1
|
Soler-Agesta R, Beltrán-Visiedo M, Sato A, Yamazaki T, Guilbaud E, Yim CY, Congenie MT, Ames TD, Anel A, Galluzzi L. Partial mitochondrial involvement in the antiproliferative and immunostimulatory effects of PT-112. Oncoimmunology 2025; 14:2507245. [PMID: 40386940 PMCID: PMC12091903 DOI: 10.1080/2162402x.2025.2507245] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2025] [Revised: 04/18/2025] [Accepted: 05/13/2025] [Indexed: 05/20/2025] Open
Abstract
PT-112 is a novel small molecule exhibiting promising clinical activity in patients with solid tumors. PT-112 kills malignant cells by inhibiting ribosome biogenesis while promoting the emission of immunostimulatory signals. Accordingly, PT-112 is an authentic immunogenic cell death (ICD) inducer and synergizes with immune checkpoint inhibitors in preclinical models of mammary and colorectal carcinoma. Moreover, PT-112 monotherapy has led to durable clinical responses, some of which persisting after treatment discontinuation. Mitochondrial outer membrane permeabilization (MOMP) regulates the cytotoxicity and immunogenicity of various anticancer agents. Here, we harnessed mouse mammary carcinoma TS/A cells to test whether genetic alterations affecting MOMP influence PT-112 activity. As previously demonstrated, PT-112 elicited robust antiproliferative and cytotoxic effects against TS/A cells, which were preceded by the ICD-associated exposure of calreticulin (CALR) on the cell surface, and accompanied by the release of HMGB1 in the culture supernatant. TS/A cells responding to PT-112 also exhibited eIF2α phosphorylation and cytosolic mtDNA accumulation, secreted type I IFN, and exposed MHC Class I molecules as well as the co-inhibitory ligand PD-L1 on their surface. Acute cytotoxicity and HMGB1 release caused by PT-112 in TS/A cells were influenced by MOMP competence. Conversely, PT-112 retained antiproliferative effects and its capacity to drive type I IFN secretion as well as CALR, MHC Class I and PD-L1 exposure on the cell surface irrespective of MOMP defects. These data indicate a partial involvement of MOMP in the mechanisms of action of PT-112, suggesting that PT-112 is active across various tumor types, including malignancies with MOMP defects.
Collapse
Affiliation(s)
- Ruth Soler-Agesta
- Department of Radiation Oncology, Weill Cornell Medical College, New York, NY, USA
- Promontory Therapeutics, New York, NY, USA
| | - Manuel Beltrán-Visiedo
- Department of Radiation Oncology, Weill Cornell Medical College, New York, NY, USA
- Cancer Signaling and Microenvironment Program, Fox Chase Cancer Center, Philadelphia, PA, USA
| | - Ai Sato
- Department of Radiation Oncology, Weill Cornell Medical College, New York, NY, USA
- Cancer Signaling and Microenvironment Program, Fox Chase Cancer Center, Philadelphia, PA, USA
| | - Takahiro Yamazaki
- Department of Radiation Oncology, Weill Cornell Medical College, New York, NY, USA
| | - Emma Guilbaud
- Department of Radiation Oncology, Weill Cornell Medical College, New York, NY, USA
- Cancer Signaling and Microenvironment Program, Fox Chase Cancer Center, Philadelphia, PA, USA
| | | | | | | | - Alberto Anel
- Biochemistry and Molecular and Cell Biology, Aragón Health Research Institute (IIS-Aragón), University of Zaragoza, Zaragoza, Spain
| | - Lorenzo Galluzzi
- Department of Radiation Oncology, Weill Cornell Medical College, New York, NY, USA
- Cancer Signaling and Microenvironment Program, Fox Chase Cancer Center, Philadelphia, PA, USA
| |
Collapse
|
2
|
Mishra Y, Kumar A, Kaundal RK. Mitochondrial Dysfunction is a Crucial Immune Checkpoint for Neuroinflammation and Neurodegeneration: mtDAMPs in Focus. Mol Neurobiol 2025; 62:6715-6747. [PMID: 39115673 DOI: 10.1007/s12035-024-04412-0] [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: 07/14/2024] [Accepted: 07/30/2024] [Indexed: 01/03/2025]
Abstract
Neuroinflammation is a pivotal factor in the progression of both age-related and acute neurodegenerative disorders, including Alzheimer's disease, Parkinson's disease, amyotrophic lateral sclerosis, multiple sclerosis, and stroke. Mitochondria, essential for neuronal health due to their roles in energy production, calcium buffering, and oxidative stress regulation, become increasingly susceptible to dysfunction under conditions of metabolic stress, aging, or injury. Impaired mitophagy in aged or injured neurons leads to the accumulation of dysfunctional mitochondria, which release mitochondrial-derived damage-associated molecular patterns (mtDAMPs). These mtDAMPs act as immune checkpoints, activating pattern recognition receptors (PRRs) and triggering innate immune signaling pathways. This activation initiates inflammatory responses in neurons and brain-resident immune cells, releasing cytokines and chemokines that damage adjacent healthy neurons and recruit peripheral immune cells, further amplifying neuroinflammation and neurodegeneration. Long-term mitochondrial dysfunction perpetuates a chronic inflammatory state, exacerbating neuronal injury and contributing additional immunogenic components to the extracellular environment. Emerging evidence highlights the critical role of mtDAMPs in initiating and sustaining neuroinflammation, with circulating levels of these molecules potentially serving as biomarkers for disease progression. This review explores the mechanisms of mtDAMP release due to mitochondrial dysfunction, their interaction with PRRs, and the subsequent activation of inflammatory pathways. We also discuss the role of mtDAMP-triggered innate immune responses in exacerbating both acute and chronic neuroinflammation and neurodegeneration. Targeting dysfunctional mitochondria and mtDAMPs with pharmacological agents presents a promising strategy for mitigating the initiation and progression of neuropathological conditions.
Collapse
Affiliation(s)
- Yogesh Mishra
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER) - SAS Nagar, SAS Nagar, Punjab, India
| | - Ashutosh Kumar
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER) - SAS Nagar, SAS Nagar, Punjab, India.
| | - Ravinder Kumar Kaundal
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER) - Raebareli, Lucknow, Uttar Pradesh, India.
| |
Collapse
|
3
|
Küng C, Lazarou M, Nguyen TN. Advances in mitophagy initiation mechanisms. Curr Opin Cell Biol 2025; 94:102493. [PMID: 40117675 DOI: 10.1016/j.ceb.2025.102493] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2025] [Revised: 02/19/2025] [Accepted: 02/20/2025] [Indexed: 03/23/2025]
Abstract
Mitophagy is an important lysosomal degradative pathway that removes damaged or unwanted mitochondria to maintain cellular and organismal homeostasis. The mechanisms behind how mitophagy is initiated to form autophagosomes around mitochondria have gained a lot of interest since they can be potentially targeted by mitophagy-inducing therapeutics. Mitophagy initiation can be driven by various autophagy receptors or adaptors that respond to different cellular and mitochondrial stimuli, ranging from mitochondrial damage to metabolic rewiring. This review will cover recent advances in our understanding of how mitophagy is initiated, and by doing so reveal the mechanistic plasticity of how autophagosome formation can begin.
Collapse
Affiliation(s)
- Catharina Küng
- Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia; Department of Medical Biology, University of Melbourne, Melbourne, Victoria, Australia
| | - Michael Lazarou
- Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia; Department of Medical Biology, University of Melbourne, Melbourne, Victoria, Australia; Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Melbourne, Australia; Aligning Science Across Parkinson's Collaborative Research Network, Chevy Chase, MD 20185, USA.
| | - Thanh Ngoc Nguyen
- Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia; Department of Medical Biology, University of Melbourne, Melbourne, Victoria, Australia; Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Melbourne, Australia; Aligning Science Across Parkinson's Collaborative Research Network, Chevy Chase, MD 20185, USA.
| |
Collapse
|
4
|
Liu Y, Hu Y, Shan ZL. Mitochondrial DNA release mediates metabolic-associated steatohepatitis via activation of inflammatory pathways. Shijie Huaren Xiaohua Zazhi 2025; 33:344-360. [DOI: 10.11569/wcjd.v33.i5.344] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/24/2025] [Revised: 04/25/2025] [Accepted: 05/19/2025] [Indexed: 05/28/2025] Open
Affiliation(s)
- Ying Liu
- Gannan Institute of Medical Innovation and Translational Medicine, Gannan Medical University, Ganzhou 431000, Jiangxi Province, China
| | - Yang Hu
- Gannan Institute of Medical Innovation and Translational Medicine, Gannan Medical University, Ganzhou 431000, Jiangxi Province, China
| | - Zhao-Liang Shan
- Gannan Institute of Medical Innovation and Translational Medicine, Gannan Medical University, Ganzhou 431000, Jiangxi Province, China
| |
Collapse
|
5
|
Zhou Q, Luo J, Chai X, Yang J, Zhong S, Zhang Z, Chang X, Wang H. Therapeutic targeting the cGAS-STING pathway associated with protein and gene: An emerging and promising novel strategy for aging-related neurodegenerative disease. Int Immunopharmacol 2025; 156:114679. [PMID: 40252469 DOI: 10.1016/j.intimp.2025.114679] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2025] [Revised: 04/11/2025] [Accepted: 04/13/2025] [Indexed: 04/21/2025]
Abstract
Neurodegenerative diseases (NDDs) represent a rapidly escalating global health challenge, contributing significantly to the worldwide disease burden and posing substantial threats to public health systems across nations. Among the many risk factors for neurodegeneration, aging is the major risk factor. In the context of aging, multiple factors lead to the release of endogenous DNA (especially mitochondrial DNA, mtDNA), which is an important trigger for the activation of the cGAS-STING innate immune pathway. Recent studies have identified an increasing role for activation of the cGAS-STING signaling pathway as a driver of senescence-associated secretory phenotypes (SASPs) in aging and NDDs. The cGAS-STING pathway mediates the immune sensing of DNA and is a key driver of chronic inflammation and functional decline during the aging process. Blocking cGAS-STING signaling may reduce the inflammatory response by preventing mtDNA release and enhancing mitophagy. Targeted inhibition of the cGAS-STING pathway by biological macromolecules such as natural products shows promise in therapeutic strategies for age-related NDDs. This review aims to systematically and comprehensively introduces the role of the cGAS-STING pathway in age-related NDDs in the context of aging while revealing the molecular mechanisms of the cGAS-STING pathway and its downstream signaling pathways and to develop more targeted and effective therapeutic strategies for NDDs.
Collapse
Affiliation(s)
- Qiongli Zhou
- Department of Toxicology, School of Public Health, Lanzhou University, Gansu 730000, China
| | - Jinghao Luo
- Department of Toxicology, School of Public Health, Lanzhou University, Gansu 730000, China
| | - Xueting Chai
- Department of Toxicology, School of Public Health, Lanzhou University, Gansu 730000, China
| | - Jirui Yang
- Department of Toxicology, School of Public Health, Lanzhou University, Gansu 730000, China
| | - Shiyin Zhong
- Department of Toxicology, School of Public Health, Lanzhou University, Gansu 730000, China
| | - Zhimin Zhang
- Department of Toxicology, School of Public Health, Lanzhou University, Gansu 730000, China
| | - Xuhong Chang
- Department of Toxicology, School of Public Health, Lanzhou University, Gansu 730000, China
| | - Hui Wang
- Department of Toxicology, School of Public Health, Lanzhou University, Gansu 730000, China.
| |
Collapse
|
6
|
Sun C, Gui J, Sheng Y, Huang L, Zhu X, Huang K. Specific signaling pathways mediated programmed cell death in tumor microenvironment and target therapies. Discov Oncol 2025; 16:776. [PMID: 40377777 PMCID: PMC12084487 DOI: 10.1007/s12672-025-02592-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2024] [Accepted: 05/06/2025] [Indexed: 05/18/2025] Open
Abstract
Increasing evidence has shown that programmed cell death (PCD) plays a crucial role in tumorigenesis and cancer progression. The components of PCD are complex and include various mechanisms such as apoptosis, necroptosis, alkaliptosis, oxeiptosis, and anoikis, all of which are interrelated in their functions and regulatory pathways. Given the significance of these processes, it is essential to conduct a comprehensive study on PCD to elucidate its multifaceted nature. Key signaling pathways, particularly the caspase signaling pathway, the RIPK1/RIPK3/MLKL pathway, and the mTOR signaling pathway, are pivotal in regulating PCD and influencing tumor progression. In this review, we briefly describe the generation mechanisms of different PCD components and focus on the regulatory mechanisms of these three major signaling pathways within the context of global PCD. Furthermore, we discuss various tumor therapeutic compounds that target different signaling axes of these pathways, which may provide novel strategies for effective tumor therapy and help improve patient outcomes in cancer treatment.
Collapse
Affiliation(s)
- Chengpeng Sun
- The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, No.1, Minde Road, Donghu District, Nanchang, 330006, Jiangxi, China
- HuanKui Academy, Jiangxi Medical College, Nanchang, 330031, China
| | - Jiawei Gui
- The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, No.1, Minde Road, Donghu District, Nanchang, 330006, Jiangxi, China
- HuanKui Academy, Jiangxi Medical College, Nanchang, 330031, China
| | - Yilei Sheng
- The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, No.1, Minde Road, Donghu District, Nanchang, 330006, Jiangxi, China
- HuanKui Academy, Jiangxi Medical College, Nanchang, 330031, China
| | - Le Huang
- The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, No.1, Minde Road, Donghu District, Nanchang, 330006, Jiangxi, China
- Jiangxi Province Key Laboratory of Neurological Diseases, Nanchang, 330006, Jiangxi, China
| | - Xingen Zhu
- The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, No.1, Minde Road, Donghu District, Nanchang, 330006, Jiangxi, China.
- Jiangxi Province Key Laboratory of Neurological Diseases, Nanchang, 330006, Jiangxi, China.
- JXHC Key Laboratory of Neurological Medicine, Nanchang, 330006, Jiangxi, China.
- Institute of Neuroscience, Jiangxi Medical College, Nanchang University, Nanchang, 330006, Jiangxi, China.
- The MOE Basic Research and Innovation Center for the Targeted Therapeutics of Solid Tumors, Jiangxi Medical College, Nanchang University, Nanchang, 330006, Jiangxi, China.
| | - Kai Huang
- The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, No.1, Minde Road, Donghu District, Nanchang, 330006, Jiangxi, China.
- Jiangxi Province Key Laboratory of Neurological Diseases, Nanchang, 330006, Jiangxi, China.
- JXHC Key Laboratory of Neurological Medicine, Nanchang, 330006, Jiangxi, China.
- Institute of Neuroscience, Jiangxi Medical College, Nanchang University, Nanchang, 330006, Jiangxi, China.
- The MOE Basic Research and Innovation Center for the Targeted Therapeutics of Solid Tumors, Jiangxi Medical College, Nanchang University, Nanchang, 330006, Jiangxi, China.
| |
Collapse
|
7
|
Guilbaud E, Naulin F, Meziani L, Deutsch E, Galluzzi L. Impact of radiation therapy on the immunological tumor microenvironment. Cell Chem Biol 2025; 32:678-693. [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] [MESH Headings] [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
|
8
|
He S, Pan T, Tian R, He Q, Cheng D, Qu H, Li R, Tan R. Fatty acid synthesis promotes mtDNA release via ETS1-mediated oligomerization of VDAC1 facilitating endothelial dysfunction in sepsis-induced lung injury. Cell Death Differ 2025:10.1038/s41418-025-01524-5. [PMID: 40369168 DOI: 10.1038/s41418-025-01524-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2024] [Revised: 04/18/2025] [Accepted: 04/30/2025] [Indexed: 05/16/2025] Open
Abstract
Sepsis involves endothelial cell dysfunction leading to the development of lung injury. Fatty acid synthesis contributes to the development of inflammatory injury in sepsis. However, the regulatory mechanisms of fatty acid synthesis-related endothelial activation remain unclear. In this study, we found that fatty acid synthesis in patients with sepsis was greatly disordered. Inhibition of fatty acid synthesis significantly alleviated sepsis-induced endothelial damage and lung injury both in vitro and in vivo. We further found that the release of mtDNA participated in fatty acid synthesis-related regulation of endothelial inflammatory and coagulation activation. Mechanistically, fatty acid synthesis promoted the oligomerization of voltage-dependent anion channel 1 (VDAC1) via ETS proto-oncogene 1 (ETS1)-mediated inhibition of VDAC1 ubiquitination, thereby leading to the increased release of mtDNA and subsequent activation of cGAS-STING signaling and pyroptosis in endothelial cells. Our findings revealed that fatty acid synthesis promoted endothelial dysfunction through mtDNA release, providing new insight into the therapeutic strategies for treating sepsis-associated lung injury.
Collapse
Affiliation(s)
- Shiyuan He
- Department of Critical Care Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, PR China
| | - Tingting Pan
- Department of Critical Care Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, PR China
| | - Rui Tian
- Department of Critical Care Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, PR China
| | - Qian He
- Department of Critical Care Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, PR China
| | - Decui Cheng
- Department of Critical Care Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, PR China
| | - Hongping Qu
- Department of Critical Care Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, PR China.
| | - Ranran Li
- Department of Critical Care Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, PR China.
| | - Ruoming Tan
- Department of Critical Care Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, PR China.
| |
Collapse
|
9
|
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
|
10
|
D'Alessandro MCB, Kanaan S, Geller M, Praticò D, Daher JPL. Mitochondrial dysfunction in Alzheimer's disease. Ageing Res Rev 2025; 107:102713. [PMID: 40023293 DOI: 10.1016/j.arr.2025.102713] [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: 11/06/2024] [Revised: 02/13/2025] [Accepted: 02/27/2025] [Indexed: 03/04/2025]
Abstract
Alzheimer's disease (AD) is a chronic neurodegenerative disease characterized by progressive cognitive decline and distinct neuropathological features. The absence of a definitive cure presents a significant challenge in neurology and neuroscience. Early clinical manifestations, such as memory retrieval deficits and apathy, underscore the need for a deeper understanding of the disease's underlying mechanisms. While amyloid-β plaques and tau neurofibrillary tangles have dominated research efforts, accumulating evidence highlights mitochondrial dysfunction as a central factor in AD pathogenesis. Mitochondria, essential cellular organelles responsible for energy production necessary for neuronal function become impaired in AD, triggering several cellular consequences. Factors such as oxidative stress, disturbances in energy metabolism, failures in the mitochondrial quality control system, and dysregulation of calcium release are associated with mitochondrial dysfunction. These abnormalities are closely linked to the neurodegenerative processes driving AD development and progression. This review explores the intricate relationship between mitochondrial dysfunction and AD pathogenesis, emphasizing its role in disease onset and progression, while also considering its potential as a biomarker and a therapeutic target.
Collapse
Affiliation(s)
- Maria Clara Bila D'Alessandro
- Universidade Federal Fluminense, Faculty of Medicine, Desembargador Athayde Parreiras road 100, Niterói, Rio de Janeiro, Brazil.
| | - Salim Kanaan
- Universidade Federal Fluminense, Faculty of Medicine, Department of Pathology, Marquês do Paraná road, 303, 2nd floor, Niterói, Rio de Janeiro, Brazil.
| | - Mauro Geller
- Unifeso, Department of Immunology and Microbiology, Alberto Torres avenue 111, Teresópolis, Rio de Janeiro, Brazil
| | - Domenico Praticò
- Department of Neurosciences, Lewis Katz School of Medicine. Temple University, 3500 North Broad Street, Philadelphia, PA, United States.
| | - João Paulo Lima Daher
- Universidade Federal Fluminense, Faculty of Medicine, Department of Pathology, Marquês do Paraná road, 303, 2nd floor, Niterói, Rio de Janeiro, Brazil.
| |
Collapse
|
11
|
Lee-Glover LP, Picard M, Shutt TE. Mitochondria - the CEO of the cell. J Cell Sci 2025; 138:jcs263403. [PMID: 40310473 PMCID: PMC12070065 DOI: 10.1242/jcs.263403] [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: 05/02/2025] Open
Abstract
As we have learned more about mitochondria over the past decades, including about their essential cellular roles and how altered mitochondrial biology results in disease, it has become apparent that they are not just powerplants pumping out ATP at the whim of the cell. Rather, mitochondria are dynamic information and energy processors that play crucial roles in directing dozens of cellular processes and behaviors. They provide instructions to enact programs that regulate various cellular operations, such as complex metabolic networks, signaling and innate immunity, and even control cell fate, dictating when cells should divide, differentiate or die. To help current and future generations of cell biologists incorporate the dynamic, multifaceted nature of mitochondria and assimilate modern discoveries into their scientific framework, mitochondria need a 21st century 'rebranding'. In this Opinion article, we argue that mitochondria should be considered as the 'Chief Executive Organelle' - the CEO - of the cell.
Collapse
Affiliation(s)
- Laurie P. Lee-Glover
- Department of Biochemistry & Molecular Biology, Cumming School of Medicine, University of Calgary, Alberta, T2N 4N1, Canada
| | - Martin Picard
- Department of Psychiatry, Division of Behavioral Medicine, Columbia University Irving Medical Center, New York, 10032, USA
- Department of Neurology, H. Houston Merritt Center for Neuromuscular and Mitochondrial Disorders, Columbia University Translational Neuroscience Initiative, Columbia University Irving Medical Center, New York, 10032, USA
- New York State Psychiatric Institute, New York, 10032, USA
- Robert N Butler Columbia Aging Center, Columbia University Mailman School of Public Health, New York, 10032, USA
| | - Timothy E. Shutt
- Department of Biochemistry & Molecular Biology, Cumming School of Medicine, University of Calgary, Alberta, T2N 4N1, Canada
- Department of Medical Genetics, Cumming School of Medicine, University of Calgary, Alberta, T2N 4N1, Canada
- Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Alberta, T2N 4N1, Canada
- Alberta Children's Hospital Research Institute, Cumming School of Medicine, University of Calgary, Alberta, T2N 4N1, Canada
- Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Alberta, T2N 4N1, Canada
| |
Collapse
|
12
|
Aparicio-Trejo OE, Hernández-Cruz EY, Reyes-Fermín LM, Ceja-Galicia ZA, Pedraza-Chaverri J. The role of redox signaling in mitochondria and endoplasmic reticulum regulation in kidney diseases. Arch Toxicol 2025; 99:1865-1891. [PMID: 40214774 DOI: 10.1007/s00204-025-04041-z] [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: 01/10/2025] [Accepted: 03/20/2025] [Indexed: 05/18/2025]
Abstract
Kidney diseases are among the fastest worldwide growing pathologies. This growth together with their high mortality rate emphasizes the importance of generating vital information about the mechanism involved in their pathophysiology to determine possible therapeutic targets. Recently, mitochondrial damage and their implication in the reactive oxygen spices (ROS) signaling and redox homeostasis have emerged as a hub point in the pathologic mechanism involved in renal pathologies. ROS in low levels are necessary to maintain cell processes as well as the mitochondria homeostasis and its association with other organelles, especially the with the endoplasmic reticulum (ER). However, the information about how redox signaling interacts and interferes with other cellular processes and the mechanism involved has not been fully integrated. Furthermore, in higher concentrations, these ROS promotes pathologic pathways linked to renal disease progression like, mitochondrial biogenesis reduction, ER stress, calcium overload, inflammation, cell death and fibrosis. Therefore, the aim of this review is to describe the molecular mechanisms involved in the redox signaling influence on mitochondrial and ER homeostasis, focusing on lipid metabolism and ß-oxidation, mitochondrial biogenesis, inflammations, ER stress and calcium homeostasis, as well as the effects of these alteration in the genesis and development of renal disease, with emphasis in acute kidney injury (AKI) and chronic kidney disease (CKD).
Collapse
Affiliation(s)
- Omar Emiliano Aparicio-Trejo
- Department of Cardio-Renal Physiopathology, National Institute of Cardiology Ignacio Chávez, 14080, Mexico City, Mexico
| | | | - Laura María Reyes-Fermín
- Laboratory F-315, Department of Biology, Faculty of Chemistry, National Autonomous University of Mexico, Mexico City, Mexico
| | | | - José Pedraza-Chaverri
- Laboratory F-315, Department of Biology, Faculty of Chemistry, National Autonomous University of Mexico, Mexico City, Mexico.
| |
Collapse
|
13
|
Zhang W, Pan X, Wang L, Li W, Dai X, Zheng M, Guo H, Chen X, Xu Y, Wu H, He Q, Yang B, Ding L. Selective BCL-2 inhibitor triggers STING-dependent antitumor immunity via inducing mtDNA release. J Immunother Cancer 2025; 13:e010889. [PMID: 40300857 PMCID: PMC12056644 DOI: 10.1136/jitc-2024-010889] [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: 10/23/2024] [Accepted: 04/10/2025] [Indexed: 05/01/2025] Open
Abstract
BACKGROUND The stimulator of interferon genes (STING) signaling pathway has been demonstrated to propagate the cancer-immunity cycle and remodel the tumor microenvironment and has emerged as an appealing target for cancer immunotherapy. Interest in STING agonist development has increased, and the candidates hold significant promise; however, most are still in the early stages of human clinical trials. We found that ABT-199 activated the STING pathway to enhance the immunotherapeutic effect, and provided a ready-to-use small molecule drug for STING signaling activation. METHODS Phosphorylation of STING, TBK1, and IRF3, as well as activation of the interferon-I (IFN-I) signaling pathway, were detected following ABT-199 treatment in various colorectal cancer cells. C57BL/6J and BALB/c mice with subcutaneous tumors were employed to evaluate the in vivo therapeutic effects of the ABT-199 and anti-PD-L1 combination. Flow cytometry and ELISA were employed to analyze the level and activity of tumor-infiltrating T lymphocytes. Immunofluorescence and quantitative real-time PCR were conducted to assess the source and accumulation of double stranded DNA (dsDNA) in the cytoplasm. Chemical cross-linking assay, co-immunoprecipitation, and CRISPR/Cas9-mediated knockout were performed to investigate the molecular mechanism underlying ABT-199-induced voltage-dependent anion channel protein 1 (VDAC1) oligomerization and mitochondrial DNA (mtDNA) release. RESULTS ABT-199 significantly activated the STING signaling pathway in various colorectal cancer cells, which was evidenced by increased phosphorylation of TBK1 and IRF3, and upregulation of C-C motif chemokine ligand 5 (CCL5), C-X-C motif chemokine ligand 10 (CXCL10), and interferon beta transcription. By promoting chemokine expression and cytotoxic T-cell infiltration, ABT-199 promoted antitumor immunity and synergized with anti-PD-L1 therapy to improve antitumor efficacy. ABT-199 induced mtDNA accumulation in the cytoplasm and triggered STING signaling via the canonical pathway. cGAS or STING-KO models significantly abolished both STING signaling activation and the antitumor efficacy of ABT-199. Mechanically, ABT-199 promoted VDAC1 oligomerization by disturbing the binding between BCL-2 and VDAC1, thereby facilitating mtDNA release into the cytoplasm. ABT-199-triggered STING signaling was attenuated when VADC1 was knocked out. Consistently, the antitumor effect of ABT-199 in vivo was abolished in the absence of VDAC1. CONCLUSIONS Our results identify a ready-to-use small molecule compound for STING activation, reveal the underlying molecular mechanism through which ABT-199 activates the STING signaling pathway, and provide a theoretical basis for the use of ABT-199 in cancer immunotherapy.
Collapse
Affiliation(s)
- Wenxin Zhang
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang, China
| | - Xiaohui Pan
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang, China
| | - Longsheng Wang
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang, China
| | - Wen Li
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang, China
| | - Xiaoyang Dai
- Center of Drug Safety Evaluation and Research, Zhejiang University, Hangzhou, Zhejiang, China
| | - Mingming Zheng
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang, China
| | - Hongjie Guo
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang, China
| | - Xi Chen
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang, China
| | - Yanjun Xu
- Department of Medical Thoracic Oncology, Zhejiang Cancer Hospital, Hangzhou, Zhejiang, China
| | - Honghai Wu
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang, China
| | - Qiaojun He
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang, China
- Cancer Center of Zhejiang University, Hangzhou, Zhejiang, China
- The Innovation Institute for Artificial Intelligence in Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Bo Yang
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang, China
- The Innovation Institute for Artificial Intelligence in Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Ling Ding
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang, China
- Nanhu Brain-computer Interface Institute, Hangzhou, Zhejiang, China
| |
Collapse
|
14
|
Lei S, Li J, Zhu M, Zhou W, Fu X, Wu S, Chen X, Zhang J, Duan X, Wang W, Men K. Chimeric Antigen Receptor-Engineered Cell Membrane-Coated Nanoparticles Promote Dual-Targeted mRNA-Based Cancer Gene Therapy. ACS NANO 2025; 19:15668-15684. [PMID: 40227438 DOI: 10.1021/acsnano.4c18153] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/15/2025]
Abstract
Gene therapy using mRNA has facilitated progress in cancer therapy. However, its application is hindered by a limited tumor-targeted delivery approach, leading to off-target effects and safety concerns. Chimeric antigen receptor (CAR) molecules enable T cells to recognize specific antigens in a major histocompatibility complex-unrestricted manner. CAR approaches provide an "off-the-shelf" solution for introducing additional targeting functionality to a cell membrane. Cancer cell membrane-coated nanoparticles with homotypic tumor-targeted properties provide a readily accessible platform for gene engineering and membrane extraction. Herein, we demonstrate a CAR-inspired cancer cell membrane-coated platform for delivering an mRNA formulation through a dual tumor-targeted mechanism. The simplified human epidermal growth factor receptor 2 (HER2)-specific CAR molecule (comprising an extracellular HER2-binding domain, a hinge, and a transmembrane domain) was engineered on the cell membrane of cancer cells to establish CAR-CT26 cells. The extracted CAR-CT26 membrane (CARM) was subsequently coated onto the lipid nanoparticle (LNP)-mRNA surface to form a CARM@LNP-mRNA complex. In vitro, the CARM-coated nanoparticles exhibited enhanced mRNA transfection efficiency toward CT26 cells overexpressing target HER2 antigens. Systemic administration of the CARM@LNP-mRNA formulation resulted in stronger tumor-targeting ability and tumor suppression in HER2+ CT26 subcutaneous tumors and peritoneal cavity metastasis models than that observed with the CT26 cell membrane-coated version. Our data suggest that CARM@LNP is a feasible choice for mRNA-based gene therapy. These results provide evidence for the systemic administration of CARM@LNP-mRNA as a promising tumor-targeted therapeutic strategy.
Collapse
Affiliation(s)
- Sibei Lei
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
- Department of Pharmacy, The First People's Hospital of Yunnan Province, The Affiliated Hospital of Kunming University of Science and Technology, Kunming 650032, China
| | - Jingmei Li
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Manfang Zhu
- Department of Pharmacy, Personalized Drug Therapy Key Laboratory of Sichuan Province Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu 610072, China
| | - Weilin Zhou
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Xizi Fu
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Shan Wu
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Xiayu Chen
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Jin Zhang
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Xingmei Duan
- Department of Pharmacy, Personalized Drug Therapy Key Laboratory of Sichuan Province Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu 610072, China
| | - Wei Wang
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Ke Men
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| |
Collapse
|
15
|
Giordano L, Ware SA, Lagranha CJ, Kaufman BA. Mitochondrial DNA signals driving immune responses: Why, How, Where? Cell Commun Signal 2025; 23:192. [PMID: 40264103 PMCID: PMC12012978 DOI: 10.1186/s12964-025-02042-0] [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: 10/29/2024] [Accepted: 01/14/2025] [Indexed: 04/24/2025] Open
Abstract
There has been a recent expansion in our understanding of DNA-sensing mechanisms. Mitochondrial dysfunction, oxidative and proteostatic stresses, instability and impaired disposal of nucleoids cause the release of mitochondrial DNA (mtDNA) from the mitochondria in several human diseases, as well as in cell culture and animal models. Mitochondrial DNA mislocalized to the cytosol and/or the extracellular compartments can trigger innate immune and inflammation responses by binding DNA-sensing receptors (DSRs). Here, we define the features that make mtDNA highly immunogenic and the mechanisms of its release from the mitochondria into the cytosol and the extracellular compartments. We describe the major DSRs that bind mtDNA such as cyclic guanosine-monophosphate-adenosine-monophosphate synthase (cGAS), Z-DNA-binding protein 1 (ZBP1), NOD-, LRR-, and PYD- domain-containing protein 3 receptor (NLRP3), absent in melanoma 2 (AIM2) and toll-like receptor 9 (TLR9), and their downstream signaling cascades. We summarize the key findings, novelties, and gaps of mislocalized mtDNA as a driving signal of immune responses in vascular, metabolic, kidney, lung, and neurodegenerative diseases, as well as viral and bacterial infections. Finally, we define common strategies to induce or inhibit mtDNA release and propose challenges to advance the field.
Collapse
Affiliation(s)
- Luca Giordano
- Center for Metabolism and Mitochondrial Medicine, Division of Cardiology, Department of Medicine, University of Pittsburgh, Pittsburgh, PA, USA.
- Heart, Lung, and Blood Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA, USA.
- Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Cardio-Pulmonary Institute (CPI), Justus-Liebig-University, Giessen, Germany.
| | - Sarah A Ware
- Center for Metabolism and Mitochondrial Medicine, Division of Cardiology, Department of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
- Heart, Lung, and Blood Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA, USA
| | - Claudia J Lagranha
- Center for Metabolism and Mitochondrial Medicine, Division of Cardiology, Department of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
- Heart, Lung, and Blood Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA, USA
| | - Brett A Kaufman
- Center for Metabolism and Mitochondrial Medicine, Division of Cardiology, Department of Medicine, University of Pittsburgh, Pittsburgh, PA, USA.
- Heart, Lung, and Blood Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA, USA.
| |
Collapse
|
16
|
Basak B, Holzbaur ELF. Mitophagy in Neurons: Mechanisms Regulating Mitochondrial Turnover and Neuronal Homeostasis. J Mol Biol 2025:169161. [PMID: 40268233 DOI: 10.1016/j.jmb.2025.169161] [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: 01/10/2025] [Revised: 04/14/2025] [Accepted: 04/15/2025] [Indexed: 04/25/2025]
Abstract
Mitochondrial quality control is instrumental in regulating neuronal health and survival. The receptor-mediated clearance of damaged mitochondria by autophagy, known as mitophagy, plays a key role in controlling mitochondrial homeostasis. Mutations in genes that regulate mitophagy are causative for familial forms of neurological disorders including Parkinson's disease (PD) and Amyotrophic lateral sclerosis (ALS). PINK1/Parkin-dependent mitophagy is the best studied mitophagy pathway, while more recent work has brought to light additional mitochondrial quality control mechanisms that operate either in parallel to or independent of PINK1/Parkin mitophagy. Here, we discuss our current understanding of mitophagy mechanisms operating in neurons to govern mitochondrial homeostasis. We also summarize progress in our understanding of the links between mitophagic dysfunction and neurodegeneration, and highlight the potential for therapeutic interventions to maintain mitochondrial health and neuronal function.
