51
|
Pan X, Zhu R, Pei J, Zhang L. Lycopene: A potent antioxidant to alleviate kidney disease. Int Immunopharmacol 2025; 151:114363. [PMID: 40022820 DOI: 10.1016/j.intimp.2025.114363] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2025] [Revised: 02/19/2025] [Accepted: 02/21/2025] [Indexed: 03/04/2025]
Abstract
Kidney disease is a major public health problem worldwide and one of the common causes of death in patients. How to effectively treat or alleviate renal diseases has not only attracted the attention of a large number of scholars, but is likewise a great challenge. Oxidative stress is the main mechanism leading to kidney injury. Natural antioxidant substances not only have efficient antioxidant capacity, but also have the advantage of high safety and low side effects. Lycopene is a naturally occurring carotenoid found mainly in tomatoes or tomato products. Epidemiologic investigations have shown that lycopene has potent antioxidant properties, scavenges reactive substances, and has a protective role in kidney disease. This paper summarizes the biochemical properties and antioxidant mechanisms of lycopene in the context of animal experiments and clinical studies of lycopene in renal diseases. We found that lycopene exerts its protective effects against kidney injury mainly through anti-oxidative stress and anti-inflammatory effects. Meanwhile, lycopene has been found to reduce the incidence of Chronic Kidney Disease (CKD) as well as mortality in some CKD patients. In addition to this, lycopene intake is negatively correlated with the incidence of kidney cancer and also mitigates the nephrotoxic damage of cisplatin. Therefore, lycopene has a promising application in the treatment of kidney diseases. However, there are relatively few clinical studies on lycopene in renal diseases, and subsequent research studies in large-scale populations are still needed to determine the value of lycopene in renal diseases even further.
Collapse
Affiliation(s)
- Xingyu Pan
- Department of Nursing, Affiliated Hospital of Zunyi Medical University, Zunyi 563100, China; Nursing School of Zunyi Medical University, Zunyi 563100, China
| | - Rong Zhu
- Department of Pediatric surgrey, Guizhou Provincial People's Hospital, Guiyang 550000, China
| | - Jun Pei
- Department of Pediatric surgrey, Guizhou Provincial People's Hospital, Guiyang 550000, China.
| | - Li Zhang
- Department of Nursing, Affiliated Hospital of Zunyi Medical University, Zunyi 563100, China; Nursing School of Zunyi Medical University, Zunyi 563100, China.
| |
Collapse
|
52
|
Stojanovic L, Abbotts R, Tripathi K, Coon CM, Rajendran S, Farid EA, Hostetter G, Guarnieri JW, Wallace DC, Liu S, Wan J, Calendo G, Marker R, Gohari Z, Inayatullah MMA, Tiwari VK, Kader T, Santagata S, Drapkin R, Kommoss S, Pfisterer J, Konecny GE, Coopergard R, Issa JPJ, Winterhoff BJN, Topper MJ, Sandusky GE, Miller KD, Baylin SB, Nephew KP, Rassool FV. ZNFX1 Functions as a Master Regulator of Epigenetically Induced Pathogen Mimicry and Inflammasome Signaling in Cancer. Cancer Res 2025; 85:1183-1198. [PMID: 39804147 PMCID: PMC11968244 DOI: 10.1158/0008-5472.can-24-1286] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Revised: 10/18/2024] [Accepted: 01/07/2025] [Indexed: 01/22/2025]
Abstract
DNA methyltransferase (DNMT) and PARP inhibitors induce a stimulator of IFN gene-dependent pathogen mimicry response (PMR) in ovarian and other cancers. In this study, we showed that combining DNMT and PARP inhibitors upregulates expression of the nucleic acid sensor NFX1-type zinc finger-containing 1 (ZNFX1) protein. ZNFX1 mediated the induction of PMR in mitochondria, serving as a gateway for stimulator of IFN gene-dependent IFN/inflammasome signaling. Loss of ZNFX1 in ovarian cancer cells promoted proliferation and spheroid formation in vitro and tumor growth in vivo. In patient ovarian cancer databases, expression of ZNFX1 was elevated in advanced stage disease, and ZNFX1 expression alone significantly correlated with an increase in overall survival in a phase III trial for patients with therapy-resistant ovarian cancer receiving bevacizumab in combination with chemotherapy. RNA sequencing revealed an association between inflammasome signaling through ZNFX1 and abnormal vasculogenesis. Together, this study identified that ZNFX1 is a tumor suppressor that controls PMR signaling through mitochondria and may serve as a biomarker to facilitate personalized therapy in patients with ovarian cancer. Significance: DNMT and PARP inhibitors induce a nucleic acid sensor, ZNFX1, that serves as a mitochondrial gateway to STING-dependent inflammasome signaling with tumor suppressor properties in ovarian cancer.
Collapse
Affiliation(s)
- Lora Stojanovic
- University of Maryland Marlene and Stewart Greenebaum Comprehensive Cancer Center, Baltimore, MD 21201, USA
- Division of Translational Radiation Sciences, Department of Radiation Oncology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Rachel Abbotts
- University of Maryland Marlene and Stewart Greenebaum Comprehensive Cancer Center, Baltimore, MD 21201, USA
- Division of Translational Radiation Sciences, Department of Radiation Oncology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Kaushlendra Tripathi
- University of Maryland Marlene and Stewart Greenebaum Comprehensive Cancer Center, Baltimore, MD 21201, USA
- Division of Translational Radiation Sciences, Department of Radiation Oncology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Collin M. Coon
- Medical Sciences Program, Indiana University School of Medicine-Bloomington, Bloomington, IN 47405, USA
| | - Saranya Rajendran
- Medical Sciences Program, Indiana University School of Medicine-Bloomington, Bloomington, IN 47405, USA
| | - Elnaz Abbasi Farid
- Medical Sciences Program, Indiana University School of Medicine-Bloomington, Bloomington, IN 47405, USA
| | | | - Joseph W. Guarnieri
- Center for Mitochondrial and Epigenomic Medicine, Division of Human Genetics, The Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Douglas C. Wallace
- Center for Mitochondrial and Epigenomic Medicine, Division of Human Genetics, The Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
- Department of Pediatrics, Division of Human Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Sheng Liu
- Department of Medical and Molecular Genetics, Indiana University School of Medicine Indianapolis, IN 46202, USA
- Department of Anatomy, Cell Biology and Physiology, Indiana University School of Medicine Indianapolis, IN 46202, USA
| | - Jun Wan
- Department of Medical and Molecular Genetics, Indiana University School of Medicine Indianapolis, IN 46202, USA
- Department of Anatomy, Cell Biology and Physiology, Indiana University School of Medicine Indianapolis, IN 46202, USA
| | | | - Rebecca Marker
- Division of Translational Radiation Sciences, Department of Radiation Oncology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Zahra Gohari
- University of Maryland Marlene and Stewart Greenebaum Comprehensive Cancer Center, Baltimore, MD 21201, USA
- Division of Translational Radiation Sciences, Department of Radiation Oncology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | | | | | - Tanjina Kader
- Laboratory of Systems Pharmacology, Harvard Medical School, Boston, MA, USA
- Ludwig Center at Harvard, Boston, MA, USA
| | - Sandro Santagata
- Laboratory of Systems Pharmacology, Harvard Medical School, Boston, MA, USA
- Ludwig Center at Harvard, Boston, MA, USA
- Department of Pathology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Ronny Drapkin
- Penn Ovarian Cancer Research Center, Department of Obstetrics and Gynecology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- Basser Center for BRCA, Abramson Cancer Center, University of Pennsylvania School of Medicine, Philadelphia, PA, USA
| | - Stefan Kommoss
- Diakonie-Klinikum Schwäbisch Hall, Germany, Institute for Health Informatics
- Kliniken Essen-Mitte, Gynäkologie und Gynäkologische Onkologie, Germany
| | | | | | - Ryan Coopergard
- Institute for Health Informatics, University of Minnesota, Minneapolis, MN 55455, USA
| | | | - Boris J. N. Winterhoff
- Department of Obstetrics, Gynecology and Women’s Health (OBGYN), Division of Gynecologic Oncology, University of Minesota, MN 55812-3011, USA
| | - Michael J. Topper
- Department of Oncology, The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, MD 21231, USA
| | - George E. Sandusky
- Pathology and Laboratory Medicine, Indiana University School of Medicine, Indianapolis, IN 46202, USA
- Indiana University Melvin and Bren Simon Comprehensive Cancer Center, Indianapolis, IN 46202, USA
| | - Kathy D. Miller
- Indiana University Melvin and Bren Simon Comprehensive Cancer Center, Indianapolis, IN 46202, USA
- Department of Medicine, Division of Hematology/Oncology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Stephen B. Baylin
- Van Andel Research Institute, Grand Rapids, MI 49503, USA
- Department of Oncology, The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, MD 21231, USA
| | - Kenneth P. Nephew
- Medical Sciences Program, Indiana University School of Medicine-Bloomington, Bloomington, IN 47405, USA
- Department of Anatomy, Cell Biology and Physiology, Indiana University School of Medicine Indianapolis, IN 46202, USA
- Indiana University Melvin and Bren Simon Comprehensive Cancer Center, Indianapolis, IN 46202, USA
| | - Feyruz V. Rassool
- University of Maryland Marlene and Stewart Greenebaum Comprehensive Cancer Center, Baltimore, MD 21201, USA
- Division of Translational Radiation Sciences, Department of Radiation Oncology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| |
Collapse
|
53
|
Liu G, Zhang Y, Cao Z, Zhao Z. Targeting KIF18A triggers antitumor immunity and enhances efficiency of PD-1 blockade in colorectal cancer with chromosomal instability phenotype. Cell Death Discov 2025; 11:130. [PMID: 40175357 PMCID: PMC11965295 DOI: 10.1038/s41420-025-02437-5] [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: 06/26/2024] [Revised: 02/27/2025] [Accepted: 03/24/2025] [Indexed: 04/04/2025] Open
Abstract
Colorectal cancer with chromosomal instability (CIN+) phenotype is immunosuppressive and refractory to immune checkpoint blockade (ICB) therapy. Recently, KIF18A is found to be a mitotic vulnerability in chromosomally unstable cancers, but whether targeting KIF18A affects antitumor immunity in CIN+ colorectal cancer is unknown. In our study, western blot, cell viability assay, transwell migration and invasion assays, flow cytometry, animal model, immunohistochemistry (IHC) staining, reverse transcription-quantitative PCR (RT-qPCR) and ELISA assay were conducted to evaluate the potential function of KIF18A in CIN+ colorectal cancer. We found that KIF18A inhibition by short hairpin RNAs (ShRNAs) or small inhibitor AM-1882 suppressed proliferation, migration, invasion and tumor growth and metastasis of CIN+ colorectal cancer cells in vitro and in vivo. Moreover, targeting KIF18A disrupted cell-cycle progression and induced G2/M arrest in CIN+ colorectal cancer cells. In addition, KIF18A inhibition promoted immune infiltration and activation in CIN+ colorectal tumors. KIF18A inhibition suppressed proliferation of Tregs and increased infiltration and activation of cytotoxic CD8+ T cells in CIN+ colorectal tumors. Mechanically, KIF18A inhibition stimulated type I IFN signaling and cGAS-STING activation in CIN+ colorectal tumors. Finally, targeting KIF18A enhanced PD-1 blockade efficiency in CIN+ colorectal tumors through T cells. Our data elucidated a novel role of KIF18A in antitumor immunity of CIN+ colorectal cancer.
Collapse
Affiliation(s)
- Gang Liu
- Senior Department of General Surgery, Chinese PLA General Hospital, Beijing, China.
| | - Yan Zhang
- Senior Department of General Surgery, Chinese PLA General Hospital, Beijing, China
| | - Zhen Cao
- Senior Department of General Surgery, Chinese PLA General Hospital, Beijing, China
| | - Zhanwei Zhao
- Senior Department of General Surgery, Chinese PLA General Hospital, Beijing, China
| |
Collapse
|
54
|
Xu WX, Wen X, Fu YT, Yang J, Cui H, Fan RF. Nuclear receptor coactive 4-mediated ferritinophagy: a key role of heavy metals toxicity. Arch Toxicol 2025; 99:1257-1270. [PMID: 39928088 DOI: 10.1007/s00204-025-03963-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: 10/01/2024] [Accepted: 01/15/2025] [Indexed: 02/11/2025]
Abstract
Nuclear receptor coactive 4 (NCOA4) is a specific receptor for ferritinophagy, transporting ferritin to lysosomal degradation, releasing free iron, and excessive iron levels may lead to cellular redox imbalance, contributing to cell death, predominantly ferroptosis. NCOA4 is regulated by a variety of transcriptional, post-transcriptional, translational, and post-translational modifications. Targeted modulation of NCOA4-mediated ferritinophagy has been successfully used as a therapeutic strategy in several disease models. Recent evidences have elucidated that ferritinophagy and ferroptosis played a major role in heavy metals toxicity. In this review, we explored the regulatory mechanism of NCOA4 as the sole receptor for ferritinophagy from multiple perspectives based on previous studies. The significant role of ferritinophagy-mediated ferroptosis in heavy metals toxicity was discussed in detail, emphasizing the great potential of NCOA4 as a target for heavy metals toxicity.
Collapse
Affiliation(s)
- Wan-Xue Xu
- College of Veterinary Medicine, Shandong Agricultural University, 7 Panhe Street, Tai'an, 271017, Shandong, China
- Shandong Provincial Key Laboratory of Zoonoses, Shandong Agricultural University, 7 Panhe Street, Tai'an, 271017, Shandong, China
| | - Xue Wen
- College of Veterinary Medicine, Shandong Agricultural University, 7 Panhe Street, Tai'an, 271017, Shandong, China
- Shandong Provincial Key Laboratory of Zoonoses, Shandong Agricultural University, 7 Panhe Street, Tai'an, 271017, Shandong, China
| | - Yi-Tong Fu
- College of Veterinary Medicine, Shandong Agricultural University, 7 Panhe Street, Tai'an, 271017, Shandong, China
- Shandong Provincial Key Laboratory of Zoonoses, Shandong Agricultural University, 7 Panhe Street, Tai'an, 271017, Shandong, China
| | - Jie Yang
- College of Veterinary Medicine, Shandong Agricultural University, 7 Panhe Street, Tai'an, 271017, Shandong, China
- Shandong Provincial Key Laboratory of Zoonoses, Shandong Agricultural University, 7 Panhe Street, Tai'an, 271017, Shandong, China
| | - Han Cui
- College of Veterinary Medicine, Shandong Agricultural University, 7 Panhe Street, Tai'an, 271017, Shandong, China
- Shandong Provincial Key Laboratory of Zoonoses, Shandong Agricultural University, 7 Panhe Street, Tai'an, 271017, Shandong, China
| | - Rui-Feng Fan
- College of Veterinary Medicine, Shandong Agricultural University, 7 Panhe Street, Tai'an, 271017, Shandong, China.
- Shandong Provincial Key Laboratory of Zoonoses, Shandong Agricultural University, 7 Panhe Street, Tai'an, 271017, Shandong, China.
| |
Collapse
|
55
|
Linderman SW, DeRidder L, Sanjurjo L, Foote MB, Alonso MJ, Kirtane AR, Langer R, Traverso G. Enhancing immunotherapy with tumour-responsive nanomaterials. Nat Rev Clin Oncol 2025; 22:262-282. [PMID: 40050505 DOI: 10.1038/s41571-025-01000-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/05/2025] [Indexed: 03/09/2025]
Abstract
The targeted delivery of immunotherapies to tumours using tumour-responsive nanomaterials is a promising area of cancer research with the potential to address the limitations of systemic administration such as on-target off-tumour toxicities and a lack of activity owing to the immunosuppressive tumour microenvironment (TME). Attempts to address these challenges include the design and functionalization of nanomaterials capable of releasing their cargoes in response to specific TME characteristics, thus facilitating the targeted delivery of immune-checkpoint inhibitors, cytokines, mRNAs, vaccines and, potentially, chimaeric antigen receptors as well as of agents that modulate the extracellular matrix and induce immunogenic cell death. In this Review, we describe these various research efforts in the context of the dynamic properties of the TME, such as pH, reductive conditions, reactive oxygen species, hypoxia, specific enzymes, high levels of ATP and locoregional aspects, which can be leveraged to enhance the specificity and efficacy of nanomaterial-based immunotherapies. Highlighting preclinical successes and ongoing clinical trials, we evaluate the current landscape and potential of these innovative approaches. We also consider future research directions as well as the most important barriers to successful clinical translation, emphasizing the transformative potential of tumour-responsive nanomaterials in overcoming the barriers that limit the activity of traditional immunotherapies, thus improving patient outcomes.
Collapse
Affiliation(s)
- Stephen W Linderman
- Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
- Division of Hospital Medicine, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Louis DeRidder
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Harvard-MIT Division of Health Science Technology, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Lucía Sanjurjo
- Health Research Institute of Santiago de Compostela (IDIS), Santiago de Compostela, Spain
- Center for Research in Molecular Medicine and Chronic Diseases (CIMUS), University of Santiago de Compostela, Santiago de Compostela, Spain
| | - Michael B Foote
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - María José Alonso
- Center for Research in Molecular Medicine and Chronic Diseases (CIMUS), University of Santiago de Compostela, Santiago de Compostela, Spain
- Department of Pharmacy and Pharmaceutical Technology, University of Santiago de Compostela, Santiago de Compostela, Spain
- IMDEA Nanosciences Institute, Madrid, Spain
| | - Ameya R Kirtane
- Department of Pharmaceutics, University of Minnesota, Minneapolis, MN, USA
| | - Robert Langer
- Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA.
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA.
| | - Giovanni Traverso
- Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA.
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA.
- Division of Gastroenterology, Hepatology and Endoscopy, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.
- Broad Institute of MIT and Harvard, Cambridge, MA, USA.
| |
Collapse
|
56
|
Han X, Wang X, Han F, Yan H, Sun J, Zhang X, Moog C, Zhang C, Su B. The cGAS-STING pathway in HIV-1 and Mycobacterium tuberculosis coinfection. Infection 2025; 53:495-511. [PMID: 39509013 DOI: 10.1007/s15010-024-02429-0] [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: 04/23/2024] [Accepted: 10/28/2024] [Indexed: 11/15/2024]
Abstract
Mycobacterium tuberculosis (M. tuberculosis) infection is the most common opportunistic infection in human immunodeficiency virus-1 (HIV-1)-infected individuals, and the mutual reinforcement of these two pathogens may accelerate disease progression and lead to rapid mortality. Therefore, HIV-1/M. tuberculosis coinfection is one of the major global public health concerns. HIV-1 infection is the greatest risk factor for M. tuberculosis infection and increases the likelihood of endogenous relapse and exogenous reinfection with M. tuberculosis. Moreover, M. tuberculosis further increases HIV-1 replication and the occurrence of chronic immune activation, accelerating the progression of HIV-1 disease. Exploring the pathogenesis of HIV-1/M. tuberculosis coinfections is essential for the development of novel treatments to reduce the global burden of tuberculosis. Innate immunity, which is the first line of host immune defense, plays a critical role in resisting HIV-1 and M. tuberculosis infections. The role of the cyclic guanosine monophosphate-adenosine monophosphate synthase (cGAS)-stimulator of interferon genes (STING) signaling pathway, which is a major DNA-sensing innate immune signaling pathway, in HIV-1 infection and M. tuberculosis infection has been intensively studied. This paper reviews the role of the cGAS-STING signaling pathway in HIV-1 infection and M. tuberculosis infection and discusses the possible role of this pathway in HIV-1/M. tuberculosis coinfection to provide new insight into the pathogenesis of HIV-1/M. tuberculosis coinfection and the development of novel therapeutic strategies.
Collapse
Affiliation(s)
- Xiaoxu Han
- Beijing Key Laboratory for HIV/AIDS Research, Clinical and Research Center for Infectious Diseases, Beijing Youan Hospital, Capital Medical University, Beijing, 100069, China
- Sino-French Joint Laboratory for HIV/AIDS Research, Sino-French Joint Laboratory for Research on Humoral Immune Response to HIV Infection, Beijing Youan Hospital, Capital Medical University, Beijing, 100069, China
| | - Xiuwen Wang
- Beijing Key Laboratory for HIV/AIDS Research, Clinical and Research Center for Infectious Diseases, Beijing Youan Hospital, Capital Medical University, Beijing, 100069, China
- Sino-French Joint Laboratory for HIV/AIDS Research, Sino-French Joint Laboratory for Research on Humoral Immune Response to HIV Infection, Beijing Youan Hospital, Capital Medical University, Beijing, 100069, China
| | - Fangping Han
- State Key Laboratory of Membrane Biology, School of Pharmaceutical Sciences, Tsinghua University, Beijing, 100084, China
- Tsinghua-Peking Center for Life Sciences, Beijing, 100084, China
| | - Hongxia Yan
- Beijing Key Laboratory for HIV/AIDS Research, Clinical and Research Center for Infectious Diseases, Beijing Youan Hospital, Capital Medical University, Beijing, 100069, China
- Sino-French Joint Laboratory for HIV/AIDS Research, Sino-French Joint Laboratory for Research on Humoral Immune Response to HIV Infection, Beijing Youan Hospital, Capital Medical University, Beijing, 100069, China
| | - Jin Sun
- Beijing Key Laboratory for HIV/AIDS Research, Clinical and Research Center for Infectious Diseases, Beijing Youan Hospital, Capital Medical University, Beijing, 100069, China
- Sino-French Joint Laboratory for HIV/AIDS Research, Sino-French Joint Laboratory for Research on Humoral Immune Response to HIV Infection, Beijing Youan Hospital, Capital Medical University, Beijing, 100069, China
| | - Xin Zhang
- Beijing Key Laboratory for HIV/AIDS Research, Clinical and Research Center for Infectious Diseases, Beijing Youan Hospital, Capital Medical University, Beijing, 100069, China
- Sino-French Joint Laboratory for HIV/AIDS Research, Sino-French Joint Laboratory for Research on Humoral Immune Response to HIV Infection, Beijing Youan Hospital, Capital Medical University, Beijing, 100069, China
| | - Christiane Moog
- Sino-French Joint Laboratory for HIV/AIDS Research, Sino-French Joint Laboratory for Research on Humoral Immune Response to HIV Infection, Beijing Youan Hospital, Capital Medical University, Beijing, 100069, China
- Laboratoire d'ImmunoRhumatologie Moléculaire, Faculté de Médecine, Fédération de Médecine Translationnelle de Strasbourg (FMTS), Institut National de la Santé et de la Recherche Médicale (INSERM) UMR_S 1109, Institut Thématique Interdisciplinaire (ITI) de Médecine de Précision de Strasbourg, Fédération Hospitalo-Universitaire OMICARE, Université de Strasbourg, Transplantex, Strasbourg, NG, 67000, France
- Vaccine Research Institute (VRI), Créteil, 94000, France
| | - Conggang Zhang
- State Key Laboratory of Membrane Biology, School of Pharmaceutical Sciences, Tsinghua University, Beijing, 100084, China.
- Tsinghua-Peking Center for Life Sciences, Beijing, 100084, China.
| | - Bin Su
- Beijing Key Laboratory for HIV/AIDS Research, Clinical and Research Center for Infectious Diseases, Beijing Youan Hospital, Capital Medical University, Beijing, 100069, China.
- Sino-French Joint Laboratory for HIV/AIDS Research, Sino-French Joint Laboratory for Research on Humoral Immune Response to HIV Infection, Beijing Youan Hospital, Capital Medical University, Beijing, 100069, China.
| |
Collapse
|
57
|
Liu M, Ye J, Yang F, Dai X, Xing C, Chang H, Gao F, Chen H, Chen J, Cao H. Based on the Sam50-MICOS-ATAD3-mtDNA axis: Exploring oligomeric proanthocyanidins to alleviate molybdenum and cadmium co-induced liver inflammation in sheep. Int J Biol Macromol 2025; 304:141035. [PMID: 39954884 DOI: 10.1016/j.ijbiomac.2025.141035] [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: 11/10/2024] [Revised: 02/06/2025] [Accepted: 02/12/2025] [Indexed: 02/17/2025]
Abstract
Molybdenum (Mo) and cadmium (Cd), well-defined hazardous pollutants in the environment, exhibit potential toxic effects on liver tissues by inducing oxidative stress and inflammatory responses. This study aims to investigate the role of the Sam50-MICOS-ATAD3-mtDNA axis in mediating the inflammatory response in liver inflammation induced by co-exposure to Mo and Cd in sheep, as well as the protective effects of oligomeric proanthocyanidins (OPC). The findings indicated that co-exposure to Mo and Cd induced cellular degeneration, rupture of hepatic mitochondrial membranes and mitochondrial dysfunction that was accompanied by the levels of ATP, SDH and GSH-Px reduced in the sheep liver tissue. Furthermore, the co-exposure downregulated the expression levels of mitochondrial membrane proteins (Sam50, MICOS and ATAD3) and degree of co-localization between Sam50 and Mic60. In addition, co-exposure to Mo and Cd elicited an increase in mtDNA content and promoted the upregulation of inflammation-related factor levels, which resulted in an augmentation of TNF-α, CRP, and IL-18 contents. However, OPC alleviated the above changes induced by the combination of Mo and Cd. In conclusion, co-exposure to Mo and Cd decreases mtDNA stability by disrupting the Sam50-MICOS-ATAD3 axis, thereby inducing liver inflammation in sheep. Nevertheless, OPC could alleviate this damage.
Collapse
Affiliation(s)
- Maokang Liu
- 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, Nanchang 330045, Jiangxi, PR China
| | - Junhua Ye
- Medicine College, Nanchang Institute of Technology, No. 901 Yingxiong Avenue, Economic and Technological Development District, Nanchang 330044, Jiangxi, PR China
| | - Fan Yang
- 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, Nanchang 330045, Jiangxi, PR China
| | - Xueyan Dai
- 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, Nanchang 330045, Jiangxi, 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, Nanchang 330045, Jiangxi, PR China
| | - Huifeng Chang
- 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, Nanchang 330045, Jiangxi, PR China
| | - Feiyan Gao
- 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, Nanchang 330045, Jiangxi, PR China
| | - Huawei 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, Nanchang 330045, Jiangxi, PR China
| | - Jing 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, Nanchang 330045, Jiangxi, PR China
| | - Huabin 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, Nanchang 330045, Jiangxi, PR China.
| |
Collapse
|
58
|
Acchioni M, Acchioni C, Hiscott J, Sgarbanti M. Origin and function of anti-interferon type I viral proteins. Virology 2025; 605:110456. [PMID: 39999585 DOI: 10.1016/j.virol.2025.110456] [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: 12/02/2024] [Revised: 02/17/2025] [Accepted: 02/19/2025] [Indexed: 02/27/2025]
Abstract
Type I interferons (IFN-I) are the most important innate immune cytokines produced by vertebrate host cells following, virus infection. Broadly speaking, detection of infecting viral nucleic acids by pattern recognition receptors (PRR) and subsequent downstream signaling triggers synthesis of a large number of IFN-I-stimulated genes (ISGs), endowed with diverse antiviral effector function. The co-evolution of virus-host interactions over million years has resulted in the emergence of viral strategies that target and inhibit host PRR-mediated detection, signal transduction pathways and IFN-I-mediated stimulation of ISGs. In this review, we illustrate the multiple mechanisms of viral immune evasion and discuss the co-evolution of anti-IFN-I viral proteins by summarizing key examples from recent literature. Due to the large number of anti-IFN-I proteins described, we provide here an evaluation of the prominent examples from different virus families. Understanding the unrelenting evolution of viral evasion strategies will provide mechanistic detail concerning these evolving interactions but will further enhance the development of tailored antiviral approaches.
Collapse
Affiliation(s)
- Marta Acchioni
- Department of Infectious Diseases, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161, Rome, Italy.
| | - Chiara Acchioni
- Department of Infectious Diseases, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161, Rome, Italy.
| | - John Hiscott
- Istituto Pasteur Italia, Fondazione Cenci Bolognetti, Viale Regina Elena 291, 00161, Rome, Italy.
| | - Marco Sgarbanti
- Department of Infectious Diseases, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161, Rome, Italy.
| |
Collapse
|
59
|
Luri-Rey C, Teijeira Á, Wculek SK, de Andrea C, Herrero C, Lopez-Janeiro A, Rodríguez-Ruiz ME, Heras I, Aggelakopoulou M, Berraondo P, Sancho D, Melero I. Cross-priming in cancer immunology and immunotherapy. Nat Rev Cancer 2025; 25:249-273. [PMID: 39881005 DOI: 10.1038/s41568-024-00785-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 12/03/2024] [Indexed: 01/31/2025]
Abstract
Cytotoxic T cell immune responses against cancer crucially depend on the ability of a subtype of professional antigen-presenting cells termed conventional type 1 dendritic cells (cDC1s) to cross-present antigens. Cross-presentation comprises redirection of exogenous antigens taken from other cells to the major histocompatibility complex class I antigen-presenting machinery. In addition, once activated and having sensed viral moieties or T helper cell cooperation via CD40-CD40L interactions, cDC1s provide key co-stimulatory ligands and cytokines to mount and sustain CD8+ T cell immune responses. This regulated process of cognate T cell activation is termed cross-priming. In cancer mouse models, CD8+ T cell cross-priming by cDC1s is crucial for the efficacy of most, if not all, immunotherapy strategies. In patients with cancer, the presence and abundance of cDC1s in the tumour microenvironment is markedly associated with the level of T cell infiltration and responsiveness to immune checkpoint inhibitors. Therapeutic strategies to increase the numbers of cDC1s using FMS-like tyrosine kinase 3 ligand (FLT3L) and/or their activation status show evidence of efficacy in cancer mouse models and are currently being tested in initial clinical trials with promising results so far.
Collapse
Affiliation(s)
- Carlos Luri-Rey
- Program of Immunology and Immunotherapy, Cima Universidad de Navarra, Pamplona, Spain
| | - Álvaro Teijeira
- Program of Immunology and Immunotherapy, Cima Universidad de Navarra, Pamplona, Spain
- Navarra Institute for Health Research (IDISNA), Pamplona, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain
| | - Stefanie K Wculek
- Innate Immune Biology Laboratory, Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology (BIST), Barcelona, Spain
| | - Carlos de Andrea
- Department of Pathology, Clínica Universidad de Navarra, Pamplona, Spain
| | - Claudia Herrero
- Program of Immunology and Immunotherapy, Cima Universidad de Navarra, Pamplona, Spain
- Department of Pathology, Clínica Universidad de Navarra, Pamplona, Spain
| | | | | | - Ignacio Heras
- Immunobiology Laboratory, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | | | - Pedro Berraondo
- Program of Immunology and Immunotherapy, Cima Universidad de Navarra, Pamplona, Spain
- Navarra Institute for Health Research (IDISNA), Pamplona, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain
| | - David Sancho
- Immunobiology Laboratory, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - Ignacio Melero
- Program of Immunology and Immunotherapy, Cima Universidad de Navarra, Pamplona, Spain.
- Navarra Institute for Health Research (IDISNA), Pamplona, Spain.
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain.
- Nuffield Department of Medicine, University of Oxford, Oxford, UK.
- Departments of Immunology and Oncology, Clínica Universidad de Navarra, Pamplona, Spain.
| |
Collapse
|
60
|
Khalilzad MA, Mohammadi J, Amirsaadat S, Najafi S, Zare S, Nilforoushzadeh MA, Khalilzad M, Amirkhani MA, Peyrovan A, Khalili SFS, Farahani A, Zare S. Therapeutic potential of apoptotic vesicles in modulating inflammation, immune responses, and tissue regeneration. J Nanobiotechnology 2025; 23:260. [PMID: 40170079 PMCID: PMC11960034 DOI: 10.1186/s12951-025-03278-1] [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: 09/20/2024] [Accepted: 02/24/2025] [Indexed: 04/03/2025] Open
Abstract
The process of apoptosis plays a crucial role in tissue homeostasis, immune system regulation, and organ formation. Apoptotic vesicles (ApoEVs) are involved in efferocytosis, the process by which phagocytes ingest dead cells. ApoEVs also have potential therapeutic applications in cancer treatment, ischemic diseases, and their anti-inflammatory properties make them incredibly versatile for medical applications. These vesicles can induce apoptosis in cancer cells, provide tumor antigens for cancer vaccines, and even serve as effective drug delivery systems. Moreover, they can target hypoxic cells, inhibit inflammatory cell death pathways, and promote tissue regeneration. Also, their potential in addressing inflammatory disorders such as gastrointestinal ailments, osteoarthritis, and diabetes is promising. Additionally, ApoEVs can polarize anti-inflammatory immune cells and suppress inflammatory immune responses which make them a viable option for addressing the unmet need for novel anti-inflammatory medications. Despite a wealth of reviews examining the applications of ApoEVs, very few have thoroughly investigated the mechanisms underlying their anti-inflammatory effects. This distinctive approach positions the current review as timely and immensely relevant, illuminating the intriguing ways these entities function beyond their established advantages.
Collapse
Affiliation(s)
- Mohammad Amin Khalilzad
- Department of Life Sciences Engineering, Faculty of New Sciences and Technologies, University of Tehran, Tehran, 143951561, Iran
- Skin and Stem Cell Research Center, Tehran University of Medical Sciences, Tehran, Iran
- School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Javad Mohammadi
- Department of Life Sciences Engineering, Faculty of New Sciences and Technologies, University of Tehran, Tehran, 143951561, Iran.
| | - Soumayeh Amirsaadat
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Sajad Najafi
- Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Biotechnology and Medicinal Plants Research Center, Ilam University of Medical Sciences, Ilam, Iran
| | - Sona Zare
- Skin and Stem Cell Research Center, Tehran University of Medical Sciences, Tehran, Iran.
- Laserin Medical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
- Stem Cell and Regenerative Medicine Institute, Sharif University of Technology, Tehran, Iran.
- Department of Mechanical Engineering, Sharif University of Technology, Tehran, Iran.
| | - Mohammad Ali Nilforoushzadeh
- Skin and Stem Cell Research Center, Tehran University of Medical Sciences, Tehran, Iran.
- Skin Repair Research Center, Jordan Dermatology and Hair Transplantation Center, Tehran, Iran.
| | - Mitra Khalilzad
- Brain Mapping Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | | | - Aysan Peyrovan
- Skin and Stem Cell Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | | | - Atefeh Farahani
- Department of Pharmaceutics, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
- Nanotechnology Research Centre, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Solmaz Zare
- Laserin Medical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
61
|
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
|
62
|
Zhang B, Xu P, Ablasser A. Regulation of the cGAS-STING Pathway. Annu Rev Immunol 2025; 43:667-692. [PMID: 40085836 DOI: 10.1146/annurev-immunol-101721-032910] [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
The cGAS-cGAMP-STING pathway is essential for immune defense against pathogens. Upon binding DNA, cGAS synthesizes cGAMP, which activates STING, leading to potent innate immune effector responses. However, lacking specific features to distinguish between self and nonself DNA, cGAS-STING immunity requires precise regulation to prevent aberrant activation. Several safeguard mechanisms acting on different levels have evolved to maintain tolerance to self DNA and ensure immune homeostasis under normal conditions. Disruption of these safeguards can lead to erroneous activation by self DNA, resulting in inflammatory conditions but also favorable antitumor immunity. Insights into structural and cellular checkpoints that control and terminate cGAS-STING signaling are essential for comprehending and manipulating DNA-triggered innate immunity in health and disease.
Collapse
Affiliation(s)
- Bing Zhang
- Global Health Institute, Swiss Federal Institute of Technology Lausanne (EPFL), Lausanne, Switzerland;
| | - Pengbiao Xu
- Global Health Institute, Swiss Federal Institute of Technology Lausanne (EPFL), Lausanne, Switzerland;
| | - Andrea Ablasser
- Global Health Institute, Swiss Federal Institute of Technology Lausanne (EPFL), Lausanne, Switzerland;
- Institute for Cancer Research (ISREC), Swiss Federal Institute of Technology Lausanne (EPFL), Lausanne, Switzerland
| |
Collapse
|
63
|
Mu W, Xu G, Li L, Wen J, Xiu Y, Zhao J, Liu T, Wei Z, Luo W, Yang H, Wu Z, Zhan X, Xiao X, Bai Z. Carnosic Acid Directly Targets STING C-Terminal Tail to Improve STING-Mediated Inflammatory Diseases. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2417686. [PMID: 39965124 PMCID: PMC11984877 DOI: 10.1002/advs.202417686] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/28/2024] [Revised: 01/30/2025] [Indexed: 02/20/2025]
Abstract
cGAS (cyclic GMP-AMP synthase)-STING (stimulator of interferon genes) signaling plays a vital role in innate immunity, while its deregulation may lead to a wide variety of autoinflammatory and autoimmune diseases. It is essential to identify specifically effective lead compounds to inhibit the signaling. Herein, it is shown that carnosic acid (CA), an active ingredient of medicinal plant Rosmarinus officinalis L., specifically suppressed cGAS-STING pathway activation and the subsequent inflammatory responses. Mechanistically, CA directly bound to STING C-terminal tail (CTT), impeded the recruitment of TANK-binding kinase 1 (TBK1) onto STING signalosome, thereby blocking the phosphorylation of STING and interferon regulatory factor 3 (IRF3) nuclear translocation. Importantly, CA dramatically attenuated STING-mediated inflammatory responses in vivo. Consistently, CA has a salient ameliorative effect on autoinflammatory disease model mediated by Trex1 deficiency, via inhibition of the cGAS-STING signaling. Notably, the study further indicates that phenolic hydroxyl groups are essential for CA-mediated STING inhibitory activity. Collectively, the results thus identify STING as one of the crucial targets of CA for mediating CA's anti-inflammatory activity, and further reveal that STING CTT may be a novel promising target for drug development.
Collapse
Affiliation(s)
- Wenqing Mu
- Department of Hepatologythe Fifth Medical Center of PLA General HospitalBeijing100039China
- School of Traditional Chinese MedicineCapital Medical UniversityBeijing100069China
- State Key Laboratory of Radiation Medicine and ProtectionInstitutes for Translational MedicineSoochow UniversityJiangsu215123China
| | - Guang Xu
- School of Traditional Chinese MedicineCapital Medical UniversityBeijing100069China
| | - Ling Li
- Beijing Institute of BiotechnologyBeijing100071China
| | - Jincai Wen
- Department of Hepatologythe Fifth Medical Center of PLA General HospitalBeijing100039China
- Military Institute of Chinese MateriaFifth Medical Center of Chinese PLA General HospitalBeijing100039China
| | - Ye Xiu
- Department of Hepatologythe Fifth Medical Center of PLA General HospitalBeijing100039China
- Military Institute of Chinese MateriaFifth Medical Center of Chinese PLA General HospitalBeijing100039China
| | - Jia Zhao
- Department of Hepatologythe Fifth Medical Center of PLA General HospitalBeijing100039China
- Military Institute of Chinese MateriaFifth Medical Center of Chinese PLA General HospitalBeijing100039China
| | - Tingting Liu
- Department of Hepatologythe Fifth Medical Center of PLA General HospitalBeijing100039China
- Military Institute of Chinese MateriaFifth Medical Center of Chinese PLA General HospitalBeijing100039China
| | - Ziying Wei
- Department of Hepatologythe Fifth Medical Center of PLA General HospitalBeijing100039China
- Military Institute of Chinese MateriaFifth Medical Center of Chinese PLA General HospitalBeijing100039China
| | - Wei Luo
- Department of Hepatologythe Fifth Medical Center of PLA General HospitalBeijing100039China
- Military Institute of Chinese MateriaFifth Medical Center of Chinese PLA General HospitalBeijing100039China
| | - Huijie Yang
- Department of Hepatologythe Fifth Medical Center of PLA General HospitalBeijing100039China
- Military Institute of Chinese MateriaFifth Medical Center of Chinese PLA General HospitalBeijing100039China
| | - Zhixin Wu
- Department of Hepatologythe Fifth Medical Center of PLA General HospitalBeijing100039China
- Military Institute of Chinese MateriaFifth Medical Center of Chinese PLA General HospitalBeijing100039China
| | - Xiaoyan Zhan
- Department of Hepatologythe Fifth Medical Center of PLA General HospitalBeijing100039China
- Military Institute of Chinese MateriaFifth Medical Center of Chinese PLA General HospitalBeijing100039China
| | - Xiaohe Xiao
- Department of Hepatologythe Fifth Medical Center of PLA General HospitalBeijing100039China
- Military Institute of Chinese MateriaFifth Medical Center of Chinese PLA General HospitalBeijing100039China
| | - Zhaofang Bai
- Department of Hepatologythe Fifth Medical Center of PLA General HospitalBeijing100039China
- Military Institute of Chinese MateriaFifth Medical Center of Chinese PLA General HospitalBeijing100039China
| |
Collapse
|
64
|
Cheng W, Peng X, He L, Ren W, Chen J, Tang X, Bao D, Liu G, Jiang L, Piao JG. Bimetallic MnZnS X Nanotheranostics for Self-Activatable Chemo-Immunotherapy of Hepatocellular Carcinoma via H₂S-Triggered Arsenic Prodrug Activation and Binary cGAS-STING Pathway Modulation. Adv Healthc Mater 2025; 14:e2404238. [PMID: 39995347 DOI: 10.1002/adhm.202404238] [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] [Revised: 02/17/2025] [Indexed: 02/26/2025]
Abstract
Arsenic trioxide (As2O3) has long been utilized in traditional Chinese medicine due to its therapeutic properties. While it exhibits potent anticancer activity, its clinical application is hindered by systemic toxicity and limited tissue specificity. In this study, an advanced therapeutic approach is developed using arsenic prodrug-loaded bimetallic sulfide MnZnSX nanorods (As-MnZnSX NRs) to enhance both the efficacy and safety of As2O3 in hepatocellular carcinoma treatment. These nanorods are engineered to release Mn2+ and H2S within the tumor microenvironment, facilitating binary-cooperative activation of the cGAS-STING pathway. This dual activation mechanism enhances immune responses while converting the arsenic prodrug into its cytotoxic form, AsIII. The results demonstrate that Mn2+ amplifies the cGAS-STING pathway by inducing TBK1 phosphorylation and IRF3 activation, leading to dendritic cell maturation and improved tumor antigen cross-presentation. Simultaneously, H2S promotes prodrug conversion and enhances immune activation, collectively driving binary stimulation of the cGAS-STING pathway. This strategy significantly augments the antitumor efficacy of As2O3 by integrating immune modulation with targeted cytotoxic effects. Furthermore, MnZnSX nanorods enable in vivo MRI, allowing real-time monitoring of treatment progression. This study represents a substantial advancement in liver cancer therapy by integrating chemoimmunotherapy with diagnostic imaging, thereby improving therapeutic precision while minimizing systemic toxicity.
Collapse
Affiliation(s)
- WeiYi Cheng
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Xuqi Peng
- State Key Laboratory of Infectious Disease Vaccine Development, Xiang'an Biomedicine Laboratory, National Innovation Platform for Industry-Education Integration in Vaccine Research, Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen, 361102, China
| | - Li He
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - WeiYe Ren
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - JingQuan Chen
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - XiaoQian Tang
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Dandan Bao
- Department of Dermatology & Cosmetology, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Traditional Chinese Medicine), Hangzhou, 310006, China
| | - Gang Liu
- State Key Laboratory of Infectious Disease Vaccine Development, Xiang'an Biomedicine Laboratory, National Innovation Platform for Industry-Education Integration in Vaccine Research, Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen, 361102, China
| | - Lai Jiang
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Ji-Gang Piao
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, China
| |
Collapse
|
65
|
Bai Y, Zhan X, Zhu Q, Ji X, Lu Y, Gao Y, Li F, Guan Z, Zhou H, Rao Z. ATG16L1 restrains macrophage NLRP3 activation and alveolar epithelial cell injury during septic lung injury. Clin Transl Med 2025; 15:e70289. [PMID: 40211890 PMCID: PMC11986372 DOI: 10.1002/ctm2.70289] [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: 09/12/2024] [Revised: 03/11/2025] [Accepted: 03/18/2025] [Indexed: 04/14/2025] Open
Abstract
BACKGROUND The lung is the organ most commonly affected by sepsis. Additionally, acute lung injury (ALI) resulting from sepsis is a major cause of death in intensive care units. Macrophages are essential for maintaining normal lung physiological functions and are implicated in various pulmonary diseases. An essential autophagy protein, autophagy-related protein 16-like 1 (ATG16L1), is crucial for the inflammatory activation of macrophages. METHODS ATG16L1 expression was measured in lung from mice with sepsis. ALI was induced in myeloid ATG16L1-, NLRP3- and STING-deficient mice by intraperitoneal injection of lipopolysaccharide (LPS, 10 mg/kg). Using immunofluorescence and flow cytometry to assess the inflammatory status of LPS-treated bone marrow-derived macrophages (BMDMs). A co-culture system of BMDMs and MLE-12 cells was established in vitro. RESULTS Myeloid ATG16L1-deficient mice exhibited exacerbated septic lung injury and a more intense inflammatory response following LPS treatment. Mechanistically, ATG16L1-deficient macrophages exhibited impaired LC3B lipidation, damaged mitochondria and reactive oxygen species (ROS) accumulation. These abnormalities led to the activation of NOD-like receptor family pyrin domain-containing protein 3 (NLRP3), subsequently enhancing proinflammatory response. Overactivated ATG16L1-deficient macrophages aggravated the damage to alveolar epithelial cells and enhanced the release of double-stranded DNA (dsDNA), thereby promoting STING activation and subsequent NLRP3 activation in macrophages, leading to positive feedback activation of macrophage NLRP3 signalling. Scavenging mitochondrial ROS or inhibiting STING activation effectively suppresses NLRP3 activation in macrophages and alleviates ALI. Furthermore, overexpression of myeloid ATG16L1 limits NLRP3 activation and reduces the severity of ALI. CONCLUSIONS Our findings reveal a new role for ATG16L1 in regulating macrophage NLRP3 feedback activation during sepsis, suggesting it as a potential therapeutic target for treating sepsis-induced ALI. KEY POINTS Myeloid-specific ATG16L1 deficiency exacerbates sepsis-induced lung injury. ATG16L1-deficient macrophages exhibit impaired LC3B lipidation and ROS accumulation, leading to NLRP3 inflammasome activation. Uncontrolled inflammatory responses in ATG16L1-deficient macrophages aggravate alveolar epithelial cell damage. Alveolar epithelial cells release dsDNA, activating the cGAS-STING-NLRP3 signaling pathway, which subsequently triggers a positive feedback activation of NLRP3. Overexpression of ATG16L1 helps mitigate lung tissue inflammation, offering a novel therapeutic direction for sepsis-induced lung injury.
Collapse
Affiliation(s)
- Yan Bai
- Department of AnesthesiologyThe First Affiliated Hospital of Nanjing Medical UniversityNanjingChina
| | - Xinyu Zhan
- Hepatobiliary CenterThe First Affiliated Hospital of Nanjing Medical University, Key Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences, NHC Key Laboratory of Living Donor Liver Transplantation, Nanjing Medical UniversityNanjingChina
| | - Qing Zhu
- Department of AnesthesiologyThe First Affiliated Hospital of Nanjing Medical UniversityNanjingChina
| | - Xingyue Ji
- Department of AnesthesiologyThe First Affiliated Hospital of Nanjing Medical UniversityNanjingChina
| | - Yingying Lu
- Department of AnesthesiologyThe First Affiliated Hospital of Nanjing Medical UniversityNanjingChina
| | - Yiyun Gao
- Hepatobiliary CenterThe First Affiliated Hospital of Nanjing Medical University, Key Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences, NHC Key Laboratory of Living Donor Liver Transplantation, Nanjing Medical UniversityNanjingChina
| | - Fei Li
- Department of AnesthesiologyThe First Affiliated Hospital of Nanjing Medical UniversityNanjingChina
| | - Zhu Guan
- Department of AnesthesiologyThe First Affiliated Hospital of Nanjing Medical UniversityNanjingChina
| | - Haoming Zhou
- Hepatobiliary CenterThe First Affiliated Hospital of Nanjing Medical University, Key Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences, NHC Key Laboratory of Living Donor Liver Transplantation, Nanjing Medical UniversityNanjingChina
| | - Zhuqing Rao
- Department of AnesthesiologyThe First Affiliated Hospital of Nanjing Medical UniversityNanjingChina
| |
Collapse
|
66
|
Wang Y, Zhao Y, Gang Q, Hao H, Gao F, Deng J, Wang Z, Zhang W, Yuan Y, Zheng Y. Circulating cell-free DNA promotes inflammation in dermatomyositis patients with anti-NXP2 antibodies via the cGAS/STING pathway. Rheumatology (Oxford) 2025; 64:2272-2281. [PMID: 39110532 DOI: 10.1093/rheumatology/keae425] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2024] [Accepted: 07/26/2024] [Indexed: 04/04/2025] Open
Abstract
OBJECTIVES DM is a rare type I IFN (IFN-I)-driven autoimmune disease, and anti-nuclear matrix protein 2 (NXP2) antibody is related to severe muscle disease and poor prognosis. Circulating cell-free DNA (ccf-DNA), including ccf-mitochondrial DNA and ccf-nuclear DNA, activates the cGAS/STING pathway to induce IFN-I production in autoimmune diseases. We investigated whether serum-derived ccf-DNA had a pathogenic effect on skeletal muscle in anti-NXP2 antibody-positive DM. METHODS Serum ccf-DNA levels were measured, and correlations between ccf-DNA and clinicopathological indicators were performed. RNA sequencing, immunofluorescence, western blotting and reverse transcriptase quantitative polymerase chain reaction were performed on skeletal muscle samples. The serum-induced expression of p-STING in C2C12 cells was assessed in vitro. RESULTS We found that increased ccf-DNA levels were positively correlated with MYOACT scores in anti-NXP2 antibody-positive DM. RNA sequencing and immunofluorescence results revealed that the cytosolic DNA-sensing pathway was upregulated and that increased cytosolic dsDNA was colocalized with cGAS in skeletal muscle in anti-NXP2 antibody-positive DM. Western blot analysis revealed activation of the cGAS/STING pathway in patients with perifascicular atrophy (PFA) but not in patients without PFA. Reverse transcriptase quantitative polymerase chain reaction showed increased IFN-I scores in both patients with PFA and patients without PFA. Sera from patients with PFA increased p-STING expression in C2C12 cells, and DNase I treatment and STING inhibitor efficiently inhibited p-STING expression, respectively. CONCLUSION Increased ccf-DNA levels may be potential biomarkers for monitoring disease activity in anti-NXP2 antibody-positive DM. Activation of the cGAS/STING pathway is associated with PFA. Our findings identified a pathogenic effect of ccf-DNA on skeletal muscle via the cGAS/STING pathway.
Collapse
Affiliation(s)
- Yikang Wang
- Department of Neurology, Peking University First Hospital, Beijing, China
| | - Yawen Zhao
- Department of Neurology, Peking University First Hospital, Beijing, China
| | - Qiang Gang
- Department of Neurology, Peking University First Hospital, Beijing, China
| | - Hongjun Hao
- Department of Neurology, Peking University First Hospital, Beijing, China
| | - Feng Gao
- Department of Neurology, Peking University First Hospital, Beijing, China
| | - Jianwen Deng
- Department of Neurology, Peking University First Hospital, Beijing, China
- Beijing Key Laboratory of Neurovascular Disease Discovery, Beijing, China
- Key Laboratory for Neuroscience, Ministry of Education/National Health Commission, Peking University, Beijing, China
| | - Zhaoxia Wang
- Department of Neurology, Peking University First Hospital, Beijing, China
- Beijing Key Laboratory of Neurovascular Disease Discovery, Beijing, China
- Key Laboratory for Neuroscience, Ministry of Education/National Health Commission, Peking University, Beijing, China
| | - Wei Zhang
- Department of Neurology, Peking University First Hospital, Beijing, China
- Beijing Key Laboratory of Neurovascular Disease Discovery, Beijing, China
| | - Yun Yuan
- Department of Neurology, Peking University First Hospital, Beijing, China
- Beijing Key Laboratory of Neurovascular Disease Discovery, Beijing, China
- Key Laboratory for Neuroscience, Ministry of Education/National Health Commission, Peking University, Beijing, China
| | - Yiming Zheng
- Department of Neurology, Peking University First Hospital, Beijing, China
| |
Collapse
|
67
|
Zhou Y, Wei Y, Li L, Yan T, Ye H. Optogenetics in medicine: innovations and therapeutic applications. Curr Opin Biotechnol 2025; 92:103262. [PMID: 39842144 DOI: 10.1016/j.copbio.2025.103262] [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: 12/01/2024] [Revised: 01/06/2025] [Accepted: 01/07/2025] [Indexed: 01/24/2025]
Abstract
Optogenetics, an innovative approach integrating photonics and genetic engineering, enables precise control over molecular and cellular processes, opening up exciting new opportunities for precision-guided medicine. In this review, we highlight recent advances in optogenetic tools and their applications across a range of medical conditions, including vision restoration in retinitis pigmentosa via light-activated ion channels, precise immune response modulation in cancer immunotherapy, and blood glucose management in diabetes through controllable drug release. Optogenetics also plays a critical role in bioelectronic medicine, enabling seamless communication between electronic systems and biological tissues to enhance therapeutic precision. Finally, we discuss the challenges and potential transition of optogenetics from experimental models to clinical therapies, emphasizing its immense potential to transform future medical treatments.
Collapse
Affiliation(s)
- Yang Zhou
- Shanghai Frontiers Science Center of Genome Editing and Cell Therapy, Biomedical Synthetic Biology Research Center, Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai 200241, China; Wuhu Hospital, Health Science Center, East China Normal University, Wuhu 241001, China
| | - Yu Wei
- Shanghai Frontiers Science Center of Genome Editing and Cell Therapy, Biomedical Synthetic Biology Research Center, Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai 200241, China
| | - Lei Li
- Shanghai Frontiers Science Center of Genome Editing and Cell Therapy, Biomedical Synthetic Biology Research Center, Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai 200241, China
| | - Tao Yan
- Shanghai Frontiers Science Center of Genome Editing and Cell Therapy, Biomedical Synthetic Biology Research Center, Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai 200241, China
| | - Haifeng Ye
- Shanghai Frontiers Science Center of Genome Editing and Cell Therapy, Biomedical Synthetic Biology Research Center, Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai 200241, China; Wuhu Hospital, Health Science Center, East China Normal University, Wuhu 241001, China; Shanghai Academy of Natural Sciences, East China Normal University, Shanghai, China.
| |
Collapse
|
68
|
Li C, Ma A, Bai Y, Liu Z, Tian L, Wang Z, Ma H, Chen Z, Gao Z, Feng S, Fu P. TRIM21 promotes type I interferon by inhibiting the autophagic degradation of STING via p62/SQSTM1 ubiquitination in systemic lupus erythematosus. Acta Biochim Biophys Sin (Shanghai) 2025. [PMID: 40165656 DOI: 10.3724/abbs.2025046] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/02/2025] Open
Abstract
The cGAS-STING signaling pathway serves as a pivotal surveillance mechanism for cytosolic double-stranded DNA (dsDNA) detection in mammalian systems. While STING-mediated type I interferon production is crucial for host defense, sustained activation of this pathway contributes to autoimmune pathologies, including systemic lupus erythematosus (SLE). Maintaining immune homeostasis requires precise regulation of STING activity to prevent hyperactivation. Our study identifies TRIM21 as a novel positive regulator of cGAS-STING signaling in SLE pathogenesis. Our results demonstrate that TRIM21 overexpression stabilizes STING by suppressing autophagic degradation, whereas TRIM21 depletion accelerates this clearance process. Mechanistically, TRIM21 catalyzes the K63-linked polyubiquitylation of the selective autophagy receptor p62/SQSTM1, disrupting its interaction with STING. This post-translational modification prevents the sequestration of STING into autophagosomes, thereby stabilizing the adaptor protein and amplifying downstream type I interferon responses. Our findings reveal a previously unrecognized regulatory circuit in which TRIM21 orchestrates cross-talk between ubiquitin signaling and autophagy to control STING turnover. The TRIM21-p62 axis represents a potential therapeutic target for attenuating pathological interferon production in STING-dependent autoimmune disorders. This work advances our understanding of immune regulation by demonstrating how E3 ligase-mediated ubiquitin modifications modulate cargo recognition in selective autophagy pathways. The identified mechanism provides new insights into the molecular interplay between protein ubiquitylation and autophagic degradation in maintaining the innate immune balance, offering novel perspectives for developing targeted therapies against interferonopathies associated with cGAS-STING hyperactivity.
Collapse
Affiliation(s)
- Chen Li
- Department of Rheumatology and Clinical Immunology, the Second Affiliated Hospital of Kunming Medical University, Kunming 650032, China
- Department of Scientific Research, Yunnan Cancer Hospital, the Third Affiliated Hospital of Kunming Medical University, Peking University Cancer Hospital Yunnan, Kunming 650118, China
| | - Ang Ma
- Department of Rheumatology and Clinical Immunology, the Second Affiliated Hospital of Kunming Medical University, Kunming 650032, China
| | - Yu Bai
- Department of Urology, Yunnan Cancer Hospital, the Third Affiliated Hospital of Kunming Medical University, Peking University Cancer Hospital Yunnan, Kunming 650118, China
| | - Zitao Liu
- Department of Scientific Research, Yunnan Cancer Hospital, the Third Affiliated Hospital of Kunming Medical University, Peking University Cancer Hospital Yunnan, Kunming 650118, China
| | - Linghan Tian
- Department of Scientific Research, Yunnan Cancer Hospital, the Third Affiliated Hospital of Kunming Medical University, Peking University Cancer Hospital Yunnan, Kunming 650118, China
| | - Ziyuan Wang
- Cancer Institute, Yunnan Cancer Hospital, the Third Affiliated Hospital of Kunming Medical University, Peking University Cancer Hospital, Kunming 650118, China
| | - Huaishun Ma
- Department of Scientific Research, Yunnan Cancer Hospital, the Third Affiliated Hospital of Kunming Medical University, Peking University Cancer Hospital Yunnan, Kunming 650118, China
| | - Zhengpu Chen
- Department of Scientific Research, Yunnan Cancer Hospital, the Third Affiliated Hospital of Kunming Medical University, Peking University Cancer Hospital Yunnan, Kunming 650118, China
| | - Zhengheng Gao
- Department of Health Management and Tumor Screening Center, Yunnan Cancer Hospital, the Third Affiliated Hospital of Kunming Medical University, Peking University Cancer Hospital Yunnan, Kunming 650118, China
| | - Shijie Feng
- Department of Rheumatology and Clinical Immunology, the Second Affiliated Hospital of Kunming Medical University, Kunming 650032, China
| | - Ping Fu
- Department of Rheumatology and Clinical Immunology, the Second Affiliated Hospital of Kunming Medical University, Kunming 650032, China
| |
Collapse
|
69
|
Wang S, Wang L, Zhao Y. ALDH1A3 Regulates Cellular Senescence and Senescence-Associated Secretome in Prostate Cancer. Cancers (Basel) 2025; 17:1184. [PMID: 40227735 PMCID: PMC11987895 DOI: 10.3390/cancers17071184] [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: 01/14/2025] [Revised: 03/19/2025] [Accepted: 03/21/2025] [Indexed: 04/15/2025] Open
Abstract
Background: Radiotherapy is a key treatment for cancer, effectively controlling local tumor growth through DNA damage that induces senescence or apoptosis in cancer cells. However, radiotherapy can trigger complex cellular reactions, such as cell senescence, which is characterized by irreversible cell cycle arrest and the secretion of pro-inflammatory factors known as the senescent-associated secretory phenotype (SASP). Methods: This study investigates the regulatory role of ALDH1A3, a key enzyme implicated in cancer cell metabolism and radiotherapy resistance, in the induction of senescence and SASP. Using in vitro models, we demonstrate that ALDH1A3 knockdown accelerates cellular senescent-like phenotype while regulating the SASP through the cGAS-STING immune response pathway. Results: Our results indicate that while ALDH1A3 knockdown promotes senescence, it reduces the secretion of pro-inflammatory factors via inhibition of the cGAS-STING pathway, potentially mitigating SASP-related tumor progression. Conclusions: These findings provide insights into the molecular mechanisms underlying prostate cancer cell senescence and suggest that ALDH1A3 could be a potential therapeutic target to enhance the efficacy of radiotherapy while controlling the adverse effects of SASP.
Collapse
Affiliation(s)
- Sen Wang
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 300192, China;
| | - Lin Wang
- School of Medicine, Nankai University, 94 Weijin Road, Tianjin 300071, China;
| | - Yu Zhao
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 300192, China;
| |
Collapse
|
70
|
Li M, Jin S, Ma H, Yang X, Zhang Z. Reciprocal regulation between ferroptosis and STING-type I interferon pathway suppresses head and neck squamous cell carcinoma growth through dendritic cell maturation. Oncogene 2025:10.1038/s41388-025-03368-2. [PMID: 40164871 DOI: 10.1038/s41388-025-03368-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2024] [Revised: 02/23/2025] [Accepted: 03/20/2025] [Indexed: 04/02/2025]
Abstract
Head and neck squamous cell carcinoma (HNSCC) presents a serious clinical challenge mainly due to its resistance to conventional therapies and its complex, immunosuppressive tumor microenvironment. While recent studies have identified ferroptosis as a new therapeutic option, its impact on the immune microenvironment in HNSCC remains controversial, which may hinder its translational application. Although the role of the stimulator of interferon genes (STING)-type I interferon (IFN-I) pathway in antitumor immune responses has been widely investigated, its relationship with ferroptosis in HNSCC has not been fully explored. In this study, we discovered that ferroptosis in HNSCC inhibited tumor growth, activated STING-IFN-I pathway and subsequently improved recruitment and maturation of dendritic cells. We further demonstrated that IFN-I could enhance ferroptosis by inhibiting xCT-glutathione peroxidase 4 (GPX4) antioxidant system. To harness this positive feedback loop, we treated HNSCC tumors with both ferroptosis inducer and STING agonist, resulting in significant tumor suppression, elevated ferroptosis levels and enhanced dendritic cell infiltration. Overall, our findings reveal a mutually regulatory relationship between ferroptosis and the intrinsic STING-IFN-I pathway, providing novel insights into immune-mediated tumor suppression and suggesting its potential as therapeutic approach in HNSCC.
Collapse
Affiliation(s)
- Mingyu Li
- Department of Oral Maxillofacial-Head and Neck Oncology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, College of Stomatology, Shanghai Jiao Tong University, No. 639, Zhizaoju Rd, Shanghai, 200011, China
- National Center for Stomatology; National Clinical Research Center for Oral Diseases; Shanghai Key Laboratory of Stomatology. No. 639, Zhizaoju Rd, Shanghai, 200011, China
- Shanghai Research Institute of Stomatology; Research Unit of Oral and Maxillofacial Regenerative Medicine, Chinese Academy of Medical Sciences, No. 639, Zhizaoju Rd, Shanghai, 200011, China
| | - Shufang Jin
- National Center for Stomatology; National Clinical Research Center for Oral Diseases; Shanghai Key Laboratory of Stomatology. No. 639, Zhizaoju Rd, Shanghai, 200011, China
- Shanghai Research Institute of Stomatology; Research Unit of Oral and Maxillofacial Regenerative Medicine, Chinese Academy of Medical Sciences, No. 639, Zhizaoju Rd, Shanghai, 200011, China
- Department of Second Dental Center, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, College of Stomatology, Shanghai Jiao Tong University, Shanghai, China
| | - Hailong Ma
- Department of Oral Maxillofacial-Head and Neck Oncology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, College of Stomatology, Shanghai Jiao Tong University, No. 639, Zhizaoju Rd, Shanghai, 200011, China.
- National Center for Stomatology; National Clinical Research Center for Oral Diseases; Shanghai Key Laboratory of Stomatology. No. 639, Zhizaoju Rd, Shanghai, 200011, China.
- Shanghai Research Institute of Stomatology; Research Unit of Oral and Maxillofacial Regenerative Medicine, Chinese Academy of Medical Sciences, No. 639, Zhizaoju Rd, Shanghai, 200011, China.
| | - Xi Yang
- Department of Oral Maxillofacial-Head and Neck Oncology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, College of Stomatology, Shanghai Jiao Tong University, No. 639, Zhizaoju Rd, Shanghai, 200011, China.
- National Center for Stomatology; National Clinical Research Center for Oral Diseases; Shanghai Key Laboratory of Stomatology. No. 639, Zhizaoju Rd, Shanghai, 200011, China.
- Shanghai Research Institute of Stomatology; Research Unit of Oral and Maxillofacial Regenerative Medicine, Chinese Academy of Medical Sciences, No. 639, Zhizaoju Rd, Shanghai, 200011, China.
| | - Zhiyuan Zhang
- Department of Oral Maxillofacial-Head and Neck Oncology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, College of Stomatology, Shanghai Jiao Tong University, No. 639, Zhizaoju Rd, Shanghai, 200011, China.
- National Center for Stomatology; National Clinical Research Center for Oral Diseases; Shanghai Key Laboratory of Stomatology. No. 639, Zhizaoju Rd, Shanghai, 200011, China.
- Shanghai Research Institute of Stomatology; Research Unit of Oral and Maxillofacial Regenerative Medicine, Chinese Academy of Medical Sciences, No. 639, Zhizaoju Rd, Shanghai, 200011, China.
| |
Collapse
|
71
|
Ji J, Li Y, Xu T, Shao Q, Sun Z, Chen S, Zhang D, Wang Q, Wang X, Ma C, Cheng F. Protective effects of berberine on MASLD: regulation of glucose and lipid metabolism through PI3K/Akt and STING pathways. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025:10.1007/s00210-025-04077-z. [PMID: 40146248 DOI: 10.1007/s00210-025-04077-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Subscribe] [Scholar Register] [Received: 01/15/2025] [Accepted: 03/18/2025] [Indexed: 03/28/2025]
Abstract
This study is aimed at exploring the therapeutic potential of berberine (BBR) in mitigating metabolic dysfunction-associated steatotic liver disease (MASLD) and at elucidating its mechanisms of action, with a focus on the modulation of glucose and lipid metabolism via the PI3K/Akt and STING signaling pathways. Male C57BL/6 J mice were fed a high-fat diet (HFD) to induce MASLD and subsequently treated with BBR or metformin. HepG2 cells were cultured in vitro, and palmitic acid (PA) was used to construct the cell model. Comprehensive analyses, including network pharmacology, transcriptome sequencing, and Western blotting, were conducted to identify critical pathways and molecular targets. Biochemical, histological, and molecular assays were performed to evaluate metabolic and inflammatory responses. BBR significantly attenuated HFD-induced hepatic steatosis, inflammation, and glucose intolerance. It effectively reduced lipid accumulation, enhanced insulin sensitivity, and modulated the expression of genes involved in lipid metabolism. Network pharmacology and transcriptome analysis highlighted the involvement of the PI3K/Akt and STING pathways. BBR activated PI3K/Akt signaling while suppressing the STING pathway, thereby reducing lipid accumulation in both in vivo and in vitro models. The inhibition of AKT negated the beneficial effects of BBR, underscoring the pivotal role of PI3K/Akt in regulating STING signaling. BBR ameliorates MASLD by activating the PI3K/Akt pathway and inhibiting the STING pathway, leading to improved glucose and lipid metabolism. These findings position BBR as a promising therapeutic candidate for the treatment of MASLD.
Collapse
Affiliation(s)
- Jing Ji
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Ying Li
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Tian Xu
- Department of Gastroenterology, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Qi Shao
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Zijin Sun
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Simin Chen
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Di Zhang
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Qingguo Wang
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Xueqian Wang
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China.
| | - Chongyang Ma
- School of Traditional Chinese Medicine, Capital Medical University, Beijing, China.
| | - Fafeng Cheng
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China.
| |
Collapse
|
72
|
Phillips CM, Smyth JW. Viral Infection and Connexin Dysfunction in the Heart. Curr Cardiol Rep 2025; 27:76. [PMID: 40146392 PMCID: PMC11950093 DOI: 10.1007/s11886-025-02227-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 03/11/2025] [Indexed: 03/28/2025]
Abstract
PURPOSE OF REVIEW Gap junctions, comprising connexin proteins, enable the direct intercellular electrical coupling of cardiomyocytes, and disruption of this process is arrhythmogenic. In addition, gap junctions effect metabolic coupling and of relevance to this review, propagate host antiviral immune responses. Accordingly, connexins have emerged as viral targets during infection. This review summarizes current knowledge regarding contributions of inflammation vs virally encoded factors in driving alterations to cardiac gap junction function. RECENT FINDINGS In addition to host immune-mediated effects on cardiac electrophysiology and gap junctions in myocarditis, there is now increasing appreciation for virally encoded factors targeting connexin function in acute/active infection. We now know diverse viral species have independently evolved to directly target connexin function during infection. Understanding both the direct and indirect effects of viral infection on cardiac gap junctions is critical to inform treatment strategies and development of novel therapeutics for acute infection as a distinct disease process from chronic myocarditis.
Collapse
Affiliation(s)
- Chelsea M Phillips
- Fralin Biomedical Research Institute at Virginia Tech Carilion, Roanoke, VA, 24016, USA
| | - James W Smyth
- Fralin Biomedical Research Institute at Virginia Tech Carilion, Roanoke, VA, 24016, USA.
- Department of Biological Sciences, College of Science, Virginia Tech, Blacksburg, VA, 24061, USA.
- Department of Basic Science Education, Virginia Tech Carilion School of Medicine, Roanoke, VA, 24016, USA.
- Center for Vascular and Heart Research, Fralin Biomedical Research Institute at Virginia Tech Carilion, Roanoke, VA, 24016, USA.
- Department of Biomedical Engineering and Mechanics, College of Engineering, Virginia Tech, Blacksburg, VA, 24061, USA.
| |
Collapse
|
73
|
Jin S, He Y, Feng C, Yuan J, Guo Y, Guo Z, Wang X. Cellular Discrepancy of Platinum Complexes in Interfering with Mitochondrial DNA. ACS CENTRAL SCIENCE 2025; 11:393-403. [PMID: 40161961 PMCID: PMC11950849 DOI: 10.1021/acscentsci.4c01941] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/12/2024] [Revised: 01/06/2025] [Accepted: 01/07/2025] [Indexed: 04/02/2025]
Abstract
Mitochondria are associated with cellular energy metabolism, proliferation, and mode of death. Damage to mitochondrial DNA (mtDNA) greatly affects mitochondrial function by interfering with energy production and the signaling pathway. Monofunctional trinuclear platinum complex MTPC demonstrates different actions on the mtDNA of cancerous and normal cells. It severely impairs the integrity and function of mitochondria in the human lung cancer A549 cells, such as dissipating mitochondrial membrane potential, decreasing the copy number of mtDNA, interfering in nucleoid proteins and polymerase gamma gene, reducing adenosine triphosphate (ATP), and inducing mitophagy, whereas it barely affects the mtDNA of the human kidney 2 (HK-2) cells. Moreover, MTPC promotes the release of mtDNA into the cytosol and stimulates the cyclic GMP-AMP synthase-stimulator of interferon genes (cGAS-STING) pathway, thus showing the potential to trigger antitumor immunity. MTPC displays significant cytotoxicity against A549 cells, while it exhibits weak toxicity toward HK-2 cells, therefore displaying great advantage to overcome the lingering nephrotoxicity of platinum anticancer drugs. Discrepant effects of a metal complex on mitochondria of different cells mean that targeting mitochondria has special significance in cancer therapy.
Collapse
Affiliation(s)
- Suxing Jin
- School
of Food Science and Pharmaceutical Engineering, Nanjing Normal University, Nanjing 210023, P. R. China
- State
Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing 210023, P. R. China
| | - Yafeng He
- State
Key Laboratory of Coordination Chemistry, School of Chemistry and
Chemical Engineering, Nanjing University, Nanjing 210023, P. R. China
| | - Chenyao Feng
- State
Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing 210023, P. R. China
| | - Jian Yuan
- State
Key Laboratory of Coordination Chemistry, School of Chemistry and
Chemical Engineering, Nanjing University, Nanjing 210023, P. R. China
| | - Yan Guo
- State
Key Laboratory of Coordination Chemistry, School of Chemistry and
Chemical Engineering, Nanjing University, Nanjing 210023, P. R. China
| | - Zijian Guo
- State
Key Laboratory of Coordination Chemistry, School of Chemistry and
Chemical Engineering, Nanjing University, Nanjing 210023, P. R. China
| | - Xiaoyong Wang
- State
Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing 210023, P. R. China
| |
Collapse
|
74
|
Chen L, Hu L, Chang H, Mao J, Ye M, Jin X. DNA-RNA hybrids in inflammation: sources, immune response, and therapeutic implications. J Mol Med (Berl) 2025:10.1007/s00109-025-02533-0. [PMID: 40131443 DOI: 10.1007/s00109-025-02533-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Revised: 03/13/2025] [Accepted: 03/14/2025] [Indexed: 03/27/2025]
Abstract
Cytoplasmic DNA-RNA hybrids are emerging as important immunogenic nucleic acids, that were previously underappreciated. DNA-RNA hybrids, formed during cellular processes like transcription and replication, or by exogenous pathogens, are recognized by pattern recognition receptors (PRRs), including cGAS, DDX41, and TLR9, which trigger immune responses. Post-translational modifications (PTMs) including ubiquitination, phosphorylation, acetylation, and palmitoylation regulate the activity of PRRs and downstream signaling molecules, fine-tuning the immune response. Targeting enzymes involved in DNA-RNA hybrid metabolism and PTMs regulation offers therapeutic potential for inflammatory diseases. Herein, we discuss the sources, immune response, and therapeutic implications of DNA-RNA hybrids in inflammation, highlighting the significance of DNA-RNA hybrids as potential targets for the treatment of inflammation.
Collapse
Affiliation(s)
- Litao Chen
- Department of Biochemistry and Molecular Biology, Zhejiang Key Laboratory of Pathophysiology, Health Science Center, Ningbo University, Ningbo, 315211, China
| | - Lechen Hu
- Department of Biochemistry and Molecular Biology, Zhejiang Key Laboratory of Pathophysiology, Health Science Center, Ningbo University, Ningbo, 315211, China
| | - Han Chang
- Department of Biochemistry and Molecular Biology, Zhejiang Key Laboratory of Pathophysiology, Health Science Center, Ningbo University, Ningbo, 315211, China
| | - Jianing Mao
- Department of Biochemistry and Molecular Biology, Zhejiang Key Laboratory of Pathophysiology, Health Science Center, Ningbo University, Ningbo, 315211, China
| | - Meng Ye
- Department of Biochemistry and Molecular Biology, Zhejiang Key Laboratory of Pathophysiology, Health Science Center, Ningbo University, Ningbo, 315211, China.
| | - Xiaofeng Jin
- Department of Biochemistry and Molecular Biology, Zhejiang Key Laboratory of Pathophysiology, Health Science Center, Ningbo University, Ningbo, 315211, China.
| |
Collapse
|
75
|
Wang Y, Zhou M, Jiang RY, Zhu CL. Pharmacological inhibition of STING-mediated GPX4 autophagic degradation by 4-octyl itaconate ameliorates sepsis-induced acute kidney injury. Apoptosis 2025:10.1007/s10495-025-02099-9. [PMID: 40119983 DOI: 10.1007/s10495-025-02099-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/04/2025] [Indexed: 03/25/2025]
Abstract
The precise pathogenic mechanisms underlying sepsis-induced acute kidney injury (AKI) remain elusive. Emerging evidence suggests a link between tubular ferroptosis and the pathogenesis of AKI, though the regulatory pathways are not fully understood. Stimulator of interferon genes (STING), previously recognized as a pivotal mediator of innate immunity via DNA-sensing pathways, is increasingly associated with lipid peroxidation, a hallmark of ferroptosis, and 4-octyl itaconate (4-OI) has been shown to inhibit STING activation, exerting anti-inflammatory effects. This study investigates the protective mechanisms of 4-OI in sepsis-AKI. Following cecal ligation and puncture (CLP), inflammation, oxidative stress, and ferroptosis levels in kidney tissue increased. Both 4-OI and ferrostatin-1 (Fer-1) mitigated renal ferroptosis, exerting anti-inflammatory and antioxidant stress effects, and improved renal function. Consistently, in vitro experiments demonstrated that 4-OI reduced ferroptosis in human renal proximal tubule (HK-2) cells induced by lipopolysaccharide (LPS). Mechanistically, 4-OI suppressed LPS-induced activation of the STING pathway and reduced levels of inflammatory cytokines in a manner independent of NF-E2-related factor 2 (Nrf2). Additionally, 4-OI inhibited STING transcription through the activation of Nrf2. These dual actions effectively suppressed LPS-induced STING pathway activation, thereby inhibiting STING-mediated autophagic degradation of glutathione peroxidase 4 (GPX4), reducing reactive oxygen species (ROS) accumulation, and alleviating ferroptosis. In summary, 4-OI is a promising therapeutic candidate, functioning both as a STING inhibitor and a ferroptosis inhibitor, with potential applications in the treatment of sepsis.
Collapse
Affiliation(s)
- Yiyang Wang
- Chongqing Medical University-University of Leicester Joint Institute, Chongqing, 400016, China
| | - Miao Zhou
- Department of Anesthesiology, Jiangsu Cancer Hospital, The Affiliated Cancer Hospital of Nanjing Medical University, Jiangsu Institute of Cancer Research, Nanjing Medical University, Nanjing, Jiangsu, 210009, China
| | - Ruo-Yu Jiang
- Department of Biochemistry and Molecular Biology, College of Basic Medical Sciences, Naval Medical University, Shanghai, 200433, China.
| | - Cheng-Long Zhu
- School of Anesthesiology, Naval Medical University, Shanghai, 200433, China.
| |
Collapse
|
76
|
Skeldon AM, Wang L, Sgarioto N, Beveridge RE, Chan S, Dorich S, Dumais V, Fradet N, Gaudreault S, LeGros P, McKay D, Seliniotakis R, Sietsema DV, Zhang L, Boily MO, Burch JD, Caron A, Fader LD, Lama L, Xie W, Patel DJ, Tuschl T, Crackower MA, Pike KA. Structural insight into the cGAS active site explains differences between therapeutically relevant species. Commun Chem 2025; 8:88. [PMID: 40121343 PMCID: PMC11929900 DOI: 10.1038/s42004-025-01481-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2024] [Accepted: 03/03/2025] [Indexed: 03/25/2025] Open
Abstract
Cyclic GMP-AMP synthase (cGAS) is an intracellular sensor of double-stranded DNA that triggers a pro-inflammatory response upon binding. The interest in cGAS as a drug discovery target has increased substantially over the past decade due to growing evidence linking its activation to numerous peripheral and neurological diseases. Here, we report the binding mode of previously described cGAS inhibitors while also uncovering the structural basis for the interspecies potency shifts within this chemotype. A single threonine to isoleucine substitution between human and mouse cGAS drives compound activity, as demonstrated by biochemical, cellular, and in vivo studies. Finally, we utilize a structurally enabled design approach to engineer a novel chemical inhibitor with excellent potency for both human and mouse enzymes by targeting key interactions within the enzyme active site. Overall, this work provides the framework for rational optimization of cGAS inhibitors and potential preclinical translational strategies.
Collapse
Affiliation(s)
| | - Li Wang
- Ventus Therapeutics, Waltham, MA, USA
| | | | | | - Silas Chan
- Ventus Therapeutics, Saint-Laurent, QC, Canada
| | | | | | | | | | | | | | | | | | | | | | | | - Alex Caron
- Ventus Therapeutics, Saint-Laurent, QC, Canada
| | - Lee D Fader
- Ventus Therapeutics, Saint-Laurent, QC, Canada
| | - Lodoe Lama
- Laboratory for RNA Molecular Biology, The Rockefeller University, New York, NY, USA
| | - Wei Xie
- Structural Biology Program, Memorial Sloan-Kettering Cancer Center, New York, NY, USA
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Dinshaw J Patel
- Structural Biology Program, Memorial Sloan-Kettering Cancer Center, New York, NY, USA
| | - Thomas Tuschl
- Laboratory for RNA Molecular Biology, The Rockefeller University, New York, NY, USA
| | | | | |
Collapse
|
77
|
Ren KX, Feng L, Wu P, Liu Y, Ren HM, Jin XW, Zhong CB, Zhou XQ, Jiang WD. Mitigation of the toxic effects of nitrite: Role and mechanism of isoleucine in mitigating mitochondrial DNA leakage-induced inflammation in grass carp (Ctenopharyngodon idella) under nitrite exposure. JOURNAL OF HAZARDOUS MATERIALS 2025; 491:138016. [PMID: 40147124 DOI: 10.1016/j.jhazmat.2025.138016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/26/2024] [Revised: 03/12/2025] [Accepted: 03/19/2025] [Indexed: 03/29/2025]
Abstract
The physiological and growth processes of fish are closely associated with their surrounding environment. This study investigated the role and underlying mechanisms of isoleucine (Ile) in alleviating mitochondrial DNA (mtDNA) leakage-induced inflammation in grass carp under nitrite exposure. Grass carp were fed six experimental diets containing different Ile levels (0.00, 3.00, 6.00, 9.00, 12.00 and 15.00 g/kg) for 9 weeks, followed by a 96-hour nitrite exposure trial. Ile supplementation mitigated the deterioration of blood parameters including glutamic oxaloacetic transaminase (GOT), glutamic alanine transaminase (GPT), glucose, cortisol and lactate dehydrogenase (LDH) induced by nitrite exposure. Additionally, Ile enhanced its transport to the liver and mitochondria, as well as increased metabolism of Ile in mitochondria. Histological analyses revealed that Ile mitigated nitrite exposure-induced liver damage and mitochondrial cristae disruption. Furthermore, Ile preserved the mitochondrial cristae homeostasis by upregulating key proteins involved in mitochondrial structure maintenance, while inhibiting mtDNA leakage. Mechanistically, Ile attenuated mtDNA leakage-induced inflammation under nitrite exposure associated with the cyclic guanosine monophosphate-adenosine monophosphate synthase (cGAS)-Stimulator of Interferon Genes (STING) and NOD-like receptor thermal protein domain associated protein 3 (NLRP3) pathways. These findings highlight the protective role of Ile in reducing inflammation triggered by environmental nitrite exposure, offering new insights into aquatic toxicology, and determined that Ile concentration of 11.13 g/kg diet could be optimal for mitigating nitrite-induced stress in grass carp, providing a theoretical basis for formulating anti- nitrite stress diets in aquaculture.
Collapse
Affiliation(s)
- Kai-Xuan Ren
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, China
| | - Lin Feng
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, China; Fish Nutrition and Safety Production University Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China; Key Laboratory of Animal Disease-Resistance Nutrition, Ministry of Education, Ministry of Agriculture and Rural Affairs, Key Laboratory of Sichuan Province, Sichuan 611130, China
| | - Pei Wu
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, China; Fish Nutrition and Safety Production University Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China; Key Laboratory of Animal Disease-Resistance Nutrition, Ministry of Education, Ministry of Agriculture and Rural Affairs, Key Laboratory of Sichuan Province, Sichuan 611130, China
| | - Yang Liu
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, China; Fish Nutrition and Safety Production University Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China; Key Laboratory of Animal Disease-Resistance Nutrition, Ministry of Education, Ministry of Agriculture and Rural Affairs, Key Laboratory of Sichuan Province, Sichuan 611130, China
| | - Hong-Mei Ren
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, China
| | - Xiao-Wan Jin
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, China
| | - Cheng-Bo Zhong
- Sichuan Animal Science Academy, Sichuan Animtech Feed Co.Ltd, Chengdu 610066, China
| | - Xiao-Qiu Zhou
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, China; Fish Nutrition and Safety Production University Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China; Key Laboratory of Animal Disease-Resistance Nutrition, Ministry of Education, Ministry of Agriculture and Rural Affairs, Key Laboratory of Sichuan Province, Sichuan 611130, China.
| | - Wei-Dan Jiang
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, China; Fish Nutrition and Safety Production University Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China; Key Laboratory of Animal Disease-Resistance Nutrition, Ministry of Education, Ministry of Agriculture and Rural Affairs, Key Laboratory of Sichuan Province, Sichuan 611130, China.
| |
Collapse
|
78
|
Yang R, Deng MY, Yang LK, Wang GZ, Ma J, Wen Q, Gao N, Qiao HL. Identification of cytochrome P450 2E1 as a novel target in neuroinflammation and development of its inhibitor Q11 as a treatment strategy. Free Radic Biol Med 2025; 234:220-232. [PMID: 40122152 DOI: 10.1016/j.freeradbiomed.2025.03.032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/27/2025] [Revised: 03/12/2025] [Accepted: 03/21/2025] [Indexed: 03/25/2025]
Abstract
Neuroinflammation is implicated in nearly all pathological processes of central nervous system (CNS) diseases. However, no medications specifically targeting neuroinflammation are clinically available, and conventional anti-inflammatory drugs exhibit limited efficacy. Here, we identified cytochrome P450 2E1 (CYP2E1) as a novel therapeutic target in neuroinflammation. Elevated CYP2E1 levels were observed in hippocampal tissues of mouse and rat neuroinflammation models, as well as in LPS-stimulated primary microglia. Genetic ablation of Cyp2e1 improved spatial learning and memory in neuroinflammatory rats and reduced pro-inflammatory cytokine levels in Cyp2e1-deficient microglia. Furthermore, Q11 (1-(4-methyl-5-thiazolyl) ethanone), a novel CYP2E1 inhibitor developed and synthesized in our laboratory, effectively ameliorated Alzheimer's disease-related spatial learning and memory functions and depression-related anxiety-like behaviors in mice/rats. Mechanistically, Q11 attenuated microglial activation, neuronal damage, oxidative stress, and neuroinflammation by suppressing the PI3K/Akt, STAT1/3, and NF-κB signaling pathways. These findings establish CYP2E1 as a druggable target for neuroinflammation and propose Q11 as a promising candidate for treating neuroinflammation-related diseases.
Collapse
Affiliation(s)
- Rui Yang
- Institute of Clinical Pharmacology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450001, Henan, China
| | - Meng-Yan Deng
- Institute of Clinical Pharmacology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450001, Henan, China
| | - Lu-Kui Yang
- Institute of Clinical Pharmacology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450001, Henan, China
| | - Guan-Zhe Wang
- Institute of Clinical Pharmacology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450001, Henan, China
| | - Jun Ma
- Institute of Clinical Pharmacology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450001, Henan, China
| | - Qiang Wen
- Institute of Clinical Pharmacology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450001, Henan, China
| | - Na Gao
- Institute of Clinical Pharmacology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450001, Henan, China
| | - Hai-Ling Qiao
- Institute of Clinical Pharmacology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450001, Henan, China.
| |
Collapse
|
79
|
Ju Y, Lv Y, Liu X, Lu J, Shi Y, Guo H, Xu S, Tian J, Yang J, Zhong J. Role of long non-coding RNAs in the regulation of ferroptosis in tumors. Front Immunol 2025; 16:1568567. [PMID: 40191204 PMCID: PMC11968707 DOI: 10.3389/fimmu.2025.1568567] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2025] [Accepted: 03/06/2025] [Indexed: 04/09/2025] Open
Abstract
Normal cells begin to grow indefinitely and immortalize to form tumor cells after an external stimulus resulting in a genetic mutation. Effective killing of tumor cells is the basis of various cancer therapies. Ferroptosis is a class of cell death types dependent on iron and cellular lipid peroxidation. Tumors themselves are iron-dependent, and conventional radiotherapy also sensitizes cancer cells to ferroptosis. Increasing the sensitivity of tumor cells to ferroptosis may be a potential therapeutic strategy to overcome the resistance mechanisms of conventional cancer therapy. Long noncoding RNAs (LncRNAs) are a class of transcripts more than 200 nucleotides in length that regulate gene expression at multiple levels and are involved in biological processes such as cell differentiation, cell cycle arrest, and maintenance of tumor stemness. Recent studies have found that lncRNAs regulate ferroptosis of tumor cells through multiple mechanisms and may influence or ameliorate tumor resistance to chemotherapeutic agents. With the continuous maturation of nanomaterials technology, it may provide new means for cancer treatment by regulating the levels of ferroptosis-related lncRNAs inside tumors as well as increasing the levels of Fe2+ and ROS inside tumors. In this paper, we systematically introduce the regulatory mechanism of lncRNAs in ferroptosis, the role of ferroptosis in tumor immunotherapy and the application of lncRNAs combined with ferroptosis in nanomaterials, which provides new perspectives for tumor therapy.
Collapse
Affiliation(s)
- Ying Ju
- Department of Gynecology, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
| | - Yuanhao Lv
- Department of Pathology, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
- Department of Pathology, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, China
| | - Xu Liu
- Department of Anesthesia and Perioperative Medicine, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
| | - Jing Lu
- Department of Gynecology, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
| | - Yashen Shi
- Department of Pathology, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
| | - Huimin Guo
- Department of Gynecology, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
| | - Siguang Xu
- Department of Pathology, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, China
| | - Jiaqi Tian
- Department of Pathology, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, China
| | - Jun Yang
- Department of Gynecology, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
| | - Jiateng Zhong
- Department of Pathology, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
- Department of Pathology, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, China
- Xinxiang Engineering Technology Research Center of Digestive Tumor Molecular Diagnosis, the First Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
| |
Collapse
|
80
|
Poddar S, Chauvin SD, Archer CH, Qian W, Castillo-Badillo JA, Yin X, Disbennett WM, Miner CA, Holley JA, Naismith TV, Stinson WA, Wei X, Ning Y, Fu J, Ochoa TA, Surve N, Zaver SA, Wodzanowski KA, Balka KR, Venkatraman R, Liu C, Rome K, Bailis W, Shiba Y, Cherry S, Shin S, Semenkovich CF, De Nardo D, Yoh S, Roberson EDO, Chanda SK, Kast DJ, Miner JJ. ArfGAP2 promotes STING proton channel activity, cytokine transit, and autoinflammation. Cell 2025; 188:1605-1622.e26. [PMID: 39947179 PMCID: PMC11928284 DOI: 10.1016/j.cell.2025.01.027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2024] [Revised: 11/03/2024] [Accepted: 01/17/2025] [Indexed: 02/23/2025]
Abstract
Stimulator of interferon genes (STING) transmits signals downstream of the cytosolic DNA sensor cyclic guanosine monophosphate-AMP synthase (cGAS), leading to transcriptional upregulation of cytokines. However, components of the STING signaling pathway, such as IRF3 and IFNAR1, are not essential for autoinflammatory disease in STING gain-of-function (STING-associated vasculopathy with onset in infancy [SAVI]) mice. Recent discoveries revealed that STING also functions as a proton channel that deacidifies the Golgi apparatus. Because pH impacts Golgi enzyme activity, protein maturation, and trafficking, we hypothesized that STING proton channel activity influences multiple Golgi functions. Here, we show that STING-mediated proton efflux non-transcriptionally regulates Golgi trafficking of protein cargos. This process requires the Golgi-associated protein ArfGAP2, a cell-type-specific dual regulator of STING-mediated proton efflux and signaling. Deletion of ArfGAP2 in hematopoietic and endothelial cells markedly reduces STING-mediated cytokine and chemokine secretion, immune cell activation, and autoinflammatory pathology in SAVI mice. Thus, ArfGAP2 facilitates STING-mediated signaling and cytokine release in hematopoietic cells, significantly contributing to autoinflammatory disease pathogenesis.
Collapse
Affiliation(s)
- Subhajit Poddar
- Division of Rheumatology, Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Samuel D Chauvin
- Division of Rheumatology, Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Christopher H Archer
- Department of Cell Biology and Physiology, Washington University School of Medicine, St Louis, MO 63110, USA
| | - Wei Qian
- Department of Medicine, Washington University School of Medicine, St Louis, MO 63110, USA
| | - Jean A Castillo-Badillo
- Department of Cell Biology and Physiology, Washington University School of Medicine, St Louis, MO 63110, USA
| | - Xin Yin
- Immunity and Pathogenesis Program, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA 92037, USA; State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin 150069, China
| | - W Miguel Disbennett
- Division of Rheumatology, Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Cathrine A Miner
- Division of Rheumatology, Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Joe A Holley
- Division of Rheumatology, Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Teresa V Naismith
- Department of Cell Biology and Physiology, Washington University School of Medicine, St Louis, MO 63110, USA
| | - W Alexander Stinson
- Department of Medicine, Washington University School of Medicine, St Louis, MO 63110, USA
| | - Xiaochao Wei
- Department of Medicine, Washington University School of Medicine, St Louis, MO 63110, USA
| | - Yue Ning
- Division of Rheumatology, Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Jiayuan Fu
- Division of Rheumatology, Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Trini A Ochoa
- Division of Rheumatology, Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Nehalee Surve
- Division of Rheumatology, Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Shivam A Zaver
- Division of Rheumatology, Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Kimberly A Wodzanowski
- Department of Microbiology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Katherine R Balka
- Department of Biochemistry and Molecular Biology, Immunity Program, Biomedicine Discovery Institute, Monash University, Clayton, VIC 3800, Australia
| | - Rajan Venkatraman
- Department of Biochemistry and Molecular Biology, Immunity Program, Biomedicine Discovery Institute, Monash University, Clayton, VIC 3800, Australia
| | - Canyu Liu
- Division of Rheumatology, Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Kelly Rome
- Department of Pathology, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Will Bailis
- Department of Pathology, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Yoko Shiba
- Faculty of Science and Engineering, Iwate University, Morioka 020-8551, Japan
| | - Sara Cherry
- Institute for Immunology and Immune Health, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA; Department of Pathology and Laboratory Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Sunny Shin
- Department of Microbiology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Clay F Semenkovich
- Department of Medicine, Washington University School of Medicine, St Louis, MO 63110, USA
| | - Dominic De Nardo
- Department of Biochemistry and Molecular Biology, Immunity Program, Biomedicine Discovery Institute, Monash University, Clayton, VIC 3800, Australia
| | - Sunnie Yoh
- Immunity and Pathogenesis Program, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA 92037, USA
| | - Elisha D O Roberson
- Department of Medicine, Washington University School of Medicine, St Louis, MO 63110, USA; Department of Genetics, Washington University School of Medicine, St Louis, MO 63110, USA
| | - Sumit K Chanda
- Immunity and Pathogenesis Program, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA 92037, USA
| | - David J Kast
- Department of Cell Biology and Physiology, Washington University School of Medicine, St Louis, MO 63110, USA.
| | - Jonathan J Miner
- Division of Rheumatology, Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA; Department of Medicine, Washington University School of Medicine, St Louis, MO 63110, USA; Department of Microbiology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA; Institute for Immunology and Immune Health, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA; Colton Center for Autoimmunity, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA.
| |
Collapse
|
81
|
Pan P, Cao S, Gao H, Qu X, Ma Y, Yang J, Pei X, Yang Y. Immp2l gene knockout induces granulosa cell senescence by activation of cGAS-STING pathway via TFAM-mediated mtDNA leakage. Int J Biol Macromol 2025; 307:142368. [PMID: 40120895 DOI: 10.1016/j.ijbiomac.2025.142368] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2024] [Revised: 02/18/2025] [Accepted: 03/19/2025] [Indexed: 03/25/2025]
Abstract
Granulosa cell-produced inflammatory factors may be key contributors to ovarian dysfunction, and Immp2l deficiency accelerates ovarian aging via granulosa cell senescence; however, the role of inflammation in granulosa cell senescence is largely unknown. Therefore, in this study, cGAS-STING-mediated inflammation was explored in Immp2l deficiency-induced granulosa cell senescence. Immp2l deficiency led to senescence-associated secretory phenotype (SASP) and granulosa cell senescence. Immp2l knockout caused mitochondrial dysfunction and mitochondrial DNA (mtDNA) leakage into the cytoplasm. The cytoplasmic mtDNA was recognized by the DNA-sensing molecule cGAS-STING, which activates cGAS-STING and key downstream interferon-stimulated genes (ISGs) and then promotes the secretion of proinflammatory factors, leading to SASP in senescent granulosa cells. Interestingly, the mitochondrial inner membrane pore protein (Cyclophilin D40) CyPD40 and the outer membrane pore protein voltage-dependent-anion channel 1 (VDAC1) were markedly increased in senescent granulosa cells, accompanied by significantly increased expression of the mtDNA stability protein mitochondrial transcription factor A (TFAM). Downregulation of TFAM with siRNA in senescent granulosa cells improved mitochondrial function, significantly decreased mtDNA in the cytoplasm, inhibited the cGAS-STING pathway and markedly decreased CyPD40 and VDAC1 protein levels in TFAM-treated senescent granulosa cells. The SASP phenotype was also alleviated. In addition, senescent granulosa cells were treated with procyanidin B2 (PCB2), which has anti-inflammatory effects, and the TFAM-mediated mtDNA-cGAS-STING pathway was inhibited, accompanied by a markedly reduced SASP phenotype and granulosa cell senescence. In conclusion, Immp2l gene knockout induced granulosa cell senescence by activation of the cGAS-STING pathway via TFAM-mediated mtDNA leakage into the cytoplasm through the CyPD40 and the VDAC1.
Collapse
Affiliation(s)
- Pengge Pan
- Key Laboratory of Fertility Preservation and Maintenance of Ministry of Education, School of Basic Medical Sciences, Ningxia Medical University, Yinchuan 750004, China
| | - Sinan Cao
- Key Laboratory of Fertility Preservation and Maintenance of Ministry of Education, School of Basic Medical Sciences, Ningxia Medical University, Yinchuan 750004, China
| | - Hui Gao
- Key Laboratory of Fertility Preservation and Maintenance of Ministry of Education, School of Basic Medical Sciences, Ningxia Medical University, Yinchuan 750004, China
| | - Xiaoya Qu
- Key Laboratory of Fertility Preservation and Maintenance of Ministry of Education, School of Basic Medical Sciences, Ningxia Medical University, Yinchuan 750004, China
| | - Yan Ma
- Key Laboratory of Fertility Preservation and Maintenance of Ministry of Education, School of Basic Medical Sciences, Ningxia Medical University, Yinchuan 750004, China
| | - Jinyi Yang
- Key Laboratory of Fertility Preservation and Maintenance of Ministry of Education, School of Basic Medical Sciences, Ningxia Medical University, Yinchuan 750004, China
| | - Xiuying Pei
- Key Laboratory of Fertility Preservation and Maintenance of Ministry of Education, School of Basic Medical Sciences, Ningxia Medical University, Yinchuan 750004, China.
| | - Yanzhou Yang
- Key Laboratory of Fertility Preservation and Maintenance of Ministry of Education, School of Basic Medical Sciences, Ningxia Medical University, Yinchuan 750004, China; Emergency Department, The First People's Hospital of Yinchuan, The Second Clinical Medical College, Ningxia Medical University, Yinchuan, Ningxia, China.
| |
Collapse
|
82
|
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] [MESH Headings] [Grants] [Track Full Text] [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
|
83
|
Yonesaka K, Kurosaki T, Tanizaki J, Kawakami H, Tanaka K, Maenishi O, Takamura S, Sakai K, Chiba Y, Teramura T, Goto H, Otsuka E, Okida H, Funabashi M, Hashimoto Y, Hirotani K, Kamai Y, Kagari T, Nishio K, Kakimi K, Hayashi H. Chromosomal Instability Is Associated with cGAS-STING Activation in EGFR-TKI Refractory Non-Small-Cell Lung Cancer. Cells 2025; 14:447. [PMID: 40136696 PMCID: PMC11941500 DOI: 10.3390/cells14060447] [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: 02/11/2025] [Revised: 03/13/2025] [Accepted: 03/14/2025] [Indexed: 03/27/2025] Open
Abstract
Epidermal growth factor receptor tyrosine kinase inhibitors (EGFR-TKIs) are standard therapies for EGFR-mutated non-small-cell lung cancer (NSCLC); however, their efficacy is inconsistent. Secondary mutations in the EGFR or other genes that lead to resistance have been identified, but resistance mechanisms have not been fully identified. Chromosomal instability (CIN) is a hallmark of cancer and results in genetic diversity. In this study, we demonstrated by transcriptomic analysis that CIN activates the cGAS-STING signaling pathway, which leads to EGFR-TKI refractoriness in a subset of EGFR-mutated NSCLC patients. Furthermore, EGFR-mutated H1975dnMCAK cells, which frequently underwent chromosomal mis-segregation, demonstrated refractoriness to the EGFR-TKI osimertinib compared to control cells. Second, H1975dnMCAK cells exhibited activation of cGAS-STING signaling and its downstream signaling, including tumor-promoting cytokine IL-6. Finally, chromosomally unstable EGFR-mutated NSCLC exhibited enhanced epithelial-mesenchymal transition (EMT). Blockade of cGAS-STING-TBK1 signaling reversed EMT, resulting in restored susceptibility to EGFR-TKIs in vitro and in vivo. These results suggest that CIN may lead to the activation of cGAS-STING signaling in some EGFR-mutated NSCLC, resulting in EMT-associated EGFR-TKI resistance.
Collapse
Affiliation(s)
- Kimio Yonesaka
- Department of Medical Oncology, Kindai University Faculty of Medicine, Osaka 589-8511, Japan
| | - Takashi Kurosaki
- Department of Medical Oncology, Kindai University Faculty of Medicine, Osaka 589-8511, Japan
| | - Junko Tanizaki
- Department of Medical Oncology, Kindai University Faculty of Medicine, Osaka 589-8511, Japan
- Department of Medical Oncology, Kishiwada City Hospital, Osaka 589-8511, Japan
| | - Hisato Kawakami
- Department of Medical Oncology, Kindai University Faculty of Medicine, Osaka 589-8511, Japan
| | - Kaoru Tanaka
- Department of Medical Oncology, Kindai University Faculty of Medicine, Osaka 589-8511, Japan
| | - Osamu Maenishi
- Department of Pathology, Kindai University Faculty of Medicine, Osaka 589-8511, Japan;
| | - Shiki Takamura
- Department of Immunology, Kindai University Faculty of Medicine, Osaka 589-8511, Japan (K.K.)
| | - Kazuko Sakai
- Department of Genome Biology, Kindai University Faculty of Medicine, Osaka 589-8511, Japan
| | - Yasutaka Chiba
- Clinical Research Center, Kindai University Hospital, Osaka 589-8511, Japan;
| | - Takeshi Teramura
- Division of Cell Biology for Regenerative Medicine, Institute of Advanced Clinical Medicine, Kindai University Faculty of Medicine, Osaka 589-8511, Japan
| | - Hiroki Goto
- Translational Research Department, Daiichi Sankyo RD Novare Co., Ltd., Tokyo 134-0081, Japan (M.F.)
| | - Eri Otsuka
- Translational Research Department, Daiichi Sankyo RD Novare Co., Ltd., Tokyo 134-0081, Japan (M.F.)
| | - Hiroaki Okida
- Translational Research Department, Daiichi Sankyo RD Novare Co., Ltd., Tokyo 134-0081, Japan (M.F.)
| | - Masanori Funabashi
- Translational Research Department, Daiichi Sankyo RD Novare Co., Ltd., Tokyo 134-0081, Japan (M.F.)
| | - Yuuri Hashimoto
- Discovery Intelligence Research Laboratories, Research Function, R&D Division, Daiichi Sankyo Co., Ltd., Tokyo 103-0023, Japan
| | - Kenji Hirotani
- Early Clinical Development Department, Development Function, R&D Division, Daiichi Sankyo Co., Ltd., Tokyo 103-0023, Japan
| | - Yasuki Kamai
- Discovery Research Laboratories I, Research Function, R&D Division, Daiichi Sankyo Co., Ltd., Tokyo 103-0023, Japan; (Y.K.)
| | - Takashi Kagari
- Discovery Research Laboratories I, Research Function, R&D Division, Daiichi Sankyo Co., Ltd., Tokyo 103-0023, Japan; (Y.K.)
| | - Kazuto Nishio
- Department of Genome Biology, Kindai University Faculty of Medicine, Osaka 589-8511, Japan
| | - Kazuhiro Kakimi
- Department of Immunology, Kindai University Faculty of Medicine, Osaka 589-8511, Japan (K.K.)
| | - Hidetoshi Hayashi
- Department of Medical Oncology, Kindai University Faculty of Medicine, Osaka 589-8511, Japan
| |
Collapse
|
84
|
Guo Y, Zhou H, Wang Y, Gu Y. Activated NETosis of bone marrow neutrophils up-regulates macrophage osteoclastogenesis via cGAS-STING/AKT2 pathway to promote osteoporosis. Exp Cell Res 2025; 446:114477. [PMID: 39988126 DOI: 10.1016/j.yexcr.2025.114477] [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: 12/02/2024] [Revised: 02/05/2025] [Accepted: 02/16/2025] [Indexed: 02/25/2025]
Abstract
Bone marrow (BM) of postmenopausal osteoporosis has been found highly inflammatory, resulting from dysregulated immune cells induced by both estrogen efficiency and body aging. NETosis of neutrophils has been found aberrantly activated in age-related chronic inflammation, while their role in postmenopausal osteoporosis remains unclear. Here we found NETosis of BM neutrophils of OVX (ovariectomy) mice was significantly activated, and we verified NETs released by neutrophils induced M1 polarization and osteoclastogenesis of RAW264.7 macrophages. Further, we demonstrated effects of NETs on osteoclastogenesis was mediated by cGAS-STING/AKT2 pathway. Finally, we found in vivo NETs-clearance through GSK484 significantly inhibited osteoclastogenesis and attenuated osteoporosis of OVX mice. Our study highlights the role of neutrophil NETosis in activating osteoclastogenesis and bone resorption of postmenopausal osteoporosis, thereby providing novel targets for bone loss treatment.
Collapse
Affiliation(s)
- Yutong Guo
- Department of Orthodontics, Peking University School and Hospital of Stomatology & National Center for Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices& Beijing Key Laboratory of Digital Stomatology & NHC Key Laboratory of Digital Stomatology & NMPA Key Laboratory for Dental Materials, No.22, Zhongguancun South Avenue, Haidian District, Beijing, 100081, PR China
| | - Hanzhang Zhou
- Department of Orthodontics, Peking University School and Hospital of Stomatology & National Center for Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices& Beijing Key Laboratory of Digital Stomatology & NHC Key Laboratory of Digital Stomatology & NMPA Key Laboratory for Dental Materials, No.22, Zhongguancun South Avenue, Haidian District, Beijing, 100081, PR China
| | - Yixiang Wang
- Central Laboratory, Peking University School and Hospital of Stomatology, No. 22 Zhongguancun Avenue South, Haidian District, Beijing, 100081, PR China.
| | - Yan Gu
- Department of Orthodontics, Peking University School and Hospital of Stomatology & National Center for Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices& Beijing Key Laboratory of Digital Stomatology & NHC Key Laboratory of Digital Stomatology & NMPA Key Laboratory for Dental Materials, No.22, Zhongguancun South Avenue, Haidian District, Beijing, 100081, PR China.
| |
Collapse
|
85
|
Chen C, Cai X, Liu Z, Zhang W, Yang J, Tang Y, Chen Y, Huang Y, Hu W, Zhang X, Zhou J, Wu Y, Yin W, Shang R, Lu Q, Sheng H, Ju Z, Luo G, He W. STING coordinates resolution of inflammation during wound repair by modulating macrophage trafficking through STAT3. J Leukoc Biol 2025; 117:qiae175. [PMID: 39119796 DOI: 10.1093/jleuko/qiae175] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Revised: 06/07/2024] [Accepted: 06/24/2024] [Indexed: 08/10/2024] Open
Abstract
Efficient cutaneous wound healing requires a coordinated transition between inflammatory phases mediated by dynamic changes in leukocyte subset populations. Here, we identify STING as a key innate immune mediator governing timely resolution of inflammation by regulating macrophage dynamics during skin repair. Using a mouse model, we show STING deficiency caused delayed wound closure associated with abnormal persistence of TNF-α+ leukocytes. This resulted from the impaired macrophage recruitment. STING controlled the trafficking of bone marrow myeloid cells into blood and wounds, intrinsically enhancing macrophage migratory capacity through STAT3 activation. Specifically, STING modulated the production of monocyte chemokines and their receptors CCR2/CCR5 to enable efficient egress and wound infiltration. Consequently, disrupted systemic and local STING-STAT3-chemokine signaling combine to delay macrophage influx. This study elucidates STING as a critical rheostat tuning macrophage responses through STAT3 to orchestrate inflammatory resolution necessary for efficient wound healing. Our findings have broad implications for targeting STING therapeutically in both regenerative medicine and inflammatory disease contexts.
Collapse
Affiliation(s)
- Cheng Chen
- State Key Laboratory of Trauma, Burn and Combined Injury, Institute of Burn Research, Southwest Hospital, Third Military Medical University (Army Medical University), 30 Gaotanyan Main Street, Shapingba District, Chongqing 400038, China
- Chongqing Key Laboratory for Disease Proteomics, 30 Gaotanyan Main Street, Shapingba District, Chongqing 400038, China
| | - Xin Cai
- State Key Laboratory of Trauma, Burn and Combined Injury, Institute of Burn Research, Southwest Hospital, Third Military Medical University (Army Medical University), 30 Gaotanyan Main Street, Shapingba District, Chongqing 400038, China
- Chongqing Key Laboratory for Disease Proteomics, 30 Gaotanyan Main Street, Shapingba District, Chongqing 400038, China
| | - Zhihui Liu
- State Key Laboratory of Trauma, Burn and Combined Injury, Institute of Burn Research, Southwest Hospital, Third Military Medical University (Army Medical University), 30 Gaotanyan Main Street, Shapingba District, Chongqing 400038, China
- Chongqing Key Laboratory for Disease Proteomics, 30 Gaotanyan Main Street, Shapingba District, Chongqing 400038, China
| | - Weiguang Zhang
- Department of Intensive Care, Southwest Hospital, Army Medical University, 30 Gaotanyan Main Street, Shapingba District, Chongqing 400038, China
| | - Jiacai Yang
- State Key Laboratory of Trauma, Burn and Combined Injury, Institute of Burn Research, Southwest Hospital, Third Military Medical University (Army Medical University), 30 Gaotanyan Main Street, Shapingba District, Chongqing 400038, China
- Chongqing Key Laboratory for Disease Proteomics, 30 Gaotanyan Main Street, Shapingba District, Chongqing 400038, China
| | - Yuanyang Tang
- Department of Infectious Diseases, Southwest Hospital, Third Military Medical University (Army Medical University), 30 Gaotanyan Main Street, Shapingba District, Chongqing 400038, China
| | - Yunxia Chen
- State Key Laboratory of Trauma, Burn and Combined Injury, Institute of Burn Research, Southwest Hospital, Third Military Medical University (Army Medical University), 30 Gaotanyan Main Street, Shapingba District, Chongqing 400038, China
- Chongqing Key Laboratory for Disease Proteomics, 30 Gaotanyan Main Street, Shapingba District, Chongqing 400038, China
| | - Yong Huang
- State Key Laboratory of Trauma, Burn and Combined Injury, Institute of Burn Research, Southwest Hospital, Third Military Medical University (Army Medical University), 30 Gaotanyan Main Street, Shapingba District, Chongqing 400038, China
- Chongqing Key Laboratory for Disease Proteomics, 30 Gaotanyan Main Street, Shapingba District, Chongqing 400038, China
| | - Wengang Hu
- State Key Laboratory of Trauma, Burn and Combined Injury, Institute of Burn Research, Southwest Hospital, Third Military Medical University (Army Medical University), 30 Gaotanyan Main Street, Shapingba District, Chongqing 400038, China
- Chongqing Key Laboratory for Disease Proteomics, 30 Gaotanyan Main Street, Shapingba District, Chongqing 400038, China
| | - Xiaorong Zhang
- State Key Laboratory of Trauma, Burn and Combined Injury, Institute of Burn Research, Southwest Hospital, Third Military Medical University (Army Medical University), 30 Gaotanyan Main Street, Shapingba District, Chongqing 400038, China
- Chongqing Key Laboratory for Disease Proteomics, 30 Gaotanyan Main Street, Shapingba District, Chongqing 400038, China
| | - Junyi Zhou
- State Key Laboratory of Trauma, Burn and Combined Injury, Institute of Burn Research, Southwest Hospital, Third Military Medical University (Army Medical University), 30 Gaotanyan Main Street, Shapingba District, Chongqing 400038, China
- Chongqing Key Laboratory for Disease Proteomics, 30 Gaotanyan Main Street, Shapingba District, Chongqing 400038, China
| | - Yanjun Wu
- State Key Laboratory of Trauma, Burn and Combined Injury, Institute of Burn Research, Southwest Hospital, Third Military Medical University (Army Medical University), 30 Gaotanyan Main Street, Shapingba District, Chongqing 400038, China
- Chongqing Key Laboratory for Disease Proteomics, 30 Gaotanyan Main Street, Shapingba District, Chongqing 400038, China
| | - Wenjing Yin
- State Key Laboratory of Trauma, Burn and Combined Injury, Institute of Burn Research, Southwest Hospital, Third Military Medical University (Army Medical University), 30 Gaotanyan Main Street, Shapingba District, Chongqing 400038, China
- Chongqing Key Laboratory for Disease Proteomics, 30 Gaotanyan Main Street, Shapingba District, Chongqing 400038, China
- Academy of Biological Engineering, Chongqing University, 174 Shazheng Street, Chongqing 400044, China
| | - Ruoyu Shang
- State Key Laboratory of Trauma, Burn and Combined Injury, Institute of Burn Research, Southwest Hospital, Third Military Medical University (Army Medical University), 30 Gaotanyan Main Street, Shapingba District, Chongqing 400038, China
- Chongqing Key Laboratory for Disease Proteomics, 30 Gaotanyan Main Street, Shapingba District, Chongqing 400038, China
| | - Qudong Lu
- Department of Urology, Army 73rd Group Military Hospital, 94 Wenyuan Road, Xiamen 361012, China
| | - Hao Sheng
- Urology Department, The Second Affiliated Hospital, Third Military Medical University (Army Medical University), NO 83 Xinqiao Main Street, Chongqing 400037, China
| | - Zhenyu Ju
- Key Laboratory of Regenerative Medicine of Ministry of Education, Guangzhou Regenerative Medicine and Health Guangdong Laboratory, Institute of Aging and Regenerative Medicine, Jinan University, No. 601, West Huangpu Avenue, Guangzhou 510632, China
| | - Gaoxing Luo
- State Key Laboratory of Trauma, Burn and Combined Injury, Institute of Burn Research, Southwest Hospital, Third Military Medical University (Army Medical University), 30 Gaotanyan Main Street, Shapingba District, Chongqing 400038, China
- Chongqing Key Laboratory for Disease Proteomics, 30 Gaotanyan Main Street, Shapingba District, Chongqing 400038, China
| | - Weifeng He
- State Key Laboratory of Trauma, Burn and Combined Injury, Institute of Burn Research, Southwest Hospital, Third Military Medical University (Army Medical University), 30 Gaotanyan Main Street, Shapingba District, Chongqing 400038, China
- Chongqing Key Laboratory for Disease Proteomics, 30 Gaotanyan Main Street, Shapingba District, Chongqing 400038, China
| |
Collapse
|
86
|
He P, Wen C, Zhang X, Yin H. Discovery of a Novel CRBN-Recruiting cGAS PROTAC Degrader for the Treatment of Ulcerative Colitis. J Med Chem 2025; 68:5551-5572. [PMID: 40012371 DOI: 10.1021/acs.jmedchem.4c02774] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/28/2025]
Abstract
Cyclic GMP-AMP synthase (cGAS), a critical cytosolic DNA sensor initiating innate immune responses in the presence of cytosolic DNA, is increasingly recognized as a promising therapeutic target for ulcerative colitis (UC). Here, we reported the design, synthesis, structure-activity relationship exploration and biological evaluation of a novel class of CRBN-recruiting cGAS-targeting PROTAC degraders. Among them, TH35 exhibited the most favorable degradation profile, achieving potent and selective degradation of cGAS, and markedly attenuated dsDNA-induced activation of cGAS signaling in both human and murine cells, with minimal cytotoxic effects. In vivo, TH35 demonstrated superior therapeutic efficacy in a dextran sulfate sodium (DSS)-induced mouse model of UC compared to the corresponding cGAS inhibitor, while also displaying acceptable pharmacokinetic properties. Collectively, TH35 as the first CRBN-recruiting cGAS PROTAC holds promise for augmenting anti-inflammatory responses and offers a new avenue for treating cGAS-driven inflammatory diseases.
Collapse
Affiliation(s)
- Peng He
- State Key Laboratory of Membrane Biology, School of Pharmaceutical Sciences, Tsinghua-Peking Center for Life Sciences, Key Laboratory of Bioorganic Phosphorous Chemistry and Chemical Biology (Ministry of Education), Tsinghua University, Beijing 100084, China
| | - Chengming Wen
- State Key Laboratory of Membrane Biology, School of Pharmaceutical Sciences, Tsinghua-Peking Center for Life Sciences, Key Laboratory of Bioorganic Phosphorous Chemistry and Chemical Biology (Ministry of Education), Tsinghua University, Beijing 100084, China
| | - Xinyu Zhang
- State Key Laboratory of Membrane Biology, School of Pharmaceutical Sciences, Tsinghua-Peking Center for Life Sciences, Key Laboratory of Bioorganic Phosphorous Chemistry and Chemical Biology (Ministry of Education), Tsinghua University, Beijing 100084, China
| | - Hang Yin
- State Key Laboratory of Membrane Biology, School of Pharmaceutical Sciences, Tsinghua-Peking Center for Life Sciences, Key Laboratory of Bioorganic Phosphorous Chemistry and Chemical Biology (Ministry of Education), Tsinghua University, Beijing 100084, China
| |
Collapse
|
87
|
Niu GH, Hsiao WC, Lee PH, Zheng LG, Yang YS, Huang WC, Hsieh CC, Chiu TY, Wang JY, Chen CP, Huang CL, You MS, Kuo YP, Wang CM, Wen ZH, Yu GY, Chen CT, Chi YH, Tung CW, Hsu SC, Yeh TK, Sung PJ, Zhang MM, Tsou LK. Orally Bioavailable and Site-Selective Covalent STING Inhibitor Derived from a Macrocyclic Marine Diterpenoid. J Med Chem 2025; 68:5471-5487. [PMID: 40014799 PMCID: PMC11912488 DOI: 10.1021/acs.jmedchem.4c02665] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Revised: 02/16/2025] [Accepted: 02/20/2025] [Indexed: 03/01/2025]
Abstract
Pharmacological inhibition of the cGAS-STING-controlled innate immune pathway is an emerging therapeutic strategy for a myriad of inflammatory diseases. Here, we report GHN105 as an orally bioavailable covalent STING inhibitor. Late-stage diversification of the briarane-type diterpenoid excavatolide B allowed the installation of solubility-enhancing functional groups while enhancing its activity as a covalent STING inhibitor against multiple human STING variants, including the S154 variant responsible for a genetic autoimmune disease. Selectively engaging the membrane-proximal Cys91 residue of STING, GHN105 dose-dependently inhibited cGAS-STING signaling and type I interferon responses in cells and in vivo. Moreover, orally administered GHN105 exhibited on-target engagement in vivo and markedly reversed key pathological features in a delayed treatment of the acute colitis mouse model. Our study provided proof of concept that the synthetic briarane analog GHN105 serves as a safe, site-selective, and orally active covalent STING inhibitor and devises a regimen that allows long-term systemic administration.
Collapse
Affiliation(s)
- Guang-Hao Niu
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Zhunan, Miaoli 35053, Taiwan
| | - Wan-Chi Hsiao
- Institute of Molecular and Genomic Medicine, National Health Research Institutes, Zhunan, Miaoli 35053, Taiwan
- Institute of Biotechnology, National Tsing Hua University, Hsinchu 30013, Taiwan
| | - Po-Hsun Lee
- Institute of Molecular and Genomic Medicine, National Health Research Institutes, Zhunan, Miaoli 35053, Taiwan
- Institute of Biotechnology, National Tsing Hua University, Hsinchu 30013, Taiwan
| | - Li-Guo Zheng
- National Museum of Marine Biology and Aquarium, Pingtung 944401, Taiwan
| | - Yu-Shao Yang
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Zhunan, Miaoli 35053, Taiwan
| | - Wei-Cheng Huang
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Zhunan, Miaoli 35053, Taiwan
| | - Chih-Chien Hsieh
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Zhunan, Miaoli 35053, Taiwan
| | - Tai-Yu Chiu
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Zhunan, Miaoli 35053, Taiwan
| | - Jing-Ya Wang
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Zhunan, Miaoli 35053, Taiwan
| | - Ching-Ping Chen
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Zhunan, Miaoli 35053, Taiwan
| | - Chen-Lung Huang
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Zhunan, Miaoli 35053, Taiwan
| | - May-Su You
- Institute of Molecular and Genomic Medicine, National Health Research Institutes, Zhunan, Miaoli 35053, Taiwan
| | - Yi-Ping Kuo
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Zhunan, Miaoli 35053, Taiwan
| | - Chien-Ming Wang
- Institute of Molecular and Genomic Medicine, National Health Research Institutes, Zhunan, Miaoli 35053, Taiwan
| | - Zhi-Hong Wen
- Department of Marine Biotechnology and Resources, National Sun Yat-Sen University, Kaohsiung 804201, Taiwan
| | - Guann-Yi Yu
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Zhunan, Miaoli 35053, Taiwan
| | - Chiung-Tong Chen
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Zhunan, Miaoli 35053, Taiwan
| | - Ya-Hui Chi
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Zhunan, Miaoli 35053, Taiwan
| | - Chun-Wei Tung
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Zhunan, Miaoli 35053, Taiwan
| | - Shu-Ching Hsu
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Zhunan, Miaoli 35053, Taiwan
| | - Teng-Kuang Yeh
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Zhunan, Miaoli 35053, Taiwan
| | - Ping-Jyun Sung
- National Museum of Marine Biology and Aquarium, Pingtung 944401, Taