreeding Engineering Research Center, School of Life Sciences, Henan University, Kaifeng 475004 China.
| | - Yuequan Zheng
- Henan Province Livestock Genome Editing and Biobreeding Engineering Research Center, School of Life Sciences, Henan University, Kaifeng 475004 China.
| | - Kunlong Li
- Henan University Kaifeng China; Henan Province Livestock Genome Editing and Biobreeding Engineering Research Center, School of Life Sciences, Henan University, Kaifeng 475004 China.
| | - Chongyuan Ye
- Henan University Kaifeng China; Henan Province Livestock Genome Editing and Biobreeding Engineering Research Center, School of Life Sciences, Henan University, Kaifeng 475004 China.
| | - Gengsheng Cao
- Henan University Kaifeng China; Henan Province Livestock Genome Editing and Biobreeding Engineering Research Center, School of Life Sciences, Henan University, Kaifeng 475004 China.
| |
Collapse
|
31
|
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] [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
|
32
|
ZHU XI, HUANG KAI, KAO XIAOMING, TANG ZHAOHUI, GUO WENJIE, WU TIANCONG, LI QIURONG. Death domain-associated protein (Daxx) impairs colon cancer chemotherapy by inhibiting the cGAS-STING pathway. Oncol Res 2025; 33:1149-1159. [PMID: 40296918 PMCID: PMC12034003 DOI: 10.32604/or.2024.054930] [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: 06/11/2024] [Accepted: 10/11/2024] [Indexed: 04/30/2025] Open
Abstract
Background Colorectal cancer (CRC) holds the third position in global cancer prevalence mortality. Although chemotherapy is a conventional treatment, recent investigations have shed light on the therapeutic potential of the cGAS cyclic GMP-AMP synthase (cGAS)-stimulator of interferon genes (STING) pathway in CRC management. Despite the primary role of the death domain-associated protein (Daxx) in cellular apoptosis, its influence on the regulation of cGAS-STING activation remains elusive. Methods The Daxx degradation and speck formation were conducted using immunofluorescence and Western blotting. The Daxx knock-down and over-expression in CRC cells were performed to detect in vivo and in vitro migration, proliferation, cGAS-STING activation, and immune responses. Results Our study reveals that treatment with irinotecan (CPT-11) and oxaliplatin (OXA) significantly accelerated the Daxx degradation and diminished the formation of Daxx specks within the nucleus of CRC cells. Genetic elimination of Daxx enhanced the irinotecan and oxaliplatin-induced suppression of proliferation and migration in CRC cells, and overexpression of Daxx resulted in similar results. Mechanistically, Daxx overexpression reduced DNA damage repair by restraining homologous recombination (HR) over non-homologous end-joining (NHEJ), which suppressed TBK1 and IRF3 phosphorylation downstream of the cGAS-STING signal. In a murine model of CT-26 tumors, Daxx knockdown amplified the OXA-mediated tumor growth inhibition by promoting STING activation and immune responses. Conclusions Our findings show that the degradation of nuclear Daxx potentiates the cGAS-STING pathway, thereby bolstering the efficacy of chemotherapy.
Collapse
Affiliation(s)
- XI ZHU
- Research Institute of General Surgery, Jinling Hospital, Nanjing Medical University, Nanjing, 210002, China
- Research Institute of General Surgery, Nanjing Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210002, China
| | - KAI HUANG
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Science, Nanjing University, Nanjing, 210093, China
| | - XIAOMING KAO
- Research Institute of General Surgery, Nanjing Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210002, China
| | - ZHAOHUI TANG
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Science, Nanjing University, Nanjing, 210093, China
| | - WENJIE GUO
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Science, Nanjing University, Nanjing, 210093, China
| | - TIANCONG WU
- Department of Radiation Oncology, Nanjing Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210002, China
| | - QIURONG LI
- Research Institute of General Surgery, Jinling Hospital, Nanjing Medical University, Nanjing, 210002, China
- Research Institute of General Surgery, Nanjing Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210002, China
| |
Collapse
|
33
|
Wang XT, Zhu X, Lian ZH, Liu Q, Yan HH, Qiu Y, Ge XY. AUP1 and UBE2G2 complex targets STING signaling and regulates virus-induced innate immunity. mBio 2025:e0060225. [PMID: 40237449 DOI: 10.1128/mbio.00602-25] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2025] [Accepted: 03/18/2025] [Indexed: 04/18/2025] Open
Abstract
Stimulator of interferon genes (STING) is an endoplasmic reticulum (ER) signaling adaptor that is essential for the host immune response triggered by DNA pathogens. Precise regulation of STING is crucial for maintaining a balanced immune response and preventing harmful autoinflammation. Activation of STING requires its translocation from the ER to the Golgi apparatus. However, the mechanisms that maintain STING in its resting state remain largely unclear. Here, we find that deficiency of the ancient ubiquitous protein 1 (AUP1) causes spontaneous activation of STING and enhances the expression of type I interferons (IFNs) under resting conditions. Furthermore, deficiency of UBE2G2, a cofactor of AUP1, also promotes the abnormal activation of STING. AUP1 deficiency significantly enhances STING signaling induced by DNA virus, and AUP1 deficiency exhibits increased resistance to DNA virus infection in vitro and in vivo. Mechanistically, AUP1 may form a complex with UBE2G2 to interact with STING, preventing its exit from the ER membrane. Notably, infection with the RNA virus vesicular stomatitis virus (VSV) promotes the accumulation of lipid droplets (LDs) and AUP1 proteins. Additionally, AUP1 deficiency markedly inhibits the replication of VSV because AUP1 deficiency reduces lipid accumulation and alters the expression of lipid metabolism genes, such as carnitine palmitoyltransferase 1A (CPT1A), monoglyceride lipase (MGLL), and sterol regulatory element-binding transcription factor 1 (SREBF1). This study uncovers the essential roles of AUP1 in the STING signaling pathway and lipid metabolism pathway, highlighting its dual role in regulating virus replication.IMPORTANCEThe stimulator of interferon genes (STING) signaling cascade plays an essential role in coordinating innate immunity against DNA pathogens and autoimmunity. Precise regulation of the innate immune response is essential for maintaining homeostasis. In this study, we demonstrate that ancient ubiquitous protein 1 (AUP1) and UBE2G2 act as negative regulators of the innate immune response by targeting STING. Notably, AUP1 interacts with STING to retain STING in the endoplasmic reticulum (ER), preventing STING translocation and thereby limiting STING signaling in the resting state. In addition, deficiency of AUP1 markedly inhibits the replication of DNA virus and RNA virus. Our findings provide new insights into the regulation of STING signaling and confirm AUP1 has a dual role in regulating virus replication.
Collapse
Affiliation(s)
- Xin-Tao Wang
- Hunan Provincial Key Laboratory of Medical Virology, Institute of Pathogen Biology and Immunology, College of Biology, Hunan University, Changsha, Hunan, China
| | - Xi Zhu
- Hunan Provincial Key Laboratory of Medical Virology, Institute of Pathogen Biology and Immunology, College of Biology, Hunan University, Changsha, Hunan, China
| | - Zhong-Hao Lian
- Hunan Provincial Key Laboratory of Medical Virology, Institute of Pathogen Biology and Immunology, College of Biology, Hunan University, Changsha, Hunan, China
| | - Qian Liu
- Hunan Provincial Key Laboratory of Medical Virology, Institute of Pathogen Biology and Immunology, College of Biology, Hunan University, Changsha, Hunan, China
| | - Hui-Hui Yan
- Hunan Provincial Key Laboratory of Medical Virology, Institute of Pathogen Biology and Immunology, College of Biology, Hunan University, Changsha, Hunan, China
| | - Ye Qiu
- Hunan Provincial Key Laboratory of Medical Virology, Institute of Pathogen Biology and Immunology, College of Biology, Hunan University, Changsha, Hunan, China
| | - Xing-Yi Ge
- Hunan Provincial Key Laboratory of Medical Virology, Institute of Pathogen Biology and Immunology, College of Biology, Hunan University, Changsha, Hunan, China
| |
Collapse
|
34
|
Zhang X, Hao C, Li T, Gao W, Ren Y, Wang J, Zhang Y. Leptin attenuates diabetic cardiomyopathy-induced cardiac remodeling via regulating cGAS/STING signaling and Opa1-mediated mitochondrial fusion. Cell Signal 2025; 132:111805. [PMID: 40246132 DOI: 10.1016/j.cellsig.2025.111805] [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: 01/08/2025] [Revised: 03/11/2025] [Accepted: 04/09/2025] [Indexed: 04/19/2025]
Abstract
PURPOSE This investigation seeks to elucidate the contribution of leptin to the pathogenesis of diabetic cardiomyopathy (DCM). METHODS Mice were rendered diabetic through the administration of streptozotocin (STZ). Leptin was delivered via subcutaneously implanted osmotic pumps. Assessments of cardiac performance, hypertrophy, and fibrosis were conducted using echocardiography, Hematoxylin and Eosin (H&E), Wheat Germ Agglutinin (WGA), and Masson trichrome staining. Myocardial apoptosis and oxidative stress were quantified through TUNEL assay and biochemical markers of oxidative stress, including Malondialdehyde (MDA), 4-Hydroxynonenal (4-HNE), and 3-Nitrotyrosine (3NT). Mitochondrial structure was examined using Transmission Electron Microscopy (TEM). Primary neonatal cardiomyocytes were subjected to high glucose (HG) conditions. The fluorescent indicators MitoTracker Green and MitoSOX Red were employed to evaluate mitochondrial morphology and function within the cardiomyocytes. RESULTS Mice with diabetes displayed marked cardiac hypertrophy and fibrosis, as indicated by H&E, WGA, and Masson staining. The administration of leptin significantly mitigated the cardiac pathological manifestations in diabetic mice. Leptin increased the expression of Opa1 and enhanced mitochondrial fusion and function in cardiomyocytes exposed to HG. The cGAS/STING signaling pathway may serve as a pivotal intermediary for leptin to facilitate Opa1-driven mitochondrial fusion. CONCLUSIONS Leptin appears to safeguard against hyperglycemia-induced mitochondrial oxidative damage and DCM by modulating the cGAS/STING signaling cascade and Opa1-mediated mitochondrial fusion. These results propose that leptin could be a promising agent for promoting mitochondrial fusion and preventing diabetes-associated cardiac pathologies.
Collapse
Affiliation(s)
| | - Chunyuan Hao
- Cardiovascular Department, Xi'an No.1 Hospital, Xi'an, Shaanxi, China
| | - Tonghua Li
- Cardiovascular Department, Xi'an No.1 Hospital, Xi'an, Shaanxi, China
| | - Weihua Gao
- Cardiovascular Department, Xi'an No.1 Hospital, Xi'an, Shaanxi, China
| | - Yang Ren
- Cardiovascular Department, Xi'an No.1 Hospital, Xi'an, Shaanxi, China
| | - Junzhe Wang
- Cardiovascular Department, Xi'an No.1 Hospital, Xi'an, Shaanxi, China
| | - Yuyang Zhang
- Cardiovascular Department, Xi'an No.1 Hospital, Xi'an, Shaanxi, China.
| |
Collapse
|
35
|
Pan X, Zhao X, Lu Y, Xie P, Liu L, Chu X. Harnessing Nanomaterials for Enhanced DNA-Based Biosensing and Therapeutic Performance. Chembiochem 2025; 26:e202400936. [PMID: 39655520 DOI: 10.1002/cbic.202400936] [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: 11/16/2024] [Revised: 12/09/2024] [Accepted: 12/09/2024] [Indexed: 12/19/2024]
Abstract
The integration of nanomaterials with DNA-based systems has emerged as a transformative approach in biosensing and therapeutic applications. Unique features of DNA, like its programmability and specificity, complement the diverse functions of nanomaterials, leading to the creation of advanced systems for detecting biomarkers and delivering treatments. Here, we review the developments in DNA-nanomaterial conjugates, emphasizing their enhanced functionalities and potential across various biomedical applications. We first discuss the methodologies for synthesizing these conjugates, distinguishing between covalent and non-covalent interactions. We then categorize DNA-nanomaterials conjugates based on the properties of the DNA and nanomaterials involved, respectively. DNA probes are classified by their application into biosensing or therapeutic uses, and, several nanomaterials are highlighted by their recent progress in living biological. Finally, we discuss the current challenges and future prospects in this field, anticipating that significant progress in DNA-nanomaterial conjugates will greatly enhance precision medicine.
Collapse
Affiliation(s)
- Xumin Pan
- State Key Laboratory of Chemo/Bio-Sensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, 410082, Changsha, P. R. China
| | - Xiaoman Zhao
- State Key Laboratory of Chemo/Bio-Sensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, 410082, Changsha, P. R. China
| | - Yanhong Lu
- State Key Laboratory of Chemo/Bio-Sensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, 410082, Changsha, P. R. China
| | - Ping Xie
- State Key Laboratory of Chemo/Bio-Sensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, 410082, Changsha, P. R. China
| | - Lan Liu
- State Key Laboratory of Chemo/Bio-Sensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, 410082, Changsha, P. R. China
| | - Xia Chu
- State Key Laboratory of Chemo/Bio-Sensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, 410082, Changsha, P. R. China
| |
Collapse
|
36
|
Ali I, Xu F, Peng Q, Qiu J. The dilemma of nuclear mechanical forces in DNA damage and repair. Biochem Biophys Res Commun 2025; 758:151639. [PMID: 40121966 DOI: 10.1016/j.bbrc.2025.151639] [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: 02/12/2025] [Revised: 03/13/2025] [Accepted: 03/14/2025] [Indexed: 03/25/2025]
Abstract
Genomic stability, encompassing DNA damage and repair mechanisms, plays a pivotal role in the onset of diseases and the aging process. The stability of DNA is intricately linked to the chemical and mechanical forces exerted on chromatin, particularly within lamina-associated domains (LADs). Mechanical stress can induce DNA damage through the deformation and rupture of the nuclear envelope, leading to DNA bending and cleavage. However, DNA can evade such mechanical stress-induced damage by relocating away from the nuclear membrane, a process facilitated by the depletion of H3K9me3-marked heterochromatin and its cleavage from the lamina. When DNA double-stranded breaks occur, they prompt the rapid recruitment of Lamin B1 and the deposition of H3K9me3. Despite these insights, the precise mechanisms underlying DNA damage and repair under mechanical stress remain unclear. In this review, we explore the interplay between mechanical forces and the nuclear envelope in the context of DNA damage, elucidate the molecular pathways through which DNA escapes force-induced damage, and discuss the corresponding repair strategies involving the nuclear cytoskeleton. By summarizing the mechanisms of force-induced DNA damage and repair, we aim to underscore the potential for developing targeted therapeutic strategies to bolster genomic stability and alleviate the impacts of aging and disease.
Collapse
Affiliation(s)
- Iqra Ali
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, College of Bioengineering, Chongqing University, Chongqing, 400030, China
| | - Fangning Xu
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, College of Bioengineering, Chongqing University, Chongqing, 400030, China
| | - Qin Peng
- Institute of Systems and Physical Biology, Shenzhen Bay Laboratory, Shenzhen, 518132, China.
| | - Juhui Qiu
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, College of Bioengineering, Chongqing University, Chongqing, 400030, China.
| |
Collapse
|
37
|
Zhang L, Liu S, Zhao Q, Liu X, Zhang Q, Liu M, Zhao W. The role of ubiquitination and deubiquitination in the pathogenesis of non-alcoholic fatty liver disease. Front Immunol 2025; 16:1535362. [PMID: 40292292 PMCID: PMC12021615 DOI: 10.3389/fimmu.2025.1535362] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2024] [Accepted: 03/19/2025] [Indexed: 04/30/2025] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD) is one of the most common chronic liver diseases and is closely associated with metabolic abnormalities. The causes of NAFLD are exceedingly complicated, and it is known that a variety of signaling pathways, endoplasmic reticulum stress, and mitochondrial dysfunction play a role in the pathogenesis of NAFLD. Recent studies have shown that ubiquitination and deubiquitination are involved in the regulation of the NAFLD pathophysiology. Protein ubiquitination is a dynamic and diverse post-translational alteration that affects various cellular biological processes. Numerous disorders, including NAFLD, exhibit imbalances in ubiquitination and deubiquitination. To highlight the significance of this post-translational modification in the pathogenesis of NAFLD and to aid in the development of new therapeutic approaches for the disease, we will discuss the role of enzymes involved in the processes of ubiquitination and deubiquitination, specifically E3 ubiquitin ligases and deubiquitinating enzymes that are important in the regulation of NAFLD.
Collapse
Affiliation(s)
- Lihui Zhang
- The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, China
- Collaborative Innovation Center of Prevention and Treatment of Major Diseases by Chinese and Western Medicine, Zhengzhou, Henan, China
- Collaborative Innovation Center of Research and Development on the Whole Industry Chain of Yu-Yao, Zhengzhou, Henan, China
| | - Sutong Liu
- The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, China
- Collaborative Innovation Center of Prevention and Treatment of Major Diseases by Chinese and Western Medicine, Zhengzhou, Henan, China
- Collaborative Innovation Center of Research and Development on the Whole Industry Chain of Yu-Yao, Zhengzhou, Henan, China
| | - Qing Zhao
- The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, China
| | - Xiaoyan Liu
- The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, China
| | - Qiang Zhang
- The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, China
| | - Minghao Liu
- The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, China
- Collaborative Innovation Center of Prevention and Treatment of Major Diseases by Chinese and Western Medicine, Zhengzhou, Henan, China
- Collaborative Innovation Center of Research and Development on the Whole Industry Chain of Yu-Yao, Zhengzhou, Henan, China
| | - Wenxiao Zhao
- The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, China
- Collaborative Innovation Center of Prevention and Treatment of Major Diseases by Chinese and Western Medicine, Zhengzhou, Henan, China
- Collaborative Innovation Center of Research and Development on the Whole Industry Chain of Yu-Yao, Zhengzhou, Henan, China
| |
Collapse
|
38
|
Gharpure A, Sulpizio A, Loeffler JR, Fernández-Quintero ML, Tran AS, Lairson LL, Ward AB. Distinct oligomeric assemblies of STING induced by non-nucleotide agonists. Nat Commun 2025; 16:3440. [PMID: 40216780 PMCID: PMC11992164 DOI: 10.1038/s41467-025-58641-5] [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: 08/02/2024] [Accepted: 03/31/2025] [Indexed: 04/14/2025] Open
Abstract
STING plays essential roles coordinating innate immune responses to processes that range from pathogenic infection to genomic instability. Its adaptor function is activated by cyclic dinucleotide (CDN) secondary messengers originating from self (2'3'-cGAMP) or bacterial sources (3'3'-CDNs). Different classes of CDNs possess distinct binding modes, stabilizing STING's ligand-binding domain (LBD) in either a closed or open conformation. The closed conformation, induced by the endogenous ligand 2'3'-cGAMP, has been extensively studied using cryo-EM. However, significant questions remain regarding the structural basis of STING activation by open conformation-inducing ligands. Using cryo-EM, we investigate potential differences in conformational changes and oligomeric assemblies of STING for closed and open conformation-inducing synthetic agonists. While we observe a characteristic 180° rotation for both classes, the open-LBD inducing agonist diABZI-3 uniquely induces a quaternary structure reminiscent but distinct from the reported autoinhibited state of apo-STING. Additionally, we observe slower rates of activation for this ligand class in functional assays, which collectively suggests the existence of a potential additional regulatory mechanism for open conformation-inducing ligands that involves head-to-head interactions and restriction of curved oligomer formation. These observations have potential implications in the selection of an optimal class of STING agonist in the context of a defined therapeutic application.
Collapse
Affiliation(s)
- Anant Gharpure
- Department of Integrative Structural and Computational Biology, Scripps Research, La Jolla, CA, USA
| | - Ariana Sulpizio
- Department of Chemistry, Scripps Research, La Jolla, CA, USA
| | - Johannes R Loeffler
- Department of Integrative Structural and Computational Biology, Scripps Research, La Jolla, CA, USA
| | | | - Andy S Tran
- Department of Integrative Structural and Computational Biology, Scripps Research, La Jolla, CA, USA
| | - Luke L Lairson
- Department of Chemistry, Scripps Research, La Jolla, CA, USA.
| | - Andrew B Ward
- Department of Integrative Structural and Computational Biology, Scripps Research, La Jolla, CA, USA.
| |
Collapse
|
39
|
Paolì A, Sadeghi S, Battistello G, Carpanese V, Checchetto V. In silico pan-cancer analysis of VRAC subunits and their prognostic roles in human cancers. Sci Rep 2025; 15:12388. [PMID: 40216864 PMCID: PMC11992229 DOI: 10.1038/s41598-025-97078-0] [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: 07/26/2024] [Accepted: 04/02/2025] [Indexed: 04/14/2025] Open
Abstract
The study focuses on the VRAC channel and its significant roles in cancer development. It addresses a research gap by conducting a pan-cancer analysis with multi-omics bioinformatics tools, integrating data from the Human Protein Atlas (HPA) and Genotype-Tissue Expression (GTEx) datasets to examine mRNA expression patterns of its Leucine Rich Repeat Containing 8 (LRRC8) subunits in various tissues and cancers. The study links variations in LRRC8s expression with patient outcomes and includes analyses of DNA and RNA methylation. The study reveals significant correlations between LRRC8s expression and immune cell infiltration, as well as a positive association with cancer-associated fibroblasts and key immune regulators such as major histocompatibility complex (MHCs) and chemokines. Furthermore, the research suggests that LRRC8s are involved in cancer-signalling pathways, which may offer new therapeutic targets. Additionally, a drug sensitivity analysis shows that LRRC8 subunits affect drug responses differently, supporting the use of personalized therapeutic strategies. In conclusion, the study emphasizes the significance of VRAC subunits in cancer biology and suggests their potential as biomarkers and targets in cancer immunotherapy and personalized medicine.
Collapse
Affiliation(s)
| | - Soha Sadeghi
- Department of Biology, University of Padova, Padua, Italy
| | | | | | | |
Collapse
|
40
|
Li T, Peng S, Zhou Y, Zhang C, Feng G, Yu Z, Xu Y, Quan M, Wang W, Song H. A novel STING1-activating mutation is identified in a patient with childhood-onset systemic lupus erythematosus. Clin Immunol 2025; 276:110493. [PMID: 40222637 DOI: 10.1016/j.clim.2025.110493] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2025] [Revised: 04/06/2025] [Accepted: 04/07/2025] [Indexed: 04/15/2025]
Abstract
Gain-of-function variants in stimulator of interferon genes (STING1) are known to cause STING-associated vasculopathy with onset in infancy (SAVI), a disorder characterized by cutaneous vasculopathy, interstitial lung disease (ILD), and systemic inflammation. Here, we report a novel STING1 N188H variant in a patient who met the classification criteria for systemic lupus erythematosus (SLE) but lacked typical SAVI features. In vitro assays demonstrated that the N188H variant drives constitutive STING activation and enhances type I interferon signaling. Consistent with this, the patient exhibited elevated interferon-stimulated genes (ISGs) expression, and RNA sequencing confirmed significant upregulation of type I IFN signaling compared to healthy controls. Our findings expand the molecular spectrum of STING-associated disease.
Collapse
Affiliation(s)
- Ting Li
- Department of Pediatrics, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Siming Peng
- Department of Pediatrics, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Yu Zhou
- Department of Pediatrics, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Caihui Zhang
- Department of Pediatrics, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Gexuan Feng
- Department of Pediatrics, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Zhongxun Yu
- Department of Pediatrics, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Yiwen Xu
- Department of Pediatrics, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Meiying Quan
- Department of Pediatrics, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Wei Wang
- Department of Pediatrics, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China.
| | - Hongmei Song
- Department of Pediatrics, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China.
| |
Collapse
|
41
|
Yu K, Yang S, Song H, Sun Z, Wang K, Zhu Y, Yang C, Hao R, Cao Y. High-Resolution Tracking of Aging-Related Small Molecules: Bridging Pollutant Exposure, Brain Aging Mechanisms, and Detection Innovations. BIOSENSORS 2025; 15:242. [PMID: 40277555 PMCID: PMC12024821 DOI: 10.3390/bios15040242] [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: 02/09/2025] [Revised: 03/23/2025] [Accepted: 03/26/2025] [Indexed: 04/26/2025]
Abstract
Brain aging is a complex process regulated by genetic, environmental, and metabolic factors, and increasing evidence suggests that environmental pollutants can significantly accelerate this process by interfering with oxidative stress, neuroinflammation, and mitochondrial function-related signaling pathways. Traditional studies have focused on the direct damage of pollutants on macromolecules (e.g., proteins, DNA), while the central role of senescence-associated small molecules (e.g., ROS, PGE2, lactate) in early regulatory mechanisms has been long neglected. In this study, we innovatively proposed a cascade framework of "small molecule metabolic imbalance-signaling pathway dysregulation-macromolecule collapse", which reveals that pollutants exacerbate the dynamics of brain aging through activation of NLRP3 inflammatory vesicles and inhibition of HIF-1α. Meanwhile, to address the technical bottleneck of small molecule spatiotemporal dynamics monitoring, this paper systematically reviews the cutting-edge detection tools such as electrochemical sensors, genetically encoded fluorescent probes and antioxidant quantum dots (AQDs). Among them, AQDs show unique advantages in real-time monitoring of ROS fluctuations and intervention of oxidative damage by virtue of their ultra-high specific surface area, controllable surface modification, and free radical scavenging ability. By integrating multimodal detection techniques and mechanism studies, this work provides a new perspective for analyzing pollutant-induced brain aging and lays a methodological foundation for early intervention strategies based on small molecule metabolic networks.
Collapse
Affiliation(s)
- Keying Yu
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing 100069, China; (K.Y.); (K.W.)
- Beijing Key Laboratory of Environment and Aging, Capital Medical University, Beijing 100069, China
| | - Sirui Yang
- School of Basic Medical Sciences, Capital Medical University, Beijing 100069, China; (S.Y.); (H.S.); (Z.S.); (Y.Z.)
| | - Hongxu Song
- School of Basic Medical Sciences, Capital Medical University, Beijing 100069, China; (S.Y.); (H.S.); (Z.S.); (Y.Z.)
| | - Zhou Sun
- School of Basic Medical Sciences, Capital Medical University, Beijing 100069, China; (S.Y.); (H.S.); (Z.S.); (Y.Z.)
| | - Kaichao Wang
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing 100069, China; (K.Y.); (K.W.)
- Beijing Key Laboratory of Environment and Aging, Capital Medical University, Beijing 100069, China
| | - Yuqi Zhu
- School of Basic Medical Sciences, Capital Medical University, Beijing 100069, China; (S.Y.); (H.S.); (Z.S.); (Y.Z.)
| | - Chengkai Yang
- Beijing Friendship Hospital, Capital Medical University, Beijing 100069, China;
| | - Rongzhang Hao
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing 100069, China; (K.Y.); (K.W.)
- Beijing Key Laboratory of Environment and Aging, Capital Medical University, Beijing 100069, China
| | - Yuanyuan Cao
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing 100069, China; (K.Y.); (K.W.)
- Beijing Key Laboratory of Environment and Aging, Capital Medical University, Beijing 100069, China
| |
Collapse
|
42
|
Wang W, Zhai Y, Yang X, Ye L, Lu G, Shi X, Zhai G. Effective design of therapeutic nanovaccines based on tumor neoantigens. J Control Release 2025; 380:17-35. [PMID: 39892648 DOI: 10.1016/j.jconrel.2025.01.078] [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: 09/05/2024] [Revised: 01/17/2025] [Accepted: 01/26/2025] [Indexed: 02/04/2025]
Abstract
Neoantigen vaccines are among the most potent immunotherapies for personalized cancer treatment. Therapeutic vaccines containing tumor-specific neoantigens that elicit specific T cell responses offer the potential for long-term clinical benefits to cancer patients. Unlike immune-checkpoint inhibitors (ICIs), which rely on pre-existing specific T cell responses, personalized neoantigen vaccines not only promote existing specific T cell responses but importantly stimulate the generation of neoantigen-specific T cells, leading to the establishment of a persistent specific memory T cell pool. The review discusses the current state of clinical research on neoantigen nanovaccines, focusing on the application of vectors, adjuvants, and combinational strategies to address a range of challenges and optimize therapeutic outcomes.
Collapse
Affiliation(s)
- Weilin Wang
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, China
| | - Yujia Zhai
- Department of Pharmaceutics and Pharmaceutical Chemistry, University of Utah, Salt Lake City, UT 84124, United States of America
| | - Xiaoye Yang
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, China
| | - Lei Ye
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, China
| | - Guoliang Lu
- Auckland Cancer Society Research Centre, Faculty of Medical and Health Sciences, The University of Auckland, Private Bag 92019, Auckland 1142, New Zealand; Maurice Wilkins Centre, University of Auckland, Private Bag 92019, Auckland 1142, New Zealand
| | - Xiaoqun Shi
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, China.
| | - Guangxi Zhai
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, China.
| |
Collapse
|
43
|
Srivastava SP, Kopasz-Gemmen O, Kunamneni A, Thurnman A, Ozukan E, Swaroop V, Yoshida S, Hong S, Inoki K. AMPK is dispensable for physiological podocyte and glomerular functions but prevents glomerular fibrosis in experimental diabetes. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.04.07.647592. [PMID: 40291739 PMCID: PMC12026990 DOI: 10.1101/2025.04.07.647592] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/30/2025]
Abstract
AMP-activated protein kinase (AMPK) has been postulated to be crucial in regulating various renal physiology and pathophysiology processes, including energy metabolism, ion and water transport, inflammation, and hypertrophy. However, the specific roles of AMPK in the podocyte, a cell critical for maintaining glomerular filtration, have not been fully explored using genetic model animals. In this study, we generated mice lacking both AMPK α1 and α2 catalytic subunits in glomerular podocytes (pmut). Our findings revealed that, surprisingly, AMPK is dispensable for normal podocyte function. These knockout mice could live as long as their wild-type littermates without showing any pathological alterations in their glomeruli or glomerular function at two years of age. However, under type 1 diabetic conditions, the diabetic pmut mice exhibited increased lipid and collagen accumulation and an elevated expression of mesenchymal proteins in their glomeruli. They also showed more significant albuminuria compared to control diabetic mice. Under high glucose culture conditions, glomeruli isolated from pmut mice demonstrated a reduced expression of mitochondrial genes (e.g., Ndufv2) and increased leakage of mitochondrial components. Additionally, there was heightened expression of genes associated with nucleotide sensing and pro-inflammatory pathways (including mb21d2, IL-1 beta, and NF-kB). These observations suggest that while AMPK is not necessary for podocyte function in healthy kidneys, it is crucial for preventing glomerular fibrosis resulting from lipotoxicity and inflammation under diabetic conditions.
Collapse
|
44
|
Simchoni N, Koide S, Likhite M, Kuchitsu Y, Kadirvel S, Law CS, Elicker BM, Kurra S, Wong MMK, Yuan B, Grossi A, Laxer RM, Volpi S, Dissanayake D, Taguchi T, Beck DB, Vogel TP, Shum AK. The common HAQ STING allele prevents clinical penetrance of COPA syndrome. J Exp Med 2025; 222:e20242179. [PMID: 40014299 PMCID: PMC11867111 DOI: 10.1084/jem.20242179] [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: 11/18/2024] [Revised: 12/20/2024] [Accepted: 12/23/2024] [Indexed: 02/28/2025] Open
Abstract
COPA syndrome, an autosomal-dominant inborn error of immunity, is nonpenetrant in ∼20% of individuals, with no known mediators of protection. Recent studies implicate STING in the pathogenesis of COPA syndrome. We show that the common HAQ STING allele mediates complete clinical protection. We sequenced 35 individuals with COPA mutations, 26 affected patients and 9 unaffected carriers, finding HAQ STING co-segregation with clinical nonpenetrance. Exome sequencing identified only the mutations comprising HAQ STING as variants shared by unaffected carriers and absent in patients. Experimentally, we found that HAQ STING acts dominantly to dampen COPA-dependent STING signaling. Expressing HAQ STING in patient cells rescued the molecular phenotype of COPA syndrome. Our study is the first report of a common and well-tolerated allele mediating complete clinical protection from a severe genetic disorder. Our findings redefine the diagnostic criteria for COPA syndrome, expose functional differences among STING alleles with broad scientific and clinical implications, and reveal a potential universal gene therapy approach for patients.
Collapse
Affiliation(s)
- Noa Simchoni
- Division of Pulmonary, Department of Medicine, University of California, San Francisco, San Francisco, CA, USA
| | - Shogo Koide
- Department of Integrative Life Sciences, Graduate School of Life Sciences, Tohoku University, Sendai, Japan
| | - Maryel Likhite
- Center for Human Genetics and Genomics, NYU Grossman School of Medicine, New York, NY, USA
| | - Yoshihiko Kuchitsu
- Department of Integrative Life Sciences, Graduate School of Life Sciences, Tohoku University, Sendai, Japan
| | | | - Christopher S. Law
- Division of Pulmonary, Department of Medicine, University of California, San Francisco, San Francisco, CA, USA
| | - Brett M. Elicker
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco, CA, USA
| | - Santosh Kurra
- Division of Pulmonary, Department of Medicine, University of California, San Francisco, San Francisco, CA, USA
| | - Margaret Mei-Kay Wong
- Center for Human Genetics and Genomics, NYU Grossman School of Medicine, New York, NY, USA
| | - Bo Yuan
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, TX, USA
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
| | - Alice Grossi
- Laboratorio Genetica e Genomica Delle Malattie Rare, Istituto Giannina Gaslini, Genoa, Italy
| | - Ronald M. Laxer
- Division of Rheumatology, Department of Paediatrics, The Hospital for Sick Children, University of Toronto, Toronto, Canada
- Division of Rheumatology, Department of Medicine, St. Michael’s Hospital, Toronto, Canada
| | - Stefano Volpi
- Unità Operativa Complessa Reumatologia e Malattie Autoinfiammatorie, Istituto di Ricovero e Cura a Carattere Scientifico Istituto Giannina Gaslini, Genoa, Italy
- Dipartimento Di Neuroscienze, Riabilitazione, Oftalmologia, Genetica e Scienze Materno-Infantili, Università Degli Studi Di Genova, Genoa, Italy
| | - Dilan Dissanayake
- Division of Rheumatology, Department of Paediatrics, The Hospital for Sick Children, University of Toronto, Toronto, Canada
| | - Tomohiko Taguchi
- Department of Integrative Life Sciences, Graduate School of Life Sciences, Tohoku University, Sendai, Japan
| | - David B. Beck
- Center for Human Genetics and Genomics, NYU Grossman School of Medicine, New York, NY, USA
- Division of Rheumatology, Department of Medicine, NYU Grossman School of Medicine, New York, NY, USA
| | - Tiphanie P. Vogel
- Division of Rheumatology, Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA
- Center for Human Immunobiology, Texas Children’s Hospital, Houston, TX, USA
| | - Anthony K. Shum
- Division of Pulmonary, Department of Medicine, University of California, San Francisco, San Francisco, CA, USA
- Cardiovascular Research Institute, University of California, San Francisco, San Francisco, CA, USA
| |
Collapse
|
45
|
Guilbaud L, Chen C, Domingues I, Kavungere EK, Marotti V, Yagoubi H, Zhang W, Malfanti A, Beloqui A. Oral Lipid-Based Nanomedicine for the Inhibition of the cGAS-STING Pathway in Inflammatory Bowel Disease Treatment. Mol Pharm 2025; 22:2108-2121. [PMID: 40032274 PMCID: PMC11979890 DOI: 10.1021/acs.molpharmaceut.4c01297] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Revised: 02/17/2025] [Accepted: 02/18/2025] [Indexed: 03/05/2025]
Abstract
Harnessing the effect of the cyclic GMP-AMP Synthase-STimulator of INterferon Genes (cGAS-STING) signaling pathway has emerged as a promising approach to developing novel strategies for the oral treatment of inflammatory bowel disease (IBD). In this work, we screened different cGAS-STING inhibitors in vitro in murine macrophages. Then, we encapsulated the cGAS-STING inhibitor H-151 within lipid nanocapsules (LNCs), owing to their inherent ability to induce the secretion of glucagon-like peptide 2 (GLP-2), a re-epithelizing peptide, upon oral administration. We demonstrated that our formulation (LNC(H-151)) could induce GLP-2 secretion and selectively target the cGAS-STING pathway and its downstream key markers (including TBK1 and pTBK1) while reducing the expression of pro-inflammatory cytokines associated with the cGAS-STING pathway (TNF-α and CXCL10) in murine macrophages. In an in vivo acute dextran sodium sulfate (DSS)-induced colitis mouse model, the oral administration of LNC(H-151) significantly reduced pro-inflammatory cytokines to levels comparable to the CTRL Healthy group while promoting mucosal healing. The therapeutic potential of this scalable and cost-effective nanomedicine warrants further investigation as an alternative for the oral treatment of IBD.
Collapse
Affiliation(s)
- Léo Guilbaud
- Louvain
Drug Research Institute, Advanced Drug Delivery and Biomaterials, UCLouvain, Université Catholique de Louvain, 1200 Brussels, Belgium
| | - Cheng Chen
- Louvain
Drug Research Institute, Advanced Drug Delivery and Biomaterials, UCLouvain, Université Catholique de Louvain, 1200 Brussels, Belgium
| | - Inês Domingues
- Louvain
Drug Research Institute, Advanced Drug Delivery and Biomaterials, UCLouvain, Université Catholique de Louvain, 1200 Brussels, Belgium
| | - Espoir K. Kavungere
- Louvain
Drug Research Institute, Advanced Drug Delivery and Biomaterials, UCLouvain, Université Catholique de Louvain, 1200 Brussels, Belgium
| | - Valentina Marotti
- Louvain
Drug Research Institute, Advanced Drug Delivery and Biomaterials, UCLouvain, Université Catholique de Louvain, 1200 Brussels, Belgium
| | - Hafsa Yagoubi
- Louvain
Drug Research Institute, Advanced Drug Delivery and Biomaterials, UCLouvain, Université Catholique de Louvain, 1200 Brussels, Belgium
| | - Wunan Zhang
- Louvain
Drug Research Institute, Advanced Drug Delivery and Biomaterials, UCLouvain, Université Catholique de Louvain, 1200 Brussels, Belgium
| | - Alessio Malfanti
- Department
of Pharmaceutical and Pharmacological Sciences, University of Padova, Via F. Marzolo 5, 35131 Padova, Italy
| | - Ana Beloqui
- Louvain
Drug Research Institute, Advanced Drug Delivery and Biomaterials, UCLouvain, Université Catholique de Louvain, 1200 Brussels, Belgium
- WEL
Research Institute, Avenue
Pasteur, 6, 1300 Wavre, Belgium
| |
Collapse
|
46
|
Gao F, Zheng Z, Liu X, Li J. CMPK2 promotes microglial activation through the cGAS-STING pathway in the neuroinflammatory mechanism. Sci Rep 2025; 15:11807. [PMID: 40189684 PMCID: PMC11973145 DOI: 10.1038/s41598-025-97232-8] [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: 09/23/2024] [Accepted: 04/03/2025] [Indexed: 04/09/2025] Open
Abstract
The activation of microglia and the resulting neuroinflammation play crucial regulatory roles in the pathogenesis and progression of neurological diseases, although the specific mechanisms remain incompletely understood. Cytidine monophosphate kinase 2 (CMPK2) is a key mitochondrial nucleotide kinase involved in cellular energy metabolism and nucleotide synthesis. Recent studies suggest that CMPK2 plays a role in microglial-mediated neuroinflammation; however, its specific impact on microglial activation remains unclear. In this study, we hypothesize that CMPK2 promotes microglial-mediated neuroinflammation by activating the cGAS-STING signaling pathway. To investigate this mechanism, we employed lipopolysaccharide (LPS)-treated microglial cells to investigate the detailed mechanisms by which CMPK2 regulates neuroinflammation. Our experimental results indicate that in the BV2 and mouse primary microglial neuroinflammation model, both CMPK2 protein and transcript levels were significantly elevated, accompanied by microglial activation phenotypes such as increased cell size, shortened processes, transformation to round or rod-like shapes, and elevated CD40 expression. Concurrently, there was an increase in pro-inflammatory cytokine levels and a decrease in anti-inflammatory cytokine levels. Further investigation revealed that in the microglial, the expression of cGAS and STING was elevated, along with an increase in oxidative products and inflammatory responses. CMA stimulation further intensified these changes, while cGAS knockdown mitigated them. Finally, we demonstrated that cGAS knockdown inhibited the oxidative stress, cell activation-related changes, and neuroinflammatory responses induced by CMPK2 overexpression in the BV2 neuroinflammation model. Molecular docking experiments showed that CMPK2 stably binds to cGAS at the protein level. These findings suggest that the cGAS-STING pathway mediates CMPK2-induced microglial activation. In summary, our study demonstrates that LPS-induced CMPK2 overactivity promotes microglial activation and neuroinflammatory through the cGAS-STING pathway.
Collapse
Affiliation(s)
- Feng Gao
- Department of Neurosurgery, Xingtai People's Hospital, Xingtai, Hebei, China.
| | - Zijian Zheng
- Department of Neurosurgery, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China.
| | - Xinjie Liu
- Department of Neurosurgery, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Jianwei Li
- Department of Neurosurgery, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| |
Collapse
|
47
|
Lee AR, Min HK, Lee SY, Jeon SB, Lee CR, Kim TH, Park JH, La Cho M. Remdesivir alleviates joint damage in collagen-induced arthritis and inhibits inflammatory cell death of RA synovial fibroblasts. Immunol Lett 2025; 275:107009. [PMID: 40189155 DOI: 10.1016/j.imlet.2025.107009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Revised: 03/18/2025] [Accepted: 03/31/2025] [Indexed: 04/24/2025]
Abstract
BACKGROUND The antiviral agent, remdesivir, is adenosine analogue which is currently also used as anti-coronavirus disease 2019. Remdesivir also had anti-inflammatory effect which reduced pro-inflammatory cytokine production, and inhibition of the cyclic GMP-AMP synthase-STING pathway. METHODS We evaluated the antiarthritic effects of remdesivir in a mouse model of High-fat diet (HFD) collagen-induced arthritis (CIA) and in fibroblast-like synoviocytes from patients with RA. Type II collagen was administered to DBA/1J mice to induce CIA. Vehicle or remdesivir was injected subcutaneously three times a week. During 7 weeks of treatment, the arthritis score and incidence were evaluated twice a week. Flow cytometry and confocal imaging were used to evaluate CD4 + T cells in the spleen. FLSs from patients with RA were stimulated in vitro with remdesivir and tumor necrosis factor (TNF)-α, and western blotting was used to measure the expression of STING and necroptosis-related markers. RESULTS Remdesivir administration suppressed the incidence and progression of arthritis in mice with CIA. Histological analysis revealed lower inflammation and cartilage damage scores in remdesivir-treated than in vehicle groups. Interleukin (IL)-17 + CD4 + T-cell differentiation was inhibited in the remdesivir-treated group. Furthermore, IL-17/-6/-1β, monocyte chemoattractant protein -1, and TNF-α expression was reduced in the remdesivir group. In vitro, remdesivir suppressed the expression of STING, nuclear factor-κB, RIPK3, and phosphorylated MLKL in RA-FLSs under TNF-α stimulation. CONCLUSIONS The antiviral agent remdesivir suppressed arthritis by regulating Th cell differentiation, pro-inflammatory cytokine expression, the STING pathway, and necroptosis.
Collapse
Affiliation(s)
- A Ram Lee
- Lab of Translational ImmunoMedicine, Catholic Research Institute of Medical Science, College of Medicine, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea; Department of Pathology, College of Medicine, The Catholic University of Korea, Seoul 06591, South Korea
| | - Hong Ki Min
- Division of Rheumatology, Department of Internal Medicine, Konkuk University Medical Center, Seoul 05030, Republic of Korea
| | - Seon-Yeong Lee
- Lab of Translational ImmunoMedicine, Catholic Research Institute of Medical Science, College of Medicine, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea; Department of Pathology, College of Medicine, The Catholic University of Korea, Seoul 06591, South Korea
| | - Su Been Jeon
- Lab of Translational ImmunoMedicine, Catholic Research Institute of Medical Science, College of Medicine, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea; Department of Pathology, College of Medicine, The Catholic University of Korea, Seoul 06591, South Korea; Department of Biomedicine & Health Sciences, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea
| | - Chae Rim Lee
- Lab of Translational ImmunoMedicine, Catholic Research Institute of Medical Science, College of Medicine, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea; Department of Pathology, College of Medicine, The Catholic University of Korea, Seoul 06591, South Korea; Department of Biomedicine & Health Sciences, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea
| | - Tae Ho Kim
- Lab of Translational ImmunoMedicine, Catholic Research Institute of Medical Science, College of Medicine, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea; Department of Pathology, College of Medicine, The Catholic University of Korea, Seoul 06591, South Korea; Department of Biomedicine & Health Sciences, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea
| | - Jin Hyung Park
- Lab of Translational ImmunoMedicine, Catholic Research Institute of Medical Science, College of Medicine, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea; Department of Pathology, College of Medicine, The Catholic University of Korea, Seoul 06591, South Korea; Department of Biomedicine & Health Sciences, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea
| | - Mi- La Cho
- Lab of Translational ImmunoMedicine, Catholic Research Institute of Medical Science, College of Medicine, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea; Department of Pathology, College of Medicine, The Catholic University of Korea, Seoul 06591, South Korea.
| |
Collapse
|
48
|
Cancado de Faria R, Silva L, Teodoro-Castro B, McCommis KS, Shashkova EV, Gonzalo S. A non-canonical cGAS-STING pathway drives cellular and organismal aging. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.04.03.645994. [PMID: 40236012 PMCID: PMC11996560 DOI: 10.1101/2025.04.03.645994] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 04/17/2025]
Abstract
Accumulation of cytosolic DNA has emerged as a hallmark of aging, inducing sterile inflammation. STING (Stimulator of Interferon Genes) protein translates the sensing of cytosolic DNA by cGAS (cyclic-GMP-AMP synthase) into an inflammatory response. However, the molecular mechanisms whereby cytosolic DNA-induced cGAS-STING pathway leads to aging remain poorly understood. We show that STING does not follow the canonical pathway of activation in human fibroblasts passaged (aging) in culture, senescent fibroblasts, or progeria fibroblasts (from Hutchinson Gilford Progeria Syndrome patients). Despite cytosolic DNA buildup, features of the canonical cGAS-STING pathway like increased cGAMP production, STING phosphorylation, and STING trafficking to perinuclear compartment are not observed in progeria/senescent/aging fibroblasts. Instead, STING localizes at endoplasmic reticulum, nuclear envelope, and chromatin. Despite the non-conventional STING behavior, aging/senescent/progeria cells activate inflammatory programs such as the senescence-associated secretory phenotype (SASP) and the interferon (IFN) response, in a cGAS and STING-dependent manner, revealing a non-canonical pathway in aging. Importantly, progeria/aging/senescent cells are hindered in their ability to activate the canonical cGAS-STING pathway with synthetic DNA, compared to young cells. This deficiency is rescued by activating vitamin D receptor signaling, unveiling new mechanisms regulating the cGAS-STING pathway in aging. Significantly, in HGPS, inhibition of the non-canonical cGAS-STING pathway ameliorates cellular hallmarks of aging, reduces tissue degeneration, and extends the lifespan of progeria mice. Our study reveals that a new feature of aging is the progressively reduced ability to activate the canonical cGAS-STING pathway in response to cytosolic DNA, triggering instead a non-canonical pathway that drives senescence/aging phenotypes. Significance Statement Our study provides novel insights into the mechanisms driving sterile inflammation in aging and progeria. We reveal a previously unrecognized characteristic of aging cells: the progressive loss of ability to activate the canonical response to foreign or self-DNA at the cytoplasm. Instead, aging, senescent, and progeria cells activate inflammatory programs via a non-conventional pathway driven by cGAS and the adaptor protein STING. Importantly, pharmacological inhibition of the non-canonical cGAS-STING pathway ameliorates cellular, tissue and organismal decline in a devastating accelerated aging disease (Hutchinson Gilford Progeria Syndrome), highlighting it as a promising therapeutic target for age-related pathologies.
Collapse
|
49
|
Pan X, Zhu R, Pei J, Zhang L. Lycopene: A potent antioxidant to alleviate kidney disease. Int Immunopharmacol 2025; 151:114363. [PMID: 40022820 DOI: 10.1016/j.intimp.2025.114363] [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: 01/05/2025] [Revised: 02/19/2025] [Accepted: 02/21/2025] [Indexed: 03/04/2025]
Abstract
Kidney disease is a major public health problem worldwide and one of the common causes of death in patients. How to effectively treat or alleviate renal diseases has not only attracted the attention of a large number of scholars, but is likewise a great challenge. Oxidative stress is the main mechanism leading to kidney injury. Natural antioxidant substances not only have efficient antioxidant capacity, but also have the advantage of high safety and low side effects. Lycopene is a naturally occurring carotenoid found mainly in tomatoes or tomato products. Epidemiologic investigations have shown that lycopene has potent antioxidant properties, scavenges reactive substances, and has a protective role in kidney disease. This paper summarizes the biochemical properties and antioxidant mechanisms of lycopene in the context of animal experiments and clinical studies of lycopene in renal diseases. We found that lycopene exerts its protective effects against kidney injury mainly through anti-oxidative stress and anti-inflammatory effects. Meanwhile, lycopene has been found to reduce the incidence of Chronic Kidney Disease (CKD) as well as mortality in some CKD patients. In addition to this, lycopene intake is negatively correlated with the incidence of kidney cancer and also mitigates the nephrotoxic damage of cisplatin. Therefore, lycopene has a promising application in the treatment of kidney diseases. However, there are relatively few clinical studies on lycopene in renal diseases, and subsequent research studies in large-scale populations are still needed to determine the value of lycopene in renal diseases even further.
Collapse
Affiliation(s)
- Xingyu Pan
- Department of Nursing, Affiliated Hospital of Zunyi Medical University, Zunyi 563100, China; Nursing School of Zunyi Medical University, Zunyi 563100, China
| | - Rong Zhu
- Department of Pediatric surgrey, Guizhou Provincial People's Hospital, Guiyang 550000, China
| | - Jun Pei
- Department of Pediatric surgrey, Guizhou Provincial People's Hospital, Guiyang 550000, China.
| | - Li Zhang
- Department of Nursing, Affiliated Hospital of Zunyi Medical University, Zunyi 563100, China; Nursing School of Zunyi Medical University, Zunyi 563100, China.
| |
Collapse
|
50
|
Stojanovic L, Abbotts R, Tripathi K, Coon CM, Rajendran S, Farid EA, Hostetter G, Guarnieri JW, Wallace DC, Liu S, Wan J, Calendo G, Marker R, Gohari Z, Inayatullah MMA, Tiwari VK, Kader T, Santagata S, Drapkin R, Kommoss S, Pfisterer J, Konecny GE, Coopergard R, Issa JPJ, Winterhoff BJN, Topper MJ, Sandusky GE, Miller KD, Baylin SB, Nephew KP, Rassool FV. ZNFX1 Functions as a Master Regulator of Epigenetically Induced Pathogen Mimicry and Inflammasome Signaling in Cancer. Cancer Res 2025; 85:1183-1198. [PMID: 39804147 PMCID: PMC11968244 DOI: 10.1158/0008-5472.can-24-1286] [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: 04/18/2024] [Revised: 10/18/2024] [Accepted: 01/07/2025] [Indexed: 01/22/2025]
Abstract
DNA methyltransferase (DNMT) and PARP inhibitors induce a stimulator of IFN gene-dependent pathogen mimicry response (PMR) in ovarian and other cancers. In this study, we showed that combining DNMT and PARP inhibitors upregulates expression of the nucleic acid sensor NFX1-type zinc finger-containing 1 (ZNFX1) protein. ZNFX1 mediated the induction of PMR in mitochondria, serving as a gateway for stimulator of IFN gene-dependent IFN/inflammasome signaling. Loss of ZNFX1 in ovarian cancer cells promoted proliferation and spheroid formation in vitro and tumor growth in vivo. In patient ovarian cancer databases, expression of ZNFX1 was elevated in advanced stage disease, and ZNFX1 expression alone significantly correlated with an increase in overall survival in a phase III trial for patients with therapy-resistant ovarian cancer receiving bevacizumab in combination with chemotherapy. RNA sequencing revealed an association between inflammasome signaling through ZNFX1 and abnormal vasculogenesis. Together, this study identified that ZNFX1 is a tumor suppressor that controls PMR signaling through mitochondria and may serve as a biomarker to facilitate personalized therapy in patients with ovarian cancer. Significance: DNMT and PARP inhibitors induce a nucleic acid sensor, ZNFX1, that serves as a mitochondrial gateway to STING-dependent inflammasome signaling with tumor suppressor properties in ovarian cancer.
Collapse
Affiliation(s)
- Lora Stojanovic
- University of Maryland Marlene and Stewart Greenebaum Comprehensive Cancer Center, Baltimore, MD 21201, USA
- Division of Translational Radiation Sciences, Department of Radiation Oncology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Rachel Abbotts
- University of Maryland Marlene and Stewart Greenebaum Comprehensive Cancer Center, Baltimore, MD 21201, USA
- Division of Translational Radiation Sciences, Department of Radiation Oncology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Kaushlendra Tripathi
- University of Maryland Marlene and Stewart Greenebaum Comprehensive Cancer Center, Baltimore, MD 21201, USA
- Division of Translational Radiation Sciences, Department of Radiation Oncology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Collin M. Coon
- Medical Sciences Program, Indiana University School of Medicine-Bloomington, Bloomington, IN 47405, USA
| | - Saranya Rajendran
- Medical Sciences Program, Indiana University School of Medicine-Bloomington, Bloomington, IN 47405, USA
| | - Elnaz Abbasi Farid
- Medical Sciences Program, Indiana University School of Medicine-Bloomington, Bloomington, IN 47405, USA
| | | | - Joseph W. Guarnieri
- Center for Mitochondrial and Epigenomic Medicine, Division of Human Genetics, The Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Douglas C. Wallace
- Center for Mitochondrial and Epigenomic Medicine, Division of Human Genetics, The Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
- Department of Pediatrics, Division of Human Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Sheng Liu
- Department of Medical and Molecular Genetics, Indiana University School of Medicine Indianapolis, IN 46202, USA
- Department of Anatomy, Cell Biology and Physiology, Indiana University School of Medicine Indianapolis, IN 46202, USA
| | - Jun Wan
- Department of Medical and Molecular Genetics, Indiana University School of Medicine Indianapolis, IN 46202, USA
- Department of Anatomy, Cell Biology and Physiology, Indiana University School of Medicine Indianapolis, IN 46202, USA
| | | | - Rebecca Marker
- Division of Translational Radiation Sciences, Department of Radiation Oncology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Zahra Gohari
- University of Maryland Marlene and Stewart Greenebaum Comprehensive Cancer Center, Baltimore, MD 21201, USA
- Division of Translational Radiation Sciences, Department of Radiation Oncology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | | | | | - Tanjina Kader
- Laboratory of Systems Pharmacology, Harvard Medical School, Boston, MA, USA
- Ludwig Center at Harvard, Boston, MA, USA
| | - Sandro Santagata
- Laboratory of Systems Pharmacology, Harvard Medical School, Boston, MA, USA
- Ludwig Center at Harvard, Boston, MA, USA
- Department of Pathology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Ronny Drapkin
- Penn Ovarian Cancer Research Center, Department of Obstetrics and Gynecology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- Basser Center for BRCA, Abramson Cancer Center, University of Pennsylvania School of Medicine, Philadelphia, PA, USA
| | - Stefan Kommoss
- Diakonie-Klinikum Schwäbisch Hall, Germany, Institute for Health Informatics
- Kliniken Essen-Mitte, Gynäkologie und Gynäkologische Onkologie, Germany
| | | | | | - Ryan Coopergard
- Institute for Health Informatics, University of Minnesota, Minneapolis, MN 55455, USA
| | | | - Boris J. N. Winterhoff
- Department of Obstetrics, Gynecology and Women’s Health (OBGYN), Division of Gynecologic Oncology, University of Minesota, MN 55812-3011, USA
| | - Michael J. Topper
- Department of Oncology, The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, MD 21231, USA
| | - George E. Sandusky
- Pathology and Laboratory Medicine, Indiana University School of Medicine, Indianapolis, IN 46202, USA
- Indiana University Melvin and Bren Simon Comprehensive Cancer Center, Indianapolis, IN 46202, USA
| | - Kathy D. Miller
- Indiana University Melvin and Bren Simon Comprehensive Cancer Center, Indianapolis, IN 46202, USA
- Department of Medicine, Division of Hematology/Oncology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Stephen B. Baylin
- Van Andel Research Institute, Grand Rapids, MI 49503, USA
- Department of Oncology, The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, MD 21231, USA
| | - Kenneth P. Nephew
- Medical Sciences Program, Indiana University School of Medicine-Bloomington, Bloomington, IN 47405, USA
- Department of Anatomy, Cell Biology and Physiology, Indiana University School of Medicine Indianapolis, IN 46202, USA
- Indiana University Melvin and Bren Simon Comprehensive Cancer Center, Indianapolis, IN 46202, USA
| | - Feyruz V. Rassool
- University of Maryland Marlene and Stewart Greenebaum Comprehensive Cancer Center, Baltimore, MD 21201, USA
- Division of Translational Radiation Sciences, Department of Radiation Oncology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| |
Collapse
|