Collapse
Affiliation(s)
- Bishal Basak
- Department of Physiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD 20815, USA
| | - Erika L F Holzbaur
- Department of Physiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD 20815, USA.
| |
Collapse
|
17
|
Zou Z, Cheng X, Chen J, Xing C, Zhang C, Guo X, Cao H, Hu G, Zhuang Y. Curcumin alleviates atrazine-induced nephrotoxicity by enhancing mitophagy through PINK1/Parkin signaling pathway in mice. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2025; 295:118118. [PMID: 40209350 DOI: 10.1016/j.ecoenv.2025.118118] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Revised: 03/26/2025] [Accepted: 03/27/2025] [Indexed: 04/12/2025]
Abstract
Atrazine (ATR), a widely used herbicide, poses significant environmental and health risks due to its high solubility and adsorption in soil. ATR exposure can lead to nephrotoxicity in humans and animals. Curcumin (Cur), an active compound in Curcuma species, is renowned for its antioxidant and anti-inflammatory properties, with potential to mitigate chronic disease risks. We hypothesized that the addition of Cur could alleviate renal impairment associated with ATR exposure and carried out experiments using mice as subjects. This study investigates whether Cur can attenuate ATR-induced nephrotoxicity in mice by modulating mitophagy and apoptotic pathways. Our findings illustrate that consumption with Cur attenuates nephrotoxicity induced by ATR, as evidenced by lowered serum concentrations of uric acid (UA), blood urea nitrogen (BUN), and creatinine (CRE), established biomarkers of renal injury. Moreover, Curcumin enhances renal antioxidant defense mechanisms in ATR-exposed mice, as indicated by elevated levels of total antioxidant capacity (T-AOC), catalase (CAT), and glutathione peroxidase (GSH-Px), alongside reduced levels of malondialdehyde (MDA). Histopathological and electron microscopy analyses further corroborate these findings, showing reduced organelle damage, particularly mitochondrial ridge breakage and vacuolization, and increased autophagic lysosomes. Cur further enhances PINK1/Parkin-mediated autophagy, as evidenced by elevated levels of PINK1, Parkin, LC3BII, and P62 compared to ATR-treated mice. Moreover, Cur mitigates the mitochondrial apoptotic pathway, indicated by the down-regulation of apoptosis-related genes (Cytochrome C (Cyto-C), Caspase3, Caspase9) and the pro-apoptotic marker (Bax), along with the up-regulation of the anti-apoptotic marker (Bcl-2) at both transcriptional and translational levels compared to ATR-treated mice. In summary, Cur demonstrates nephroprotective properties against ATR-induced injury through the enhancement of mitochondrial autophagy and display of anti-apoptotic actions, underscoring its curative potency as a treatment for nephrotoxicity caused by ATR.
Collapse
Affiliation(s)
- Zhenyu Zou
- Jiangxi Provincial Key Laboratory for Animal Health, Institute of Animal Population Health, College of Animal Science and Technology, Jiangxi Agricultural University, No. 1101 Zhimin Avenue, Economic and Technological Development District, Jiangx, Nanchang 330045, PR China
| | - Xinyi Cheng
- Jiangxi Provincial Key Laboratory for Animal Health, Institute of Animal Population Health, College of Animal Science and Technology, Jiangxi Agricultural University, No. 1101 Zhimin Avenue, Economic and Technological Development District, Jiangx, Nanchang 330045, PR China
| | - Jinyan Chen
- Jiangxi Provincial Key Laboratory for Animal Health, Institute of Animal Population Health, College of Animal Science and Technology, Jiangxi Agricultural University, No. 1101 Zhimin Avenue, Economic and Technological Development District, Jiangx, Nanchang 330045, PR China
| | - Chenghong Xing
- Jiangxi Provincial Key Laboratory for Animal Health, Institute of Animal Population Health, College of Animal Science and Technology, Jiangxi Agricultural University, No. 1101 Zhimin Avenue, Economic and Technological Development District, Jiangx, Nanchang 330045, PR China
| | - Caiying Zhang
- Jiangxi Provincial Key Laboratory for Animal Health, Institute of Animal Population Health, College of Animal Science and Technology, Jiangxi Agricultural University, No. 1101 Zhimin Avenue, Economic and Technological Development District, Jiangx, Nanchang 330045, PR China
| | - Xiaoquan Guo
- Jiangxi Provincial Key Laboratory for Animal Health, Institute of Animal Population Health, College of Animal Science and Technology, Jiangxi Agricultural University, No. 1101 Zhimin Avenue, Economic and Technological Development District, Jiangx, Nanchang 330045, PR China
| | - Huabing Cao
- Jiangxi Provincial Key Laboratory for Animal Health, Institute of Animal Population Health, College of Animal Science and Technology, Jiangxi Agricultural University, No. 1101 Zhimin Avenue, Economic and Technological Development District, Jiangx, Nanchang 330045, PR China
| | - Guoliang Hu
- Jiangxi Provincial Key Laboratory for Animal Health, Institute of Animal Population Health, College of Animal Science and Technology, Jiangxi Agricultural University, No. 1101 Zhimin Avenue, Economic and Technological Development District, Jiangx, Nanchang 330045, PR China.
| | - Yu Zhuang
- Jiangxi Provincial Key Laboratory for Animal Health, Institute of Animal Population Health, College of Animal Science and Technology, Jiangxi Agricultural University, No. 1101 Zhimin Avenue, Economic and Technological Development District, Jiangx, Nanchang 330045, PR China.
| |
Collapse
|
18
|
Shi J, Yu Y, Yuan H, Li Y, Xue Y. Mitochondrial dysfunction in AMI: mechanisms and therapeutic perspectives. J Transl Med 2025; 23:418. [PMID: 40211347 PMCID: PMC11987341 DOI: 10.1186/s12967-025-06406-5] [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: 01/03/2025] [Accepted: 03/20/2025] [Indexed: 04/13/2025] Open
Abstract
Acute myocardial infarction (AMI) and the myocardial ischemia-reperfusion injury (MI/RI) that typically ensues represent a significant global health burden, accounting for a considerable number of deaths and disabilities. In the context of AMI, percutaneous coronary intervention (PCI) is the preferred treatment option for reducing acute ischemic damage to the heart. Despite the modernity of PCI therapy, pathological damage to cardiomyocytes due to MI/RI remains an important target for intervention that affects the long-term prognosis of patients. In recent years, mitochondrial dysfunction during AMI has been increasingly recognized as a critical factor in cardiomyocyte death. Damaged mitochondria play an active role in the formation of an inflammatory environment by triggering key signaling pathways, including those mediated by cyclic GMP-AMP synthase, NOD-like receptors and Toll-like receptors. This review emphasizes the dual role of mitochondria as both contributors to and regulators of inflammation. The aim is to explore the complex mechanisms of mitochondrial dysfunction in AMI and its profound impact on immune dysregulation. Specific interventions including mitochondrial-targeted antioxidants, membrane-stabilizing peptides, and mitochondrial transplantation therapies have demonstrated efficacy in preclinical AMI models.
Collapse
Affiliation(s)
- Jingle Shi
- Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Yiding Yu
- Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Huajing Yuan
- Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Yan Li
- Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China.
| | - Yitao Xue
- Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China.
| |
Collapse
|
19
|
Croce CM, Vaux D, Strasser A, Opferman JT, Czabotar PE, Fesik SW. The BCL-2 protein family: from discovery to drug development. Cell Death Differ 2025:10.1038/s41418-025-01481-z. [PMID: 40204952 DOI: 10.1038/s41418-025-01481-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2025] [Revised: 02/24/2025] [Accepted: 03/14/2025] [Indexed: 04/11/2025] Open
Abstract
The landmark discovery of the BCL-2 gene and then its function marked the identification of inhibition of apoptotic cell death as a crucial novel mechanism driving cancer development and launched the quest to discover the molecular control of apoptosis. This work culminated in the generation of specific inhibitors that are now in clinical use, saving and improving tens of thousands of lives annually. Here, some of the original players of this story, describe the sequence of critical discoveries. The t(14;18) chromosomal translocation, frequently observed in follicular lymphoma, allowed the identification and the cloning of a novel oncogene (BCL-2) juxtaposed to the immunoglobulin heavy chain gene locus (IgH). Of note, BCL-2 acted in a distinct manner as compared to then already known oncogenic proteins like ABL and c-MYC. BCL-2 did not promote cell proliferation but inhibited cell death, as originally shown in growth factor dependent haematopoietic progenitor cell lines (e.g., FDC-P1) and in Eμ-Myc/Eμ-Bcl-2 double transgenic mice. Following a rapid expansion of the BCL-2 protein family, the Abbott Laboratories solved the first structure of BCL-XL and subsequently the BCL-XL/BAK peptide complex, opening the way to understanding the structures of other BCL-2 family members and, finally, to the generation of inhibitors of the different pro-survival BCL-2 proteins, thanks to the efforts of Servier/Norvartis, Genentech/WEHI, AbbVie, Amgen, Prelude and Gilead. Although the BCL-2 inhibitor Venetoclax is in clinical use and inhibitors of BCL-XL and MCL-1 are undergoing clinical trials, several questions remain on whether therapeutic windows can be achieved and what other agents should be used in combination with BH3 mimetics to achieve optimal therapeutic impact for cancer therapy. Finally, the control of the expression of BH3-only proteins and pro-survival BCL-2 family members needs to be better understood as this may identify novel targets for cancer therapy. This story is still not concluded!
Collapse
Affiliation(s)
- Carlo M Croce
- Department of Cancer Biology and Genetics and Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA.
| | - David Vaux
- The Walter and Eliza Hall Institute, Parkville, VIC, Australia.
- Department of Medical Biology, University of Melbourne, Parkville, VIC, Australia.
| | - Andreas Strasser
- The Walter and Eliza Hall Institute, Parkville, VIC, Australia.
- Department of Medical Biology, University of Melbourne, Parkville, VIC, Australia.
| | - Joseph T Opferman
- Department of Cell and Molecular Biology, St. Jude Children's Research Hospital, Memphis, TN, USA.
| | - Peter E Czabotar
- The Walter and Eliza Hall Institute, Parkville, VIC, Australia.
- Department of Medical Biology, University of Melbourne, Parkville, VIC, Australia.
| | - Stephen W Fesik
- Department of Biochemistry, Pharmacology and Chemistry, Vanderbilt University, Nashville, TN, USA.
| |
Collapse
|
20
|
Liu D, Liu Z, Hu Y, Xiong W, Wang D, Zeng Z. MOMP: A critical event in cell death regulation and anticancer treatment. Biochim Biophys Acta Rev Cancer 2025; 1880:189280. [PMID: 39947442 DOI: 10.1016/j.bbcan.2025.189280] [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: 11/04/2024] [Revised: 01/25/2025] [Accepted: 02/04/2025] [Indexed: 02/21/2025]
Abstract
Mitochondrial outer membrane permeabilization (MOMP) refers to the increase in permeability of the mitochondrial outer membrane, allowing proteins, DNA, and other molecules to pass through the intermembrane space into the cytosol. As a crucial event in the induction of apoptosis, MOMP plays a significant role in regulating various forms of cell death, including apoptosis, ferroptosis, and pyroptosis. Importantly, MOMP is not a binary process of "all-or-nothing." Under sub-lethal stress stimuli, cells may experience a phenomenon referred to as minority MOMP (miMOMP), where only a subset of mitochondria undergo functional impairment, thereby disrupting the normal life cycle of the cell. This can lead to pathological and physiological changes such as tumor formation, cellular senescence, innate immune dysfunction, and chronic inflammation. This review focuses on the diversity of MOMP events to elucidate how varying degrees of MOMP under different stress conditions influence cell fate. Additionally, it summarizes the current research progress on novel antitumor therapeutic strategies targeting MOMP in clinical contexts.
Collapse
Affiliation(s)
- Dan Liu
- NHC Key Laboratory of Carcinogenesis and Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China; Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute and Xiangya School of Basic Medical Sciences, Central South University, Changsha, Hunan, China
| | - Ziqi Liu
- Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute and Xiangya School of Basic Medical Sciences, Central South University, Changsha, Hunan, China
| | - Yan Hu
- Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute and Xiangya School of Basic Medical Sciences, Central South University, Changsha, Hunan, China
| | - Wei Xiong
- Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute and Xiangya School of Basic Medical Sciences, Central South University, Changsha, Hunan, China
| | - Dan Wang
- NHC Key Laboratory of Carcinogenesis and Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China; Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute and Xiangya School of Basic Medical Sciences, Central South University, Changsha, Hunan, China.
| | - Zhaoyang Zeng
- NHC Key Laboratory of Carcinogenesis and Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China; Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute and Xiangya School of Basic Medical Sciences, Central South University, Changsha, Hunan, China.
| |
Collapse
|
21
|
Smarduch S, Moreno-Velasquez SD, Ilic D, Dadsena S, Morant R, Ciprinidis A, Pereira G, Binder M, García-Sáez AJ, Acebrón SP. A novel biosensor for the spatiotemporal analysis of STING activation during innate immune responses to dsDNA. EMBO J 2025; 44:2157-2182. [PMID: 39984755 PMCID: PMC11962129 DOI: 10.1038/s44318-025-00370-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Revised: 12/23/2024] [Accepted: 01/10/2025] [Indexed: 02/23/2025] Open
Abstract
The cGAS-STING signalling pathway has a central role in the innate immune response to extrinsic and intrinsic sources of cytoplasmic dsDNA. At the core of this pathway is cGAS-dependent production of the intra- and extra-cellular messenger cGAMP, which activates STING and leads to IRF3-dependent expression of cytokines and interferons. Despite its relevance to viral and bacterial infections, cell death, and genome instability, the lack of specific live-cell reporters has precluded spatiotemporal analyses of cGAS-STING signalling. Here, we generate a fluorescent biosensor termed SIRF (STING-IRF3), which reports on the functional interaction between activated STING and IRF3 at the Golgi. We show that cells harbouring SIRF react in a time- and concentration-dependent manner both to STING agonists and to microenvironmental cGAMP. We demonstrate that the new biosensor is suitable for single-cell characterisation of immune responses to HSV-1 infection, mtDNA release upon apoptosis, or other sources of cytoplasmic dsDNA. Furthermore, our results indicate that STING signalling is not activated by ruptured micronuclei, suggesting that other cytosolic pattern recognition receptors underlie the interferon responses to chromosomal instability.
Collapse
Affiliation(s)
- Steve Smarduch
- Centre for Organismal Studies (COS), Heidelberg University, Heidelberg, Germany
| | | | - Doroteja Ilic
- Division of Virus-associated Carcinogenesis, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Shashank Dadsena
- Institute of Genetics, CECAD, University of Cologne, Cologne, Germany
| | - Ryan Morant
- Centre for Organismal Studies (COS), Heidelberg University, Heidelberg, Germany
| | - Anja Ciprinidis
- Centre for Organismal Studies (COS), Heidelberg University, Heidelberg, Germany
| | - Gislene Pereira
- Centre for Organismal Studies (COS), Heidelberg University, Heidelberg, Germany
- Molecular Biology of Centrosome and Cilia, German Cancer Research Centre (DKFZ), Heidelberg, Germany
| | - Marco Binder
- Division of Virus-associated Carcinogenesis, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Ana J García-Sáez
- Institute of Genetics, CECAD, University of Cologne, Cologne, Germany
- Max Planck Institute of Biophysics, Frankfurt, Germany
| | - Sergio P Acebrón
- Centre for Organismal Studies (COS), Heidelberg University, Heidelberg, Germany.
- IKERBASQUE, Basque Foundation of Science, Bilbao, Spain.
- University of the Basque Country (UPV/EHU), Leioa, Spain.
| |
Collapse
|
22
|
McGill Percy KC, Liu Z, Qi X. Mitochondrial dysfunction in Alzheimer's disease: Guiding the path to targeted therapies. Neurotherapeutics 2025; 22:e00525. [PMID: 39827052 PMCID: PMC12047401 DOI: 10.1016/j.neurot.2025.e00525] [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: 08/31/2024] [Revised: 01/07/2025] [Accepted: 01/07/2025] [Indexed: 01/22/2025] Open
Abstract
Alzheimer's disease (AD) is characterized by progressive neurodegeneration, marked by the accumulation of amyloid-β (Aβ) plaques and tau tangles. Emerging evidence suggests that mitochondrial dysfunction plays a pivotal role in AD pathogenesis, driven by impairments in mitochondrial quality control (MQC) mechanisms. MQC is crucial for maintaining mitochondrial integrity through processes such as proteostasis, mitochondrial dynamics, mitophagy, and precise communication with other subcellular organelles. In AD, disruptions in these processes lead to bioenergetic failure, gene dysregulation, the accumulation of damaged mitochondria, neuroinflammation, and lipid homeostasis impairment, further exacerbating neurodegeneration. This review elucidates the molecular pathways involved in MQC and their pathological relevance in AD, highlighting recent discoveries related to mitochondrial mechanisms underlying neurodegeneration. Furthermore, we explore potential therapeutic strategies targeting mitochondrial dysfunction, including gene therapy and pharmacological interventions, offering new avenues for slowing AD progression. The complex interplay between mitochondrial health and neurodegeneration underscores the need for innovative approaches to restore mitochondrial function and mitigate the onset and progression of AD.
Collapse
Affiliation(s)
- Kyle C McGill Percy
- Department of Physiology & Biophysics, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA
| | - Zunren Liu
- Department of Physiology & Biophysics, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA
| | - Xin Qi
- Department of Physiology & Biophysics, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA; Center for Mitochondrial Research and Therapeutics, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA.
| |
Collapse
|
23
|
Yuan S, Sun R, Shi H, Chapman NM, Hu H, Guy C, Rankin S, Kc A, Palacios G, Meng X, Sun X, Zhou P, Yang X, Gottschalk S, Chi H. VDAC2 loss elicits tumour destruction and inflammation for cancer therapy. Nature 2025; 640:1062-1071. [PMID: 40108474 PMCID: PMC12018455 DOI: 10.1038/s41586-025-08732-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2024] [Accepted: 02/03/2025] [Indexed: 03/22/2025]
Abstract
Tumour cells often evade immune pressure exerted by CD8+ T cells or immunotherapies through mechanisms that are largely unclear1,2. Here, using complementary in vivo and in vitro CRISPR-Cas9 genetic screens to target metabolic factors, we established voltage-dependent anion channel 2 (VDAC2) as an immune signal-dependent checkpoint that curtails interferon-γ (IFNγ)-mediated tumour destruction and inflammatory reprogramming of the tumour microenvironment. Targeting VDAC2 in tumour cells enabled IFNγ-induced cell death and cGAS-STING activation, and markedly improved anti-tumour effects and immunotherapeutic responses. Using a genome-scale genetic interaction screen, we identified BAK as the mediator of VDAC2-deficiency-induced effects. Mechanistically, IFNγ stimulation increased BIM, BID and BAK expression, with VDAC2 deficiency eliciting uncontrolled IFNγ-induced BAK activation and mitochondrial damage. Consequently, mitochondrial DNA was aberrantly released into the cytosol and triggered robust activation of cGAS-STING signalling and type I IFN response. Importantly, co-deletion of STING signalling components dampened the therapeutic effects of VDAC2 depletion in tumour cells, suggesting that targeting VDAC2 integrates CD8+ T cell- and IFNγ-mediated adaptive immunity with a tumour-intrinsic innate immune-like response. Together, our findings reveal VDAC2 as a dual-action target to overcome tumour immune evasion and establish the importance of coordinately destructing and inflaming tumours to enable efficacious cancer immunotherapy.
Collapse
Affiliation(s)
- Sujing Yuan
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Renqiang Sun
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Hao Shi
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Nicole M Chapman
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Haoran Hu
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Cliff Guy
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Sherri Rankin
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Anil Kc
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Gustavo Palacios
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Xiaoxi Meng
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Xiang Sun
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Peipei Zhou
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Xiaoyang Yang
- Experimental Cellular Therapeutics Laboratory, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Stephen Gottschalk
- Department of Bone Marrow Transplantation and Cellular Therapy, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Hongbo Chi
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN, USA.
| |
Collapse
|
24
|
Chen Z, Behrendt R, Wild L, Schlee M, Bode C. Cytosolic nucleic acid sensing as driver of critical illness: mechanisms and advances in therapy. Signal Transduct Target Ther 2025; 10:90. [PMID: 40102400 PMCID: PMC11920230 DOI: 10.1038/s41392-025-02174-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Revised: 01/14/2025] [Accepted: 02/11/2025] [Indexed: 03/20/2025] Open
Abstract
Nucleic acids from both self- and non-self-sources act as vital danger signals that trigger immune responses. Critical illnesses such as acute respiratory distress syndrome, sepsis, trauma and ischemia lead to the aberrant cytosolic accumulation and massive release of nucleic acids that are detected by antiviral innate immune receptors in the endosome or cytosol. Activation of receptors for deoxyribonucleic acids and ribonucleic acids triggers inflammation, a major contributor to morbidity and mortality in critically ill patients. In the past decade, there has been growing recognition of the therapeutic potential of targeting nucleic acid sensing in critical care. This review summarizes current knowledge of nucleic acid sensing in acute respiratory distress syndrome, sepsis, trauma and ischemia. Given the extensive research on nucleic acid sensing in common pathological conditions like cancer, autoimmune disorders, metabolic disorders and aging, we provide a comprehensive summary of nucleic acid sensing beyond critical illness to offer insights that may inform its role in critical conditions. Additionally, we discuss potential therapeutic strategies that specifically target nucleic acid sensing. By examining nucleic acid sources, sensor activation and function, as well as the impact of regulating these pathways across various acute diseases, we highlight the driving role of nucleic acid sensing in critical illness.
Collapse
Affiliation(s)
- Zhaorong Chen
- Department of Anesthesiology and Intensive Care Medicine, University Hospital Bonn, 53127, Bonn, Germany
| | - Rayk Behrendt
- Institute of Clinical Chemistry and Clinical Pharmacology, University Hospital Bonn, 53127, Bonn, Germany
| | - Lennart Wild
- Department of Anesthesiology and Intensive Care Medicine, University Hospital Bonn, 53127, Bonn, Germany
| | - Martin Schlee
- Institute of Clinical Chemistry and Clinical Pharmacology, University Hospital Bonn, 53127, Bonn, Germany
| | - Christian Bode
- Department of Anesthesiology and Intensive Care Medicine, University Hospital Bonn, 53127, Bonn, Germany.
| |
Collapse
|
25
|
Wang MX, Mauch BE, Williams AF, Barazande-Pour T, Araujo Hoffmann F, Harris SH, Lathrop CP, Turkal CE, Yung BS, Paw MH, Gervasio DAG, Tran T, Stuhlfire AE, Guo T, Daniels GA, Park SJ, Gutkind JS, Hangauer MJ. Antigenic cancer persister cells survive direct T cell attack. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.14.643359. [PMID: 40166148 PMCID: PMC11956947 DOI: 10.1101/2025.03.14.643359] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
Drug-tolerant persister cancer cells were first reported fifteen years ago as a quiescent, reversible cell state which tolerates unattenuated cytotoxic drug stress. It remains unknown whether a similar phenomenon contributes to immune evasion. Here we report a persister state which survives weeks of direct cytotoxic T lymphocyte (CTL) attack. In contrast to previously known immune evasion mechanisms that avoid immune attack, antigenic persister cells robustly activate CTLs which deliver Granzyme B, secrete IFNγ, and induce tryptophan starvation resulting in apoptosis initiation. Instead of dying, persister cells paradoxically leverage apoptotic caspase activity to avoid inflammatory death. Furthermore, persister cells acquire mutations and epigenetic changes which enable outgrowth of CTL-resistant cells. Persister cell features are enriched in inflamed tumors which regressed during immunotherapy in vivo and in surgically resected human melanoma tissue under immune stress ex vivo. These findings reveal a persister cell state which is a barrier to immune-mediated tumor clearance.
Collapse
Affiliation(s)
- Michael X Wang
- Department of Dermatology, University of California San Diego
| | - Brandon E Mauch
- Department of Dermatology, University of California San Diego
| | | | | | | | - Sophie H Harris
- Department of Dermatology, University of California San Diego
| | | | - Claire E Turkal
- Department of Dermatology, University of California San Diego
| | - Bryan S Yung
- Department of Pharmacology, University of California San Diego
- Moores Cancer Center, University of California San Diego
| | - Michelle H Paw
- Department of Dermatology, University of California San Diego
| | | | - Tiffany Tran
- Department of Dermatology, University of California San Diego
| | | | - Theresa Guo
- Moores Cancer Center, University of California San Diego
- Department of Otolaryngology, University of California San Diego
| | - Gregory A Daniels
- Moores Cancer Center, University of California San Diego
- Division of Hematology-Oncology, Department of Medicine, University of California San Diego
| | - Soo J Park
- Moores Cancer Center, University of California San Diego
- Division of Hematology-Oncology, Department of Medicine, University of California San Diego
| | - J Silvio Gutkind
- Department of Pharmacology, University of California San Diego
- Moores Cancer Center, University of California San Diego
| | - Matthew J Hangauer
- Department of Dermatology, University of California San Diego
- Moores Cancer Center, University of California San Diego
| |
Collapse
|
26
|
Shu C, Li J, Rui J, Fan D, Niu Q, Bai R, Cicka D, Doyle S, Wahafu A, Zheng X, Du Y, Ivanov AA, Doxie DB, Dhodapkar KM, Carlisle J, Owonikoko T, Sica G, Liu Y, Ramalingam S, Dhodapkar MV, Zhou W, Mo X, Fu H. Uncovering the rewired IAP-JAK regulatory axis as an immune-dependent vulnerability of LKB1-mutant lung cancer. Nat Commun 2025; 16:2324. [PMID: 40057483 PMCID: PMC11890758 DOI: 10.1038/s41467-025-57297-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Accepted: 02/13/2025] [Indexed: 05/13/2025] Open
Abstract
Harnessing the power of immune system to treat cancer has become a core clinical approach. However, rewiring of intrinsic circuitry by genomic alterations enables tumor cells to escape immune surveillance, leading to therapeutic failure. Uncovering the molecular basis of how tumor mutations induce therapeutic resistance may guide the development of intervention approaches to advance precision immunotherapy. Here we report the identification of the Liver Kinase B1 (LKB1)-Inhibitor of Apoptosis Protein (IAP)- Janus Kinase 1 (JAK1) dynamic complex as a molecular determinant for immune response of LKB1-mut lung cancer cells. LKB1 alteration exposes a critical dependency of lung cancer cells on IAP for their immune resistance. Indeed, pharmacological inhibition of IAP re-establishes JAK1-regulated Stimulator of interferon genes (STING) expression and DNA sensing signaling, enhances cytotoxic immune cell infiltration, and augmentes immune-dependent anti-tumor activity in an LKB1-mutant immune-competent mouse model. Thus, IAP-JAK1-targeted strategies, like IAP inhibitors, may offer a promising therapeutic approach to restore the responsiveness of immunologically-cold LKB1-mutant tumors to immune checkpoint inhibitors or STING-directed therapies.
Collapse
Affiliation(s)
- Changfa Shu
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, GA, USA
- Department of Obstetrics and Gynecology, The Third Xiangya Hospital, Central South University, Changsha, Hunan, P R China
| | - Jianfeng Li
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, GA, USA
| | - Jin Rui
- Department of Hematology and Medical Oncology, Emory University, Atlanta, GA, USA
| | - Dacheng Fan
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, GA, USA
| | - Qiankun Niu
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, GA, USA
| | - Ruiyang Bai
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, GA, USA
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China
| | - Danielle Cicka
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, GA, USA
| | - Sean Doyle
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, GA, USA
| | - Alafate Wahafu
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, GA, USA
- The First Affiliated Hospital, Medical School of Xi'an Jiaotong University, Xi'an, Shannxi, P R China
| | - Xi Zheng
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, GA, USA
- Cancer Institute, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, P R China
| | - Yuhong Du
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, GA, USA
- Emory Chemical Biology Discovery Center, Emory University School of Medicine, Atlanta, GA, USA
- Winship Cancer Institute of Emory University, Atlanta, GA, USA
| | - Andrey A Ivanov
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, GA, USA
- Emory Chemical Biology Discovery Center, Emory University School of Medicine, Atlanta, GA, USA
- Winship Cancer Institute of Emory University, Atlanta, GA, USA
| | - Deon B Doxie
- Department of Hematology and Medical Oncology, Emory University, Atlanta, GA, USA
| | - Kavita M Dhodapkar
- Winship Cancer Institute of Emory University, Atlanta, GA, USA
- Aflac Cancer and Blood Disorders Center, Children's Healthcare of Atlanta, Emory University, Atlanta, GA, USA
| | - Jennifer Carlisle
- Department of Hematology and Medical Oncology, Emory University, Atlanta, GA, USA
- Winship Cancer Institute of Emory University, Atlanta, GA, USA
| | - Taofeek Owonikoko
- Department of Hematology and Medical Oncology, Emory University, Atlanta, GA, USA
- Winship Cancer Institute of Emory University, Atlanta, GA, USA
| | - Gabriel Sica
- Winship Cancer Institute of Emory University, Atlanta, GA, USA
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA, USA
| | - Yuan Liu
- Winship Cancer Institute of Emory University, Atlanta, GA, USA
- Rollins School of Public Health, Emory University, Atlanta, GA, USA
| | - Suresh Ramalingam
- Department of Hematology and Medical Oncology, Emory University, Atlanta, GA, USA
- Winship Cancer Institute of Emory University, Atlanta, GA, USA
| | - Madhav V Dhodapkar
- Department of Hematology and Medical Oncology, Emory University, Atlanta, GA, USA
- Winship Cancer Institute of Emory University, Atlanta, GA, USA
| | - Wei Zhou
- Department of Hematology and Medical Oncology, Emory University, Atlanta, GA, USA.
- Winship Cancer Institute of Emory University, Atlanta, GA, USA.
| | - Xiulei Mo
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, GA, USA.
- Winship Cancer Institute of Emory University, Atlanta, GA, USA.
| | - Haian Fu
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, GA, USA.
- Department of Hematology and Medical Oncology, Emory University, Atlanta, GA, USA.
- Emory Chemical Biology Discovery Center, Emory University School of Medicine, Atlanta, GA, USA.
- Winship Cancer Institute of Emory University, Atlanta, GA, USA.
| |
Collapse
|
27
|
Quan S, Fu X, Cai H, Ren Z, Xu Y, Jia L. The neuroimmune nexus: unraveling the role of the mtDNA-cGAS-STING signal pathway in Alzheimer's disease. Mol Neurodegener 2025; 20:25. [PMID: 40038765 PMCID: PMC11877805 DOI: 10.1186/s13024-025-00815-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Accepted: 02/17/2025] [Indexed: 03/06/2025] Open
Abstract
The relationship between Alzheimer's disease (AD) and neuroimmunity has gradually begun to be unveiled. Emerging evidence indicates that cyclic GMP-AMP synthase (cGAS) acts as a cytosolic DNA sensor, recognizing cytosolic damage-associated molecular patterns (DAMPs), and inducing the innate immune response by activating stimulator of interferon genes (STING). Dysregulation of this pathway culminates in AD-related neuroinflammation and neurodegeneration. A substantial body of evidence indicates that mitochondria are involved in the critical pathogenic mechanisms of AD, whose damage leads to the release of mitochondrial DNA (mtDNA) into the extramitochondrial space. This leaked mtDNA serves as a DAMP, activating various pattern recognition receptors and immune defense networks in the brain, including the cGAS-STING pathway, ultimately leading to an imbalance in immune homeostasis. Therefore, modulation of the mtDNA-cGAS-STING pathway to restore neuroimmune homeostasis may offer promising prospects for improving AD treatment outcomes. In this review, we focus on the mechanisms of mtDNA release during stress and the activation of the cGAS-STING pathway. Additionally, we delve into the research progress on this pathway in AD, and further discuss the primary directions and potential hurdles in developing targeted therapeutic drugs, to gain a deeper understanding of the pathogenesis of AD and provide new approaches for its therapy.
Collapse
Affiliation(s)
- Shuiyue Quan
- Innovation Center for Neurological Disorders and Department of Neurology, Xuanwu Hospital, Capital Medical University, National Clinical Research Center for Geriatric Diseases, 45 Changchun St, Beijing, 100053, China
| | - Xiaofeng Fu
- Innovation Center for Neurological Disorders and Department of Neurology, Xuanwu Hospital, Capital Medical University, National Clinical Research Center for Geriatric Diseases, 45 Changchun St, Beijing, 100053, China
| | - Huimin Cai
- Innovation Center for Neurological Disorders and Department of Neurology, Xuanwu Hospital, Capital Medical University, National Clinical Research Center for Geriatric Diseases, 45 Changchun St, Beijing, 100053, China
| | - Ziye Ren
- Innovation Center for Neurological Disorders and Department of Neurology, Xuanwu Hospital, Capital Medical University, National Clinical Research Center for Geriatric Diseases, 45 Changchun St, Beijing, 100053, China
| | - Yinghao Xu
- Innovation Center for Neurological Disorders and Department of Neurology, Xuanwu Hospital, Capital Medical University, National Clinical Research Center for Geriatric Diseases, 45 Changchun St, Beijing, 100053, China
| | - Longfei Jia
- Innovation Center for Neurological Disorders and Department of Neurology, Xuanwu Hospital, Capital Medical University, National Clinical Research Center for Geriatric Diseases, 45 Changchun St, Beijing, 100053, China.
| |
Collapse
|
28
|
Wang Y, Liu S, Zhou Q, Feng Y, Xu Q, Luo L, Lv H. Bioinformatics for the Identification of STING-Related Genes in Diabetic Retinopathy. Curr Eye Res 2025; 50:320-333. [PMID: 39704112 DOI: 10.1080/02713683.2024.2430223] [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: 04/13/2024] [Revised: 09/06/2024] [Accepted: 11/11/2024] [Indexed: 12/21/2024]
Abstract
PURPOSE Diabetic retinopathy (DR) is the most common complication of diabetes mellitus. Stimulator of interferon genes (STING) plays an important regulatory role in the transcription of several genes. This study aimed to mine and identify hub genes relevant to STING in DR. METHODS The STING-related genes (STING-RGs) were extracted from MSigDB database. Differentially expressed STING-RGs (DE-STING-RGs) were filtered by overlapping differentially expressed genes (DEGs) between DR and NC specimens and STING-RGs. A PPI network was established to mine hub genes. The ability of the hub genes to differentiate between DR and NC specimens was evaluated. Additionally, a ceRNA network was established to investigate the regulatory mechanisms of hub genes. Subsequently, the discrepancies in immune infiltration between DR and NC specimens were further explored. Additionally, we performed drug predictions. Finally, RT-qPCR of peripheral blood samples was used to validate the bioinformatics results. RESULTS A grand total of four genes (IKBKG, STAT6, NFKBIA, and FCGR2A) related to STING were identified for DR. The AUC values of all four hub genes were greater than 0.7, which indicated that the diagnostic value was acceptable. The ceRNA network contained four hub genes, 170 miRNAs, and 135 lncRNAs. In addition, immunoinfiltration analysis demonstrated that the abundance of activated B cells was notably different between the DR and NC specimens. Moreover, 32 drugs were included in the drug-gene network, with twelve drugs targeting STAT6, nine drugs targeting NFKBIA, four drugs targeted IKBKG, and seven drugs targeted FCGR2A. The expression of the four hub genes in blood samples determined by RT-qPCR was consistent with our analysis. CONCLUSION In conclusion, four hub genes (IKBKG, STAT6, NFKBIA, and FCGR2A) related to STING with a diagnostic value for DR were identified by bioinformatics analysis, which might provide new insights into the evaluation and treatment of DR.
Collapse
Affiliation(s)
- Yu Wang
- Department of Ophthalmology, Affiliatied Hospital of Southwest Medical University, Sichuan Province, Luzhou, China
| | - Siyan Liu
- Department of Ophthalmology, Affiliatied Hospital of Southwest Medical University, Sichuan Province, Luzhou, China
| | - Qi Zhou
- Department of Ophthalmology, Affiliatied Hospital of Southwest Medical University, Sichuan Province, Luzhou, China
| | - Yalin Feng
- Department of Ophthalmology, Affiliatied Hospital of Southwest Medical University, Sichuan Province, Luzhou, China
| | - Qin Xu
- Department of Ophthalmology, Affiliatied Hospital of Southwest Medical University, Sichuan Province, Luzhou, China
| | - Linbi Luo
- Department of Ophthalmology, Affiliatied Hospital of Southwest Medical University, Sichuan Province, Luzhou, China
| | - Hongbin Lv
- Department of Ophthalmology, Affiliatied Hospital of Southwest Medical University, Sichuan Province, Luzhou, China
| |
Collapse
|
29
|
Izquierdo JM. Mitochondria-cGAS-STING axis is a potential therapeutic target for senescence-dependent inflammaging-associated neurodegeneration. Neural Regen Res 2025; 20:805-807. [PMID: 38886948 PMCID: PMC11433916 DOI: 10.4103/nrr.nrr-d-24-00245] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Revised: 03/25/2024] [Accepted: 04/21/2024] [Indexed: 06/20/2024] Open
Affiliation(s)
- José M. Izquierdo
- Centro de Biología Molecular Severo Ochoa (CBM), Consejo Superior de Investigaciones Científicas-Universidad Autónoma de Madrid (CSIC-UAM), Madrid, Spain
| |
Collapse
|
30
|
Mallick K, Islam SR, Krishna V, Manna SK, Banerjee S. Role of AIM2 and cGAS-STING signaling in high fat high carbohydrate diet-induced gut dysbiosis associated neurodegeneration. Life Sci 2025; 363:123392. [PMID: 39805489 DOI: 10.1016/j.lfs.2025.123392] [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: 01/09/2025] [Accepted: 01/10/2025] [Indexed: 01/16/2025]
Abstract
AIMS Gut dysbiosis modulates CNS complications and cognitive decline through the gut-brain axis. The study aims to investigate the molecular mechanisms involved in gut dysbiosis-associated cognitive changes and the potential effects of probiotics in high fat-high carbohydrate diet-induced gut dysbiosis-associated neurodegeneration. MATERIALS AND METHODS We used high fat, high-carbohydrate diet (HFHCD) and high-fat diet (HFD) to induce gut dysbiosis-associated neurodegeneration in C57BL/6 mice. IVIS imaging system and biochemical changes using ELISA measured intestinal inflammation. We used fecal samples for qPCR profiling of intestinal bacteria, and serum was used for inflammatory marker analysis using ELISA. Behavioral studies measured cognitive changes, while histopathology, immunohistochemistry, and western blot analysis of hippocampal samples measured protein changes. KEY FINDINGS The behavioral studies showed a significant decrease in cognitive function associated with gut dysbiosis in HFHCD and HFD animals. Gut dysbiosis was associated with intestinal inflammation and increased intestinal permeability, followed by systemic and neuroinflammatory changes. Molecular signaling studies showed the involvement of AIM2 inflammasome and cGAS-STING signaling pathways in neurodegeneration for HFHCD animals. Administration of probiotics restored the above processes and prevented gut dysbiosis-associated memory decline in mice. SIGNIFICANCE The study shows that alteration in microbial composition due to prolonged HFHCD could contribute to intestinal inflammation and increased intestinal permeability, facilitating the translocation of microbial toxins like LPS, leading to systemic inflammation, which eventually leads to neuroinflammation and neurodegeneration.
Collapse
Affiliation(s)
- Keya Mallick
- Department of Pharmacology & Toxicology, National Institute of Pharmaceutical Education and Research, Chunilal Bhawan, 168, Maniktala Main Rd, Kolkata, West Bengal 700054, India
| | - Sk Ramiz Islam
- Biophysics & Structural Genomics Division, Saha Institute of Nuclear Physics, 1/AF Bidhannagar, Kolkata, West Bengal 700 064, India
| | - Vamsi Krishna
- Department of Pharmacology & Toxicology, National Institute of Pharmaceutical Education and Research, Chunilal Bhawan, 168, Maniktala Main Rd, Kolkata, West Bengal 700054, India
| | - Soumen Kanti Manna
- Biophysics & Structural Genomics Division, Saha Institute of Nuclear Physics, 1/AF Bidhannagar, Kolkata, West Bengal 700 064, India; Homi Bhabha National Institute, BARC Training School Complex, Anushaktinagar, Mumbai, Maharashtra 400 094, India
| | - Sugato Banerjee
- Department of Pharmacology & Toxicology, National Institute of Pharmaceutical Education and Research, Chunilal Bhawan, 168, Maniktala Main Rd, Kolkata, West Bengal 700054, India.
| |
Collapse
|
31
|
Sun Y, Aliyari SR, Parvatiyar K, Wang L, Zhen A, Sun W, Han X, Zhang A, Kato E, Shi H, De Schutter E, McBride WH, French SW, Cheng G. STING directly interacts with PAR to promote apoptosis upon acute ionizing radiation-mediated DNA damage. Cell Death Differ 2025:10.1038/s41418-025-01457-z. [PMID: 39939798 DOI: 10.1038/s41418-025-01457-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Revised: 01/13/2025] [Accepted: 01/29/2025] [Indexed: 02/14/2025] Open
Abstract
Acute ionizing radiation (IR) causes severe DNA damage, leading to cell cycle arrest, cell death, and activation of the innate immune system. The role and signaling pathway of stimulator of interferon genes (STING) in IR-induced tissue damage and cell death are not well understood. This study revealed that STING is crucial for promoting apoptosis in response to DNA damage caused by acute IR both in vitro and in vivo. STING binds to poly (ADP‒ribose) (PAR) produced by activated poly (ADP‒ribose) polymerase-1 (PARP1) upon IR. Compared with that in WT cells, apoptosis was suppressed in Stinggt-/gt- cells. Excessive PAR production by PARP1 due to DNA damage enhances STING phosphorylation, and inhibiting PARP1 reduces cell apoptosis after IR. In vivo, IR-induced crypt cell death was significantly lower in Stinggt-/gt- mice or with low-dose PARP1 inhibitor, PJ34, resulting in substantial resistance to abdominal irradiation. STING deficiency or inhibition of PARP1 function can reduce the expression of the proapoptotic gene PUMA, decrease the localization of Bax on the mitochondrial membrane, and thus reduce cell apoptosis. Our findings highlight crucial roles for STING and PAR in the IR-mediated induction of apoptosis, which may have therapeutic implications for controlling radiation-induced apoptosis or acute radiation symptoms. STING responds to acute ionizing radiation-mediated DNA damage by directly binding to poly (ADP-ribose) (PAR) produced by activated poly (ADP-ribose) polymerase-1 (PARP1), and mainly induces cell apoptosis through Puma-Bax interaction. STING deficiency or reduced production of PAR protected mice against Acute Radiation Syndrome.
Collapse
Affiliation(s)
- Yirong Sun
- CAS Key Laboratory of Regenerative Biology, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China.
- Department of Microbiology, Immunology and Molecular Genetics, University of California-Los Angeles, Los Angeles, CA, USA.
| | - Saba R Aliyari
- Department of Microbiology, Immunology and Molecular Genetics, University of California-Los Angeles, Los Angeles, CA, USA
| | - Kislay Parvatiyar
- Department of Microbiology, Immunology and Molecular Genetics, University of California-Los Angeles, Los Angeles, CA, USA
- Department of Microbiology & Immunology, Tulane University School of Medicine, New Orleans, LA, USA
| | - Lulan Wang
- Department of Microbiology, Immunology and Molecular Genetics, University of California-Los Angeles, Los Angeles, CA, USA
| | - Anjie Zhen
- Department of Microbiology, Immunology and Molecular Genetics, University of California-Los Angeles, Los Angeles, CA, USA
| | - Wei Sun
- CAS Key Laboratory of Regenerative Biology, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Xiaobo Han
- CAS Key Laboratory of Regenerative Biology, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Adele Zhang
- Department of Microbiology, Immunology and Molecular Genetics, University of California-Los Angeles, Los Angeles, CA, USA
| | - Ethan Kato
- Department of Microbiology, Immunology and Molecular Genetics, University of California-Los Angeles, Los Angeles, CA, USA
| | - Helen Shi
- Department of Microbiology, Immunology and Molecular Genetics, University of California-Los Angeles, Los Angeles, CA, USA
| | - Elena De Schutter
- Department of Microbiology, Immunology and Molecular Genetics, University of California-Los Angeles, Los Angeles, CA, USA
| | - William H McBride
- Department of Radiation Oncology, University of California-Los Angeles, Los Angeles, CA, USA
| | - Samuel W French
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California-Los Angeles, Los Angeles, CA, USA
| | - Genhong Cheng
- Department of Microbiology, Immunology and Molecular Genetics, University of California-Los Angeles, Los Angeles, CA, USA.
| |
Collapse
|
32
|
Zhang G, Wei H, Zhao A, Yan X, Zhang X, Gan J, Guo M, Wang J, Zhang F, Jiang Y, Liu X, Yang Z, Jiang X. Mitochondrial DNA leakage: underlying mechanisms and therapeutic implications in neurological disorders. J Neuroinflammation 2025; 22:34. [PMID: 39920753 PMCID: PMC11806845 DOI: 10.1186/s12974-025-03363-0] [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: 12/03/2024] [Accepted: 01/29/2025] [Indexed: 02/09/2025] Open
Abstract
Mitochondrial dysfunction is a pivotal instigator of neuroinflammation, with mitochondrial DNA (mtDNA) leakage as a critical intermediary. This review delineates the intricate pathways leading to mtDNA release, which include membrane permeabilization, vesicular trafficking, disruption of homeostatic regulation, and abnormalities in mitochondrial dynamics. The escaped mtDNA activates cytosolic DNA sensors, especially cyclic gmp-amp synthase (cGAS) signalling and inflammasome, initiating neuroinflammatory cascades via pathways, exacerbating a spectrum of neurological pathologies. The therapeutic promise of targeting mtDNA leakage is discussed in detail, underscoring the necessity for a multifaceted strategy that encompasses the preservation of mtDNA homeostasis, prevention of membrane leakage, reestablishment of mitochondrial dynamics, and inhibition the activation of cytosolic DNA sensors. Advancing our understanding of the complex interplay between mtDNA leakage and neuroinflammation is imperative for developing precision therapeutic interventions for neurological disorders.
Collapse
Affiliation(s)
- Guangming Zhang
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, No. 10, Poyang Lake Road, Tuanbo New City West District, Jinghai District, Tianjin, 301617, China
| | - Huayuan Wei
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, No. 10, Poyang Lake Road, Tuanbo New City West District, Jinghai District, Tianjin, 301617, China
| | - Anliu Zhao
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, No. 10, Poyang Lake Road, Tuanbo New City West District, Jinghai District, Tianjin, 301617, China
| | - Xu Yan
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, No. 10, Poyang Lake Road, Tuanbo New City West District, Jinghai District, Tianjin, 301617, China
| | - Xiaolu Zhang
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, No. 10, Poyang Lake Road, Tuanbo New City West District, Jinghai District, Tianjin, 301617, China
| | - Jiali Gan
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, No. 10, Poyang Lake Road, Tuanbo New City West District, Jinghai District, Tianjin, 301617, China
| | - Maojuan Guo
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, No. 10, Poyang Lake Road, Tuanbo New City West District, Jinghai District, Tianjin, 301617, China
| | - Jie Wang
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, No. 10, Poyang Lake Road, Tuanbo New City West District, Jinghai District, Tianjin, 301617, China
| | - Fayan Zhang
- Heart Disease Department, Tianjin Academy of Traditional Chinese Medicine Affiliated Hospital, Tianjin, China
| | - Yifang Jiang
- School of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Xinxing Liu
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, No. 10, Poyang Lake Road, Tuanbo New City West District, Jinghai District, Tianjin, 301617, China
| | - Zhen Yang
- School of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, No. 10, Poyang Lake Road, Tuanbo New City West District, Jinghai District, Tianjin, China.
- Tianjin Key Laboratory of Therapeutic Substance of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, No. 10, Poyang Lake Road, Tuanbo New City West District, Jinghai District, Tianjin, China.
| | - Xijuan Jiang
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, No. 10, Poyang Lake Road, Tuanbo New City West District, Jinghai District, Tianjin, 301617, China.
| |
Collapse
|
33
|
Rai P, Fessler MB. Mechanisms and effects of activation of innate immunity by mitochondrial nucleic acids. Int Immunol 2025; 37:133-142. [PMID: 39213393 DOI: 10.1093/intimm/dxae052] [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: 07/10/2024] [Accepted: 08/28/2024] [Indexed: 09/04/2024] Open
Abstract
In recent years, a growing number of roles have been identified for mitochondria in innate immunity. One principal mechanism is that the translocation of mitochondrial nucleic acid species from the mitochondrial matrix to the cytosol and endolysosomal lumen in response to an array of microbial and non-microbial environmental stressors has been found to serve as a second messenger event in the cell signaling of the innate immune response. Thus, mitochondrial DNA and RNA have been shown to access the cytosol through several regulated mechanisms involving remodeling of the mitochondrial inner and outer membranes and to access lysosomes via vesicular transport, thereby activating cytosolic [e.g. cyclic GMP-AMP synthase (cGAS), retinoic acid-inducible gene I (RIG-I)-like receptors], and endolysosomal (Toll-like receptor 7, 9) nucleic acid receptors that induce type I interferons and pro-inflammatory cytokines. In this mini-review, we discuss these molecular mechanisms of mitochondrial nucleic acid mislocalization and their roles in host defense, autoimmunity, and auto-inflammatory disorders. The emergent paradigm is one in which host-derived DNA interestingly serves as a signal amplifier in the innate immune response and also as an alarm signal for disturbances in organellar homeostasis. The apparent vast excess of mitochondria and mitochondrial DNA nucleoids per cell may thus serve to sensitize the cell response to stressors while ensuring an underlying reserve of intact mitochondria to sustain cellular metabolism. An improved understanding of these molecular mechanisms will hopefully afford future opportunities for therapeutic intervention in human disease.
Collapse
Affiliation(s)
- Prashant Rai
- Immunity, Inflammation and Disease Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC 27709, USA
| | - Michael B Fessler
- Immunity, Inflammation and Disease Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC 27709, USA
| |
Collapse
|
34
|
Miao HT, Wang J, Shao JJ, Song RX, Li WG, Sun JK, Jia SY, Zhang DX, Li XM, Zhao JY, Zhang LM. Astrocytic NLRP3 cKO mitigates depression-like behaviors induced by mild TBI in mice. Neurobiol Dis 2025; 205:106785. [PMID: 39793767 DOI: 10.1016/j.nbd.2024.106785] [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: 08/13/2024] [Revised: 12/31/2024] [Accepted: 12/31/2024] [Indexed: 01/13/2025] Open
Abstract
BACKGROUND Reports indicate that depression is a common mental health issue following traumatic brain injury (TBI). Our prior research suggests that Nucleotide-binding oligomerization domain-like receptor protein 3 (NLRP3)-related neuroinflammation, modulated by glial cells such as astrocytes, is likely to play a crucial role in the progression of anxiety and cognitive dysfunction. However, there is limited understanding of the potential of astrocytic NLRP3 in treating depression under mild TBI condition. This study aimed to determine whether astrocytic NLRP3 knockout (KO) could mitigate depressive-like behaviors following mild TBI and explore potential variations in such behaviors between genders post-mild TBI. METHODS Mild TBI was induced in mice using Feeney's weight-drop method. Behavioral assessments included neurological severity scores (NSS), social interaction test (SI), tail suspension test (TST), and forced swimming test (FST). Pathological changes were evaluated through immunofluorescence and local field potential (LFP) recordings at various time points post-injury. RESULTS Our findings indicated that astrocyte-specific NLRP3 KO decreased cleaved caspase-1 colocalized with astrocytes, decreased pathogenic astrocytes and increased Postsynaptic density protein 95 (PSD95) intensity, and significantly alleviated mild TBI-induced depression-like behaviors. It also led to the upregulation of protective astrocytes and apoptosis-associated factors, including cleaved caspase-3 post-mild TBI. Additionally, astrocyte-specific NLRP3 deletion resulting in improved θ and γ power and θ-γ phase coupling in the social interaction test (SI). Notably, under mild TBI conditions, astrocyte-specific NLRP3 exhibited greater neuroprotective effects in female knockout mice compared to males. CONCLUSION Astrocyte NLRP3 knockout demonstrated a protective mechanism in mice subjected to mild TBI, possibly attributed to the inhibition of pyroptosis through the NLRP3 signaling pathway in astrocytes.
Collapse
Affiliation(s)
- Hui-Tao Miao
- Department of Anesthesiology, Hebei Province, Cangzhou Hospital of Integrated Traditional and Western Medicine, Cangzhou, China,; Hebei Province Key Laboratory of Integrated Traditional and Western Medicine in Neurological Rehabilitation, Cangzhou, China; Hebei Key Laboratory of Integrated Traditional and Western Medicine in Osteoarthrosis Research (Preparing), Cangzhou, China
| | - Jun Wang
- Department of Orthopaedics, Tianjin Hospital, Tianjin University, Tianjin, China
| | - Jing-Jing Shao
- Department of Anesthesiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Rong-Xin Song
- Department of Anesthesiology, Hebei Province, Cangzhou Hospital of Integrated Traditional and Western Medicine, Cangzhou, China
| | - Wen-Guang Li
- Graduated School, Hebei Medical University, Shijiazhuang, China
| | - Jian-Kai Sun
- Graduated School, Hebei Medical University, Shijiazhuang, China
| | - Shi-Yan Jia
- Hebei Province Key Laboratory of Integrated Traditional and Western Medicine in Neurological Rehabilitation, Cangzhou, China
| | - Dong-Xue Zhang
- Department of Gerontology, Cangzhou Central Hospital, Cangzhou, China
| | - Xiao-Ming Li
- Hebei Key Laboratory of Integrated Traditional and Western Medicine in Osteoarthrosis Research (Preparing), Cangzhou, China
| | - Jian-Yong Zhao
- Hebei Key Laboratory of Integrated Traditional and Western Medicine in Osteoarthrosis Research (Preparing), Cangzhou, China
| | - Li-Min Zhang
- Department of Anesthesiology, Hebei Province, Cangzhou Hospital of Integrated Traditional and Western Medicine, Cangzhou, China,.
| |
Collapse
|
35
|
Guo Z, Liu Y, Chen D, Sun Y, Li D, Meng Y, Zhou Q, Zeng F, Deng G, Chen X. Targeting regulated cell death: Apoptosis, necroptosis, pyroptosis, ferroptosis, and cuproptosis in anticancer immunity. J Transl Int Med 2025; 13:10-32. [PMID: 40115032 PMCID: PMC11921819 DOI: 10.1515/jtim-2025-0004] [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] [Indexed: 03/22/2025] Open
Abstract
In the evolving landscape of cancer treatment, the strategic manipulation of regulated cell death (RCD) pathways has emerged as a crucial component of effective anti-tumor immunity. Evidence suggests that tumor cells undergoing RCD can modify the immunogenicity of the tumor microenvironment (TME), potentially enhancing its ability to suppress cancer progression and metastasis. In this review, we first explore the mechanisms of apoptosis, necroptosis, pyroptosis, ferroptosis, and cuproptosis, along with the crosstalk between these cell death modalities. We then discuss how these processes activate antigen-presenting cells, facilitate the cross-priming of CD8+ T cells, and trigger anti-tumor immune responses, highlighting the complex effects of novel forms of tumor cell death on TME and tumor biology. Furthermore, we summarize potential drugs and nanoparticles that can induce or inhibit these emerging RCD pathways and their therapeutic roles in cancer treatment. Finally, we put forward existing challenges and future prospects for targeting RCD in anti-cancer immunity. Overall, this review enhances our understanding of the molecular mechanisms and biological impacts of RCD-based therapies, providing new perspectives and strategies for cancer treatment.
Collapse
Affiliation(s)
- Ziyu Guo
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha 410008, Hunan Province, China
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Changsha 410008, Hunan Province, China
- Furong Laboratory, Changsha 410008, Hunan Province, China
- Hunan Key Laboratory of Skin Cancer and Psoriasis, Hunan Engineering Research Center of Skin Health and Disease, Xiangya Hospital, Central South University, Changsha 410008, Hunan Province, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, Hunan Province, China
| | - Yihuang Liu
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha 410008, Hunan Province, China
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Changsha 410008, Hunan Province, China
- Furong Laboratory, Changsha 410008, Hunan Province, China
- Hunan Key Laboratory of Skin Cancer and Psoriasis, Hunan Engineering Research Center of Skin Health and Disease, Xiangya Hospital, Central South University, Changsha 410008, Hunan Province, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, Hunan Province, China
| | - Danyao Chen
- Department of Thoracic Surgery, Xiangya Hospital, Central South University, Changsha 410008, Hunan Province, China
| | - Yuming Sun
- Department of Oncology, Xiangya Hospital, Central South University, Changsha 410008, Hunan Province, China
| | - Daishi Li
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha 410008, Hunan Province, China
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Changsha 410008, Hunan Province, China
- Furong Laboratory, Changsha 410008, Hunan Province, China
- Hunan Key Laboratory of Skin Cancer and Psoriasis, Hunan Engineering Research Center of Skin Health and Disease, Xiangya Hospital, Central South University, Changsha 410008, Hunan Province, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, Hunan Province, China
| | - Yu Meng
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha 410008, Hunan Province, China
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Changsha 410008, Hunan Province, China
- Furong Laboratory, Changsha 410008, Hunan Province, China
- Hunan Key Laboratory of Skin Cancer and Psoriasis, Hunan Engineering Research Center of Skin Health and Disease, Xiangya Hospital, Central South University, Changsha 410008, Hunan Province, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, Hunan Province, China
| | - Qian Zhou
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha 410008, Hunan Province, China
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Changsha 410008, Hunan Province, China
- Furong Laboratory, Changsha 410008, Hunan Province, China
- Hunan Key Laboratory of Skin Cancer and Psoriasis, Hunan Engineering Research Center of Skin Health and Disease, Xiangya Hospital, Central South University, Changsha 410008, Hunan Province, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, Hunan Province, China
| | - Furong Zeng
- Department of Plastic and Cosmetic Surgery, Xiangya Hospital, Central South University, Changsha 410008, Hunan Province, China
| | - Guangtong Deng
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha 410008, Hunan Province, China
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Changsha 410008, Hunan Province, China
- Furong Laboratory, Changsha 410008, Hunan Province, China
- Hunan Key Laboratory of Skin Cancer and Psoriasis, Hunan Engineering Research Center of Skin Health and Disease, Xiangya Hospital, Central South University, Changsha 410008, Hunan Province, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, Hunan Province, China
| | - Xiang Chen
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha 410008, Hunan Province, China
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Changsha 410008, Hunan Province, China
- Furong Laboratory, Changsha 410008, Hunan Province, China
- Hunan Key Laboratory of Skin Cancer and Psoriasis, Hunan Engineering Research Center of Skin Health and Disease, Xiangya Hospital, Central South University, Changsha 410008, Hunan Province, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, Hunan Province, China
| |
Collapse
|
36
|
Kamarulzaman NT, Makpol S. The link between Mitochondria and Sarcopenia. J Physiol Biochem 2025; 81:1-20. [PMID: 39969761 PMCID: PMC11958477 DOI: 10.1007/s13105-024-01062-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Accepted: 11/12/2024] [Indexed: 02/20/2025]
Abstract
Sarcopenia, a widespread condition, is characterized by a variety of factors influencing its development. The causes of sarcopenia differ depending on the age of the individual. It is defined as the combination of decreased muscle mass and impaired muscle function, primarily observed in association with ageing. As people age from 20 to 80 years old, there is an approximate 30% reduction in muscle mass and a 20% decline in cross-sectional area. This decline is attributed to a decrease in the size and number of muscle fibres. The regression of muscle mass and strength increases the risk of fractures, frailty, reduced quality of life, and loss of independence. Muscle cells, fibres, and tissues shrink, resulting in diminished muscle power, volume, and strength in major muscle groups. One prominent theory of cellular ageing posits a strong positive relationship between age and oxidative damage. Heightened oxidative stress leads to early-onset sarcopenia, characterized by neuromuscular innervation breakdown, muscle atrophy, and dysfunctional mitochondrial muscles. Ageing muscles generate more reactive oxygen species (ROS), and experience decreased oxygen consumption and ATP synthesis compared to younger muscles. Additionally, changes in mitochondrial protein interactions, cristae structure, and networks may contribute to ADP insensitivity, which ultimately leads to sarcopenia. Within this framework, this review provides a comprehensive summary of our current understanding of the role of mitochondria in sarcopenia and other muscle degenerative diseases, highlighting the crucial need for further research in these areas.
Collapse
Affiliation(s)
- Nurul Tihani Kamarulzaman
- Department of Biochemistry, Faculty of Medicine, Universiti Kebangsaan Malaysia, Jalan Yaacob Latif, Bandar Tun Razak, Cheras, Kuala Lumpur, 56000, Malaysia
| | - Suzana Makpol
- Department of Biochemistry, Faculty of Medicine, Universiti Kebangsaan Malaysia, Jalan Yaacob Latif, Bandar Tun Razak, Cheras, Kuala Lumpur, 56000, Malaysia.
| |
Collapse
|
37
|
Ruan J, Wei X, Li S, Ye Z, Hu L, Zhuang R, Cao Y, Wang S, Wu S, Peng D, Chen S, Yuan S, Xu A. Apaf-1 is an evolutionarily conserved DNA sensor that switches the cell fate between apoptosis and inflammation. Cell Discov 2025; 11:4. [PMID: 39833169 PMCID: PMC11747288 DOI: 10.1038/s41421-024-00750-4] [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: 06/12/2023] [Accepted: 11/04/2024] [Indexed: 01/22/2025] Open
Abstract
Apoptotic protease activating factor 1 (Apaf-1) was traditionally defined as a scaffold protein in mammalian cells for assembling a caspase activation platform known as the 'apoptosome' after its binding to cytochrome c. Although Apaf-1 structurally resembles animal NOD-like receptor (NLR) and plant resistance (R) proteins, whether it is directly involved in innate immunity is still largely unknown. Here, we found that Apaf-1-like molecules from lancelets, fruit flies, mice, and humans have conserved DNA sensing functionality. Mechanistically, mammalian Apaf-1 recruits receptor-interacting protein 2 (RIP2, also known as RIPK2) via its WD40 repeat domain and promotes RIP2 oligomerization to initiate NF-κB-driven inflammation upon cytoplasmic DNA recognition. Furthermore, DNA binding of Apaf-1 determines cell fate by switching the cellular processes between intrinsic stimuli-activated apoptosis and inflammation. These findings suggest that Apaf-1 is an evolutionarily conserved DNA sensor and may serve as a cell fate checkpoint, which determines whether cells initiate inflammation or undergo apoptosis by distinct ligand binding.
Collapse
Affiliation(s)
- Jie Ruan
- Guangdong Key Laboratory of Pharmaceutical Functional Genes, MOE Key Laboratory of Gene Function and Regulation, MOE Engineering Center of South China Sea Marine Biotechnology, Southern Laboratory of Ocean Science and Engineering (Zhuhai), State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Xuxia Wei
- Guangdong Key Laboratory of Pharmaceutical Functional Genes, MOE Key Laboratory of Gene Function and Regulation, MOE Engineering Center of South China Sea Marine Biotechnology, Southern Laboratory of Ocean Science and Engineering (Zhuhai), State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Suizhi Li
- Guangdong Key Laboratory of Pharmaceutical Functional Genes, MOE Key Laboratory of Gene Function and Regulation, MOE Engineering Center of South China Sea Marine Biotechnology, Southern Laboratory of Ocean Science and Engineering (Zhuhai), State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Zijian Ye
- Guangdong Key Laboratory of Pharmaceutical Functional Genes, MOE Key Laboratory of Gene Function and Regulation, MOE Engineering Center of South China Sea Marine Biotechnology, Southern Laboratory of Ocean Science and Engineering (Zhuhai), State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Linyi Hu
- Guangdong Key Laboratory of Pharmaceutical Functional Genes, MOE Key Laboratory of Gene Function and Regulation, MOE Engineering Center of South China Sea Marine Biotechnology, Southern Laboratory of Ocean Science and Engineering (Zhuhai), State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Ru Zhuang
- Guangdong Key Laboratory of Pharmaceutical Functional Genes, MOE Key Laboratory of Gene Function and Regulation, MOE Engineering Center of South China Sea Marine Biotechnology, Southern Laboratory of Ocean Science and Engineering (Zhuhai), State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Yange Cao
- Guangdong Key Laboratory of Pharmaceutical Functional Genes, MOE Key Laboratory of Gene Function and Regulation, MOE Engineering Center of South China Sea Marine Biotechnology, Southern Laboratory of Ocean Science and Engineering (Zhuhai), State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Shaozhou Wang
- Guangdong Key Laboratory of Pharmaceutical Functional Genes, MOE Key Laboratory of Gene Function and Regulation, MOE Engineering Center of South China Sea Marine Biotechnology, Southern Laboratory of Ocean Science and Engineering (Zhuhai), State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Shengpeng Wu
- Guangdong Key Laboratory of Pharmaceutical Functional Genes, MOE Key Laboratory of Gene Function and Regulation, MOE Engineering Center of South China Sea Marine Biotechnology, Southern Laboratory of Ocean Science and Engineering (Zhuhai), State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Dezhi Peng
- Guangdong Key Laboratory of Pharmaceutical Functional Genes, MOE Key Laboratory of Gene Function and Regulation, MOE Engineering Center of South China Sea Marine Biotechnology, Southern Laboratory of Ocean Science and Engineering (Zhuhai), State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Shangwu Chen
- Guangdong Key Laboratory of Pharmaceutical Functional Genes, MOE Key Laboratory of Gene Function and Regulation, MOE Engineering Center of South China Sea Marine Biotechnology, Southern Laboratory of Ocean Science and Engineering (Zhuhai), State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Shaochun Yuan
- Guangdong Key Laboratory of Pharmaceutical Functional Genes, MOE Key Laboratory of Gene Function and Regulation, MOE Engineering Center of South China Sea Marine Biotechnology, Southern Laboratory of Ocean Science and Engineering (Zhuhai), State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, Guangdong, China.
- Laboratory for Marine Biology and Biotechnology, Qingdao National Laboratory for Marine Science and Technology, Qingdao, Shandong, China.
| | - Anlong Xu
- Guangdong Key Laboratory of Pharmaceutical Functional Genes, MOE Key Laboratory of Gene Function and Regulation, MOE Engineering Center of South China Sea Marine Biotechnology, Southern Laboratory of Ocean Science and Engineering (Zhuhai), State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, Guangdong, China.
- Sun Yat-sen University Institute of Advanced Studies Hong Kong, Hong Kong, China.
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, China.
| |
Collapse
|
38
|
Guan X, Li H, Zhang L, Zhi H. Mechanisms of mitochondrial damage-associated molecular patterns associated with inflammatory response in cardiovascular diseases. Inflamm Res 2025; 74:18. [PMID: 39806203 DOI: 10.1007/s00011-025-01993-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: 10/20/2024] [Revised: 12/25/2024] [Accepted: 01/02/2025] [Indexed: 01/16/2025] Open
Abstract
Cardiovascular diseases (CVDs) continue to be a substantial global healthcare burden despite considerable progress in therapies. The inflammatory response during the progression of CVD has attracted considerable attention. Mitochondria serve as the principal energy source for the heart. In cardiovascular illnesses, mitochondrial homeostasis is disrupted, accompanied by structural and functional impairments. During mitochondrial stress or injury, mitochondrial damage-associated molecular patterns (mtDAMPs), such as mitochondrial DNA, cardiolipin, N-formyl peptide, and adenosine triphosphate, are released to activate pattern recognition receptors and trigger immunological responses. Inflammatory responses mediated by mtDAMPs substantially contribute to the pathophysiology of cardiovascular illnesses. In this review, we discuss the molecular mechanisms by which different mtDAMPs control the inflammatory response, address the pathological consequences of mtDAMPs in inducing or exacerbating the inflammatory response in CVDs, and summarize potential therapeutic targets in relevant experimental studies. Preventing or reducing mtDAMP release may play a role in CVD progression by alleviating the inflammatory response.
Collapse
Affiliation(s)
- Xiuju Guan
- School of Graduate Studies, Tianjin University of Traditional Chinese Medicine, Tianjin, People's Republic of China
| | - Haitao Li
- Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, 250014, Shandong, People's Republic of China
| | - Lijuan Zhang
- Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, 250014, Shandong, People's Republic of China.
| | - Hongwei Zhi
- Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, 250014, Shandong, People's Republic of China.
| |
Collapse
|
39
|
Moyer A, Tanaka K, Cheng EH. Apoptosis in Cancer Biology and Therapy. ANNUAL REVIEW OF PATHOLOGY 2025; 20:303-328. [PMID: 39854189 DOI: 10.1146/annurev-pathmechdis-051222-115023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/26/2025]
Abstract
Since its inception, the study of apoptosis has been intricately linked to the field of cancer. The term apoptosis was coined more than five decades ago following its identification in both healthy tissues and malignant neoplasms. The subsequent elucidation of its molecular mechanisms has significantly enhanced our understanding of how cancer cells hijack physiological processes to evade cell death. Moreover, it has shed light on the pathways through which most anticancer therapeutics induce tumor cell death, including targeted therapy and immunotherapy. These mechanistic studies have paved the way for the development of therapeutics directly targeting either pro- or antiapoptotic proteins. Notably, the US Food and Drug Administration (FDA) approved the BCL-2 inhibitor venetoclax in 2016, with additional agents currently undergoing clinical trials. Recent research has brought to the forefront both the anti- and proinflammatory effects of individual apoptotic pathways. This underscores the ongoing imperative to deepen our comprehension of apoptosis, particularly as we navigate the evolving landscape of immunotherapy.
Collapse
Affiliation(s)
- Allison Moyer
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA;
- Tri-Institutional MD-PhD Program, Weill Cornell Medicine, New York, NY, USA
| | - Kosuke Tanaka
- Division of Cancer Immunology, National Cancer Center Research Institute, Tokyo, Japan
- Division of Cancer Immunology, Exploratory Oncology Research and Clinical Trial Center, National Cancer Center, Chiba, Japan
| | - Emily H Cheng
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA;
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY, USA
| |
Collapse
|
40
|
Larrañaga-SanMiguel A, Bengoa-Vergniory N, Flores-Romero H. Crosstalk between mitochondria-ER contact sites and the apoptotic machinery as a novel health meter. Trends Cell Biol 2025; 35:33-45. [PMID: 39379268 DOI: 10.1016/j.tcb.2024.08.007] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Revised: 08/27/2024] [Accepted: 08/29/2024] [Indexed: 10/10/2024]
Abstract
Mitochondria-endoplasmic reticulum (ER) contact sites (MERCS) function as transient signaling platforms that regulate essential cellular functions. MERCS are enriched in specific proteins and lipids that connect mitochondria and the ER together and modulate their activities. Dysregulation of MERCS is associated with several human pathologies including Alzheimer's disease (AD), Parkinson's disease (PD), and cancer. BCL-2 family proteins can locate at MERCS and control essential cellular functions such as calcium signaling and autophagy in addition to their role in mitochondrial apoptosis. Moreover, the BCL-2-mediated apoptotic machinery was recently found to trigger cGAS-STING pathway activation and a proinflammatory response, a recognized hallmark of these diseases that requires mitochondria-ER interplay. This review underscores the pivotal role of MERCS in regulating essential cellular functions, focusing on their crosstalk with BCL-2 family proteins, and discusses how their dysregulation is linked to disease.
Collapse
Affiliation(s)
| | - Nora Bengoa-Vergniory
- Achucarro Basque Center for Neuroscience, Leioa, Spain; Ikerbasque, Basque Foundation for Science, 48013 Bilbao, Spain; Oxford Parkinson's Disease Centre and Department of Physiology, Anatomy, and Genetics, University of Oxford, South Parks Road, Oxford OX1 3QU, UK; Department of Neuroscience, University of the Basque Country (UPV/EHU), Leioa, Spain
| | - Hector Flores-Romero
- Achucarro Basque Center for Neuroscience, Leioa, Spain; Ikerbasque, Basque Foundation for Science, 48013 Bilbao, Spain.
| |
Collapse
|
41
|
Glytsou C. Electron Microscopy to Visualize and Quantify Mitochondrial Structure and Organellar Interactions in Cultured Cells During Senescence. Methods Mol Biol 2025; 2906:229-242. [PMID: 40082359 DOI: 10.1007/978-1-0716-4426-3_13] [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: 03/16/2025]
Abstract
Mitochondria are multifunctional organelles that play a crucial role in numerous cellular processes, including oncogene-induced senescence. Recent studies have demonstrated that mitochondria undergo notable morphological and functional changes during senescence, with mitochondria dysregulation being a critical factor contributing to the induction of this state. To elucidate the intricate and dynamic structure of these organelles, high-resolution visualization techniques are imperative. Electron microscopy offers nanometer-scale resolution images, enabling the comprehensive study of organelles' architecture. This chapter provides a detailed guide for preparing fixed samples from cultured cells for electron microscopy imaging. It also describes various quantification methods to accurately assess organellar parameters, including morphometric measurements of mitochondrial shape, cristae structure, and mitochondria-endoplasmic reticulum contact sites. These analyses yield valuable insights into the status of subcellular organelles, advancing our understanding of their involvement in cellular senescence and disease.
Collapse
Affiliation(s)
- Christina Glytsou
- Department of Chemical Biology, Ernest Mario School of Pharmacy, Rutgers, The State University of New Jersey, Piscataway, NJ, USA.
- Department of Pediatrics at Robert Wood Johnson Medical School, Rutgers, The State University of New Jersey, New Brunswick, NJ, USA.
- Rutgers Cancer Institute, New Brunswick, NJ, USA.
| |
Collapse
|
42
|
Bhattacharya M, Bhowmik D, Yin Q. In Vitro Cleavage Assay to Characterize DENV NS2B3 Antagonism of cGAS. Methods Mol Biol 2025; 2854:153-170. [PMID: 39192127 DOI: 10.1007/978-1-0716-4108-8_16] [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: 08/29/2024]
Abstract
cGAS is a key cytosolic dsDNA receptor that senses viral infection and elicits interferon production through the cGAS-cGAMP-STING axis. cGAS is activated by dsDNA from viral and bacterial origins as well as dsDNA leaked from damaged mitochondria and nucleus. Eventually, cGAS activation launches the cell into an antiviral state to restrict the replication of both DNA and RNA viruses. Throughout the long co-evolution, viruses devise many strategies to evade cGAS detection or suppress cGAS activation. We recently reported that the Dengue virus protease NS2B3 proteolytically cleaves human cGAS in its N-terminal region, effectively reducing cGAS binding to DNA and consequent production of the second messenger cGAMP. Several other RNA viruses likely adopt the cleavage strategy. Here, we describe a protocol for the purification of recombinant human cGAS and Dengue NS2B3 protease, as well as the in vitro cleavage assay.
Collapse
Affiliation(s)
| | - Debipreeta Bhowmik
- Department of Biological Science, Florida State University, Tallahassee, FL, USA
| | - Qian Yin
- Institute of Molecular Biophysics, Florida State University, Tallahassee, FL, USA.
- Department of Biological Science, Florida State University, Tallahassee, FL, USA.
| |
Collapse
|
43
|
Szmyd R, Casolin S, French L, Manjón AG, Walter M, Cavalli L, Nelson CB, Page SG, Dhawan A, Hau E, Pickett HA, Gee HE, Cesare AJ. Homologous recombination promotes non-immunogenic mitotic cell death upon DNA damage. Nat Cell Biol 2025; 27:59-72. [PMID: 39805921 PMCID: PMC11735404 DOI: 10.1038/s41556-024-01557-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: 09/06/2023] [Accepted: 10/14/2024] [Indexed: 01/16/2025]
Abstract
Double-strand breaks (DSBs) can initiate mitotic catastrophe, a complex oncosuppressive phenomenon characterized by cell death during or after cell division. Here we unveil how cell cycle-regulated DSB repair guides disparate cell death outcomes through single-cell analysis of extended live imaging. Following DSB induction in S or G2, passage of unresolved homologous recombination intermediates into mitosis promotes non-immunogenic intrinsic apoptosis in the immediate attempt at cell division. Conversely, non-homologous end joining, microhomology-mediated end joining and single-strand annealing cooperate to enable damaged G1 cells to complete the first cell cycle with an aberrant cell division at the cost of delayed extrinsic lethality and interferon production. Targeting non-homologous end joining, microhomology-mediated end joining or single-strand annealing promotes mitotic death, while suppressing mitotic death enhances interferon production. Together the data indicate that a temporal repair hierarchy, coupled with cumulative DSB load, serves as a reliable predictor of mitotic catastrophe outcomes following genome damage. In this pathway, homologous recombination suppresses interferon production by promoting mitotic lethality.
Collapse
Affiliation(s)
- Radoslaw Szmyd
- Genome Integrity Unit, Children's Medical Research Institute, University of Sydney, Westmead, New South Wales, Australia
- Radiation Oncology Network, Western Sydney Local Health District, Sydney, New South Wales, Australia
| | - Sienna Casolin
- Genome Integrity Unit, Children's Medical Research Institute, University of Sydney, Westmead, New South Wales, Australia
- Radiation Oncology Network, Western Sydney Local Health District, Sydney, New South Wales, Australia
| | - Lucy French
- Genome Integrity Unit, Children's Medical Research Institute, University of Sydney, Westmead, New South Wales, Australia
- Radiation Oncology Network, Western Sydney Local Health District, Sydney, New South Wales, Australia
| | - Anna G Manjón
- Genome Integrity Unit, Children's Medical Research Institute, University of Sydney, Westmead, New South Wales, Australia
- Radiation Oncology Network, Western Sydney Local Health District, Sydney, New South Wales, Australia
| | - Melanie Walter
- Genome Integrity Unit, Children's Medical Research Institute, University of Sydney, Westmead, New South Wales, Australia
- Radiation Oncology Network, Western Sydney Local Health District, Sydney, New South Wales, Australia
| | - Léa Cavalli
- Genome Integrity Unit, Children's Medical Research Institute, University of Sydney, Westmead, New South Wales, Australia
| | - Christopher B Nelson
- Telomere Length Regulation Unit, Children's Medical Research Institute, University of Sydney, Westmead, New South Wales, Australia
| | - Scott G Page
- Genome Integrity Unit, Children's Medical Research Institute, University of Sydney, Westmead, New South Wales, Australia
| | - Andrew Dhawan
- Neurological Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Eric Hau
- Radiation Oncology Network, Western Sydney Local Health District, Sydney, New South Wales, Australia
- Westmead Clinical School, University of Sydney, Westmead, New South Wales, Australia
- Translational Radiation Biology and Oncology Laboratory, Centre for Cancer Research, The Westmead Institute for Medical Research, Westmead, New South Wales, Australia
| | - Hilda A Pickett
- Telomere Length Regulation Unit, Children's Medical Research Institute, University of Sydney, Westmead, New South Wales, Australia
| | - Harriet E Gee
- Genome Integrity Unit, Children's Medical Research Institute, University of Sydney, Westmead, New South Wales, Australia.
- Radiation Oncology Network, Western Sydney Local Health District, Sydney, New South Wales, Australia.
- Westmead Clinical School, University of Sydney, Westmead, New South Wales, Australia.
- Translational Radiation Biology and Oncology Laboratory, Centre for Cancer Research, The Westmead Institute for Medical Research, Westmead, New South Wales, Australia.
| | - Anthony J Cesare
- Genome Integrity Unit, Children's Medical Research Institute, University of Sydney, Westmead, New South Wales, Australia.
| |
Collapse
|
44
|
Li Y, Wei J, Ye Z, Ji C, Li W, Xu L, Zhou Z. Essential Oils from Citrus Peels Promote Calcium Overload-Induced Calcicoptosis in U251 Cells. Antioxidants (Basel) 2024; 14:11. [PMID: 39857347 PMCID: PMC11762846 DOI: 10.3390/antiox14010011] [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: 12/03/2024] [Revised: 12/22/2024] [Accepted: 12/23/2024] [Indexed: 01/27/2025] Open
Abstract
Citrus peel essential oils (CPEOs) have demonstrated substantial medicinal potential for glioblastoma treatment because of their extensive antitumor effects, low potential for drug resistance, and ability to cross the human blood-brain barrier. In this study, the chemical compositions of five CPEOs were analyzed via gas chromatography-mass spectrometry (GC-MS). CCK8 assays were used to evaluate the ability of five CPEOs to inhibit U251 human glioblastoma cells, and XLB and RA were selected for further investigation. Through wound healing assays and cell cycle and apoptosis analyses via flow cytometry, it was revealed that these CPEOs inhibited cell migration, arrested the cell cycle at G1/G0, and induced apoptosis with similar levels of inhibition. After CPEOs treatment, the intracellular Ca2+ content and reactive oxygen species levels in U251 cells increased significantly, whereas the mitochondrial membrane potential decreased. Additionally, the antioxidant enzyme system (SOD, POD, CAT, and GR) and the nonenzymatic defense system (GSH) were inhibited, leading to an increase in lipid peroxidation. qRT-PCR indicated the significant upregulation of intracellular calcium ion signaling pathways and the upregulation of mitochondrial apoptosis-related genes. Additionally, the activation of calcicoptosis-related indicators induced by the CPEOs could be reversed by inhibitor treatment, confirming that both of the selected CPEOs inhibit tumors by activating calcicoptosis-related pathways. These findings highlight the immense potential of CPEOs in healthcare and pharmaceutical applications by not only providing a scientific basis for the potential application of CPEOs in the treatment of glioblastoma but also offering new insights for the development of novel antitumor drugs.
Collapse
Affiliation(s)
- Yurong Li
- Key Laboratory of Agricultural Biosafety and Green Production of Upper Yangtze River (Ministry of Education), College of Horticulture and Landscape Architecture, Southwest University, Beibei District, Chongqing 400715, China; (Y.L.); (J.W.); (Z.Y.); (C.J.)
| | - Juanjuan Wei
- Key Laboratory of Agricultural Biosafety and Green Production of Upper Yangtze River (Ministry of Education), College of Horticulture and Landscape Architecture, Southwest University, Beibei District, Chongqing 400715, China; (Y.L.); (J.W.); (Z.Y.); (C.J.)
| | - Zimao Ye
- Key Laboratory of Agricultural Biosafety and Green Production of Upper Yangtze River (Ministry of Education), College of Horticulture and Landscape Architecture, Southwest University, Beibei District, Chongqing 400715, China; (Y.L.); (J.W.); (Z.Y.); (C.J.)
| | - Chen Ji
- Key Laboratory of Agricultural Biosafety and Green Production of Upper Yangtze River (Ministry of Education), College of Horticulture and Landscape Architecture, Southwest University, Beibei District, Chongqing 400715, China; (Y.L.); (J.W.); (Z.Y.); (C.J.)
| | - Wenji Li
- School of Design, Chongqing Industry Polytechnic College, Chongqing 401120, China;
| | - Li Xu
- College of Sericulture, Textile and Biomass Sciences, Southwest University, Beibei District, Chongqing 400715, China
| | - Zhiqin Zhou
- Key Laboratory of Agricultural Biosafety and Green Production of Upper Yangtze River (Ministry of Education), College of Horticulture and Landscape Architecture, Southwest University, Beibei District, Chongqing 400715, China; (Y.L.); (J.W.); (Z.Y.); (C.J.)
- Southwest Institute of Fruits Nutrition, Banan District, Chongqing 400054, China
| |
Collapse
|
45
|
Paludan SR, Pradeu T, Pichlmair A, Wray KB, Mikkelsen JG, Olagnier D, Mogensen TH. Early host defense against virus infections. Cell Rep 2024; 43:115070. [PMID: 39675007 DOI: 10.1016/j.celrep.2024.115070] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Revised: 10/25/2024] [Accepted: 11/22/2024] [Indexed: 12/17/2024] Open
Abstract
Early host defense eliminates many viruses before infections are established while clearing others so they remain subclinical or cause only mild disease. The field of immunology has been shaped by broad concepts, including the pattern recognition theory that currently dominates innate immunology. Focusing on early host responses to virus infections, we analyze the literature to build a working hypothesis for the principles that govern the early line of cellular antiviral defense. Aiming to ultimately arrive at a criteria-based theory with strong explanatory power, we propose that both controlling infection and limiting inflammation are key drivers for the early cellular antiviral response. This response, which we suggest is exerted by a set of "microbe- and inflammation-restricting mechanisms," directly restrict viral replication while also counteracting inflammation. Exploring the mechanisms and physiological importance of the early layer of cellular antiviral defense may open further lines of research in immunology.
Collapse
Affiliation(s)
- Søren R Paludan
- Department of Biomedicine, Aarhus University, Aarhus, Denmark; Center for Immunology of Viral Infections, Aarhus University, Aarhus, Denmark; Department of Rheumatology and Inflammation Research, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden.
| | - Thomas Pradeu
- CNRS UMR 5164 ImmunoConcept, University of Bordeaux, Bordeaux, France; Department of Biological and Medical Sciences, University of Bordeaux, Bordeaux, France; Chapman University, Orange, CA, USA
| | - Andreas Pichlmair
- Center for Immunology of Viral Infections, Aarhus University, Aarhus, Denmark; Technical University of Munich, School of Medicine, Institute of Virology, Munich, Germany; German Center for Infection Research (DZIF), Partner Site Munich, Munich, Germany
| | - K Brad Wray
- Center for Immunology of Viral Infections, Aarhus University, Aarhus, Denmark; Centre for Science Studies, Aarhus University, Aarhus, Denmark; Aarhus Institute of Advanced Studies, Aarhus University, Aarhus, Denmark
| | - Jacob Giehm Mikkelsen
- Department of Biomedicine, Aarhus University, Aarhus, Denmark; Center for Immunology of Viral Infections, Aarhus University, Aarhus, Denmark
| | - David Olagnier
- Department of Biomedicine, Aarhus University, Aarhus, Denmark; Center for Immunology of Viral Infections, Aarhus University, Aarhus, Denmark
| | - Trine H Mogensen
- Department of Biomedicine, Aarhus University, Aarhus, Denmark; Center for Immunology of Viral Infections, Aarhus University, Aarhus, Denmark; Department of Infectious Diseases, Aarhus University Hospital, Aarhus, Denmark
| |
Collapse
|
46
|
Chen F, Xu K, Han Y, Ding J, Ren J, Wang Y, Ma Z, Cao F. Mitochondrial dysfunction in pancreatic acinar cells: mechanisms and therapeutic strategies in acute pancreatitis. Front Immunol 2024; 15:1503087. [PMID: 39776917 PMCID: PMC11703726 DOI: 10.3389/fimmu.2024.1503087] [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: 09/28/2024] [Accepted: 12/09/2024] [Indexed: 01/11/2025] Open
Abstract
Acute pancreatitis (AP) is an inflammatory disease of the pancreas and a complex process involving multiple factors, with mitochondrial damage playing a crucial role. Mitochondrial dysfunction is now considered a key driver in the development of AP. This dysfunction often presents as increased oxidative stress, altered membrane potential and permeability, and mitochondrial DNA damage and mutations. Under stress conditions, mitochondrial dynamics and mitochondrial ROS production increase, leading to decreased mitochondrial membrane potential, imbalanced calcium homeostasis, and activation of the mitochondrial permeability transition pore. The release of mitochondrial DNA (mtDNA), recognized as damage-associated molecular patterns, can activate the cGAS-STING1 and NF-κB pathway and induce pro-inflammatory factor expression. Additionally, mtDNA can activate inflammasomes, leading to interleukin release and subsequent tissue damage and inflammation. This review summarizes the relationship between mitochondria and AP and explores mitochondrial protective strategies in the diagnosis and treatment of this disease. Future research on the treatment of acute pancreatitis can benefit from exploring promising avenues such as antioxidants, mitochondrial inhibitors, and new therapies that target mitochondrial dysfunction.
Collapse
Affiliation(s)
- Fan Chen
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Kedong Xu
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
- Pancreatic Disease Center of Xi’an Jiaotong University, Xi’an, China
| | - Yimin Han
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Jiachun Ding
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Jiaqiang Ren
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Yaochun Wang
- Center for Translational Medicine, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Zhenhua Ma
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
- Pancreatic Disease Center of Xi’an Jiaotong University, Xi’an, China
| | - Fang Cao
- Center for Translational Medicine, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| |
Collapse
|
47
|
Liu Y, Guo L, Zhang G, Sun W, Yang X, Liu Y. Nogo-A exacerbates sepsis-associated encephalopathy by modulating microglial SHP-2/NLRP3 balance and inducing ROS and M1 polarization. BIOMOLECULES & BIOMEDICINE 2024; 25:210-225. [PMID: 39151100 PMCID: PMC11647263 DOI: 10.17305/bb.2024.10822] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/07/2024] [Revised: 08/12/2024] [Accepted: 08/12/2024] [Indexed: 08/18/2024]
Abstract
Sepsis, a systemic inflammatory response caused by infection, can lead to sepsis-associated encephalopathy (SAE), characterized by brain dysfunction without direct central nervous system infection. The pathogenesis of SAE involves blood-brain barrier disruption, neuroinflammation and neuronal death, with neuroinflammation being the core process. Nogo-A, a neurite growth-inhibitory protein in the central nervous system, is not well understood in sepsis. This study explores Nogo-A's mechanisms in sepsis, focusing on SAE. Using in vivo and in vitro methods, healthy SPF C57BL/6J male mice were divided into Sham, Nogo-A-NC-Model, and Nogo-A-KD-Model groups, with sepsis induced by abdominal ligation and puncture. Morris water maze tests assessed learning and memory, and brain tissues underwent hematoxylin-eosin (HE) staining, Nissl staining, and Western blot analysis. In vitro, Nogo-A gene knockdown models were constructed using BV-2 microglia cells to study inflammation and oxidative stress. Results showed Nogo-A expression affected learning and memory in septic mice, with knockdown reducing neuronal damage. Bioinformatics analysis suggested Nogo-A may activate reactive oxygen species (ROS) to inhibit p-SHP2, activating mitochondrial autophagy and promoting neuronal apoptosis. Western blot results confirmed that Nogo-A affects mitochondrial autophagy and neuronal survival by inhibiting SHP2 and activating ROS. Nogo-A's role in neuroinflammation and neuroprotection was emphasized, revealing its impact on endoplasmic reticulum (ER) stress, mitochondrial autophagy, and NLRP3 inflammasome activation. This study provides a theoretical basis for SAE treatment, suggesting further multi-gene and multi-pathway analyses and validation in clinical samples. Developing gene therapy and drug interventions targeting Nogo-A pathways will offer more effective treatment strategies.
Collapse
Affiliation(s)
- Ying Liu
- Science and Technology Experiment Center, Cangzhou Medical College, Cangzhou, China
| | - Lei Guo
- Department of Clinical Laboratory, Cangzhou Central Hospital, Cangzhou, China
| | - Guoan Zhang
- University Nanobody Application Technology Research and Development Center of Hebei Provice, Cangzhou, China
| | - Wenjie Sun
- Science and Technology Experiment Center, Cangzhou Medical College, Cangzhou, China
| | - Xiaohui Yang
- University Nanobody Application Technology Research and Development Center of Hebei Provice, Cangzhou, China
| | - Yingfu Liu
- University Nanobody Application Technology Research and Development Center of Hebei Provice, Cangzhou, China
| |
Collapse
|
48
|
Qin Y, Gao Y, Wu D, Liu QQ, Su C, Liu G, Yang L, Zhao MG, Yao JY. Ailanthone blocks mitophagy to promote mtDNA leakage through BAX-BAK1 pores and suppress hepatocellular carcinoma cell proliferation. Front Pharmacol 2024; 15:1509482. [PMID: 39723259 PMCID: PMC11668963 DOI: 10.3389/fphar.2024.1509482] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2024] [Accepted: 11/25/2024] [Indexed: 12/28/2024] Open
Abstract
Introduction Hepatocellular carcinoma (HCC), the third leading cancer mortality worldwide, shows rising incidence. The mitochondria in HCC cells are prone to damage from metabolic stress and oxidative stress, necessitating heightened mitophagy for mitochondrial homeostasis and cell survival. Thus, mitophagy inhibition is a promising HCC therapy. The traditional Chinese medicinal herb ailanthone have proved promote mitochondrial dysfunction and inhibits HCC. However, the underlying mechanism remains unclear. Methods CCK8 assay was applied to detect the proliferation. JC-1, MitoTracker Red/Green and MitoSOX staining were applied to detect the mitochondrial homeostasis. Inflammatory factors were analysed via ELISA and WB assay. Mitochondria and cytoplasm separation, genome extraction and qPCR were used to detect mitochondrial DNA (mtDNA) leakage. Mitochondria ultrastructure was detected by transmission electron microscopy. WB and IHC experiments were applied to detect protein expression. Protein-protein interactions detected by immunoprecipitation and immunofluorescence imaging. The in vivo antitumor effect was validated by the xenograft mouse model. Results In this study, we demonstrated the potent anti-HCC properties of ailanthone and revealed its molecular mechanism. In vitro studies demonstrated that ailanthone effectively inhibited PINK1-PRKN mediated mitophagy and promoted BAX-BAK1 mitochondrial pores formation through PRKN inhibition. This process led to the mitochondrial mtDNA leakage into the cytoplasm, which subsequently triggered the induction of inflammatory factors. The inhibition of mitophagy and the activation of inflammatory response ultimately led to HCC proliferation inhibition. In vivo studies demonstrated that ailanthone exhibited stronger anti-HCC activity than 5-Fluorouracil (5-FU), with no significant adverse effects on animal body weight or the physiological functions of vital organs. Conclusion This study highlighted the efficacy of ailanthone against HCC and elucidated its underlying molecular mechanisms, suggesting the promising therapeutic potential of ailanthone for HCC.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Le Yang
- Precision Pharmacy and Drug Development Center, Department of Pharmacy, Tangdu Hospital, Air Force Medical University, Xi’an, Shaanxi, China
| | - Ming-Gao Zhao
- Precision Pharmacy and Drug Development Center, Department of Pharmacy, Tangdu Hospital, Air Force Medical University, Xi’an, Shaanxi, China
| | - Jing-Yue Yao
- Precision Pharmacy and Drug Development Center, Department of Pharmacy, Tangdu Hospital, Air Force Medical University, Xi’an, Shaanxi, China
| |
Collapse
|
49
|
He R, Liu Y, Fu W, He X, Liu S, Xiao D, Tao Y. Mechanisms and cross-talk of regulated cell death and their epigenetic modifications in tumor progression. Mol Cancer 2024; 23:267. [PMID: 39614268 PMCID: PMC11606237 DOI: 10.1186/s12943-024-02172-y] [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: 08/21/2024] [Accepted: 11/07/2024] [Indexed: 12/01/2024] Open
Abstract
Cell death is a fundamental part of life for metazoans. To maintain the balance between cell proliferation and metabolism of human bodies, a certain number of cells need to be removed regularly. Hence, the mechanisms of cell death have been preserved during the evolution of multicellular organisms. Tumorigenesis is closely related with exceptional inhibition of cell death. Mutations or defects in cell death-related genes block the elimination of abnormal cells and enhance the resistance of malignant cells to chemotherapy. Therefore, the investigation of cell death mechanisms enables the development of drugs that directly induce tumor cell death. In the guidelines updated by the Cell Death Nomenclature Committee (NCCD) in 2018, cell death was classified into 12 types according to morphological, biochemical and functional classification, including intrinsic apoptosis, extrinsic apoptosis, mitochondrial permeability transition (MPT)-driven necrosis, necroptosis, ferroptosis, pyroptosis, PARP-1 parthanatos, entotic cell death, NETotic cell death, lysosome-dependent cell death, autophagy-dependent cell death, immunogenic cell death, cellular senescence and mitotic catastrophe. The mechanistic relationships between epigenetic controls and cell death in cancer progression were previously unclear. In this review, we will summarize the mechanisms of cell death pathways and corresponding epigenetic regulations. Also, we will explore the extensive interactions between these pathways and discuss the mechanisms of cell death in epigenetics which bring benefits to tumor therapy.
Collapse
Affiliation(s)
- Ruimin He
- Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, 410078, China
- Cancer Research Institute and School of Basic Medicine, Central South University, Changsha, Hunan, 410078, China
- Department of Pathology, Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Xiangya Hospital, Central South University, Hunan, 410078, China
| | - Yifan Liu
- Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, 410078, China
- Cancer Research Institute and School of Basic Medicine, Central South University, Changsha, Hunan, 410078, China
- Department of Pathology, Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Xiangya Hospital, Central South University, Hunan, 410078, China
| | - Weijie Fu
- Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, 410078, China
- Cancer Research Institute and School of Basic Medicine, Central South University, Changsha, Hunan, 410078, China
- Department of Pathology, Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Xiangya Hospital, Central South University, Hunan, 410078, China
| | - Xuan He
- Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, 410078, China
- Cancer Research Institute and School of Basic Medicine, Central South University, Changsha, Hunan, 410078, China
- Department of Pathology, Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Xiangya Hospital, Central South University, Hunan, 410078, China
| | - Shuang Liu
- Department of Oncology, Institute of Medical Sciences, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China.
| | - Desheng Xiao
- Department of Pathology, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China.
| | - Yongguang Tao
- Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, 410078, China.
- Cancer Research Institute and School of Basic Medicine, Central South University, Changsha, Hunan, 410078, China.
- Department of Pathology, Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Xiangya Hospital, Central South University, Hunan, 410078, China.
- Department of Pathology, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China.
- Department of Thoracic Surgery, Hunan Key Laboratory of Early Diagnosis and Precision Therapy in Lung Cancer, Second Xiangya Hospital, Central South University, Changsha, 410011, China.
- Furong Laboratory, Xiangya School of Medicine, Central South University, Hunan, 410078, China.
| |
Collapse
|
50
|
Zhang X, Tang B, Luo J, Yang Y, Weng Q, Fang S, Zhao Z, Tu J, Chen M, Ji J. Cuproptosis, ferroptosis and PANoptosis in tumor immune microenvironment remodeling and immunotherapy: culprits or new hope. Mol Cancer 2024; 23:255. [PMID: 39543600 PMCID: PMC11566504 DOI: 10.1186/s12943-024-02130-8] [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: 07/14/2024] [Accepted: 09/19/2024] [Indexed: 11/17/2024] Open
Abstract
Normal life requires cell division to produce new cells, but cell death is necessary to maintain balance. Dysregulation of cell death can lead to the survival and proliferation of abnormal cells, promoting tumor development. Unlike apoptosis, necrosis, and autophagy, the newly recognized forms of regulated cell death (RCD) cuproptosis, ferroptosis, and PANoptosis provide novel therapeutic strategies for tumor treatment. Increasing research indicates that the death of tumor and immune cells mediated by these newly discovered forms of cell death can regulate the tumor microenvironment (TME) and influence the effectiveness of tumor immunotherapy. This review primarily elucidates the molecular mechanisms of cuproptosis, ferroptosis, and PANoptosis and their complex effects on tumor cells and the TME. This review also summarizes the exploration of nanoparticle applications in tumor therapy based on in vivo and in vitro evidence derived from the induction or inhibition of these new RCD pathways.
Collapse
Affiliation(s)
- Xiaojie Zhang
- Zhejiang Key Laboratory of Imaging and Interventional Medicine, Zhejiang Engineering Research Csaenter of Interventional Medicine Engineering and Biotechnology, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui, Zhejiang, 323000, China
| | - Bufu Tang
- Zhejiang Key Laboratory of Imaging and Interventional Medicine, Zhejiang Engineering Research Csaenter of Interventional Medicine Engineering and Biotechnology, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui, Zhejiang, 323000, China
- Department of Radiation Oncology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Jinhua Luo
- Zhejiang Key Laboratory of Imaging and Interventional Medicine, Zhejiang Engineering Research Csaenter of Interventional Medicine Engineering and Biotechnology, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui, Zhejiang, 323000, China
| | - Yang Yang
- Zhejiang Key Laboratory of Imaging and Interventional Medicine, Zhejiang Engineering Research Csaenter of Interventional Medicine Engineering and Biotechnology, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui, Zhejiang, 323000, China
- Key Laboratory of Precision Medicine of Lishui City, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui, 323000, China
- School of Medcine, Clinical College of The Affiliated Central Hospital, Lishui University, Lishui, 323000, China
| | - Qiaoyou Weng
- Zhejiang Key Laboratory of Imaging and Interventional Medicine, Zhejiang Engineering Research Csaenter of Interventional Medicine Engineering and Biotechnology, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui, Zhejiang, 323000, China
- Key Laboratory of Precision Medicine of Lishui City, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui, 323000, China
- School of Medcine, Clinical College of The Affiliated Central Hospital, Lishui University, Lishui, 323000, China
| | - Shiji Fang
- Zhejiang Key Laboratory of Imaging and Interventional Medicine, Zhejiang Engineering Research Csaenter of Interventional Medicine Engineering and Biotechnology, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui, Zhejiang, 323000, China
- Key Laboratory of Precision Medicine of Lishui City, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui, 323000, China
- School of Medcine, Clinical College of The Affiliated Central Hospital, Lishui University, Lishui, 323000, China
| | - Zhongwei Zhao
- Zhejiang Key Laboratory of Imaging and Interventional Medicine, Zhejiang Engineering Research Csaenter of Interventional Medicine Engineering and Biotechnology, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui, Zhejiang, 323000, China
- Key Laboratory of Precision Medicine of Lishui City, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui, 323000, China
- School of Medcine, Clinical College of The Affiliated Central Hospital, Lishui University, Lishui, 323000, China
| | - Jianfei Tu
- Zhejiang Key Laboratory of Imaging and Interventional Medicine, Zhejiang Engineering Research Csaenter of Interventional Medicine Engineering and Biotechnology, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui, Zhejiang, 323000, China.
- Key Laboratory of Precision Medicine of Lishui City, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui, 323000, China.
- School of Medcine, Clinical College of The Affiliated Central Hospital, Lishui University, Lishui, 323000, China.
| | - Minjiang Chen
- Zhejiang Key Laboratory of Imaging and Interventional Medicine, Zhejiang Engineering Research Csaenter of Interventional Medicine Engineering and Biotechnology, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui, Zhejiang, 323000, China.
- Key Laboratory of Precision Medicine of Lishui City, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui, 323000, China.
- School of Medcine, Clinical College of The Affiliated Central Hospital, Lishui University, Lishui, 323000, China.
| | - Jiansong Ji
- Zhejiang Key Laboratory of Imaging and Interventional Medicine, Zhejiang Engineering Research Csaenter of Interventional Medicine Engineering and Biotechnology, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui, Zhejiang, 323000, China.
- Key Laboratory of Precision Medicine of Lishui City, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui, 323000, China.
- School of Medcine, Clinical College of The Affiliated Central Hospital, Lishui University, Lishui, 323000, China.
| |
Collapse
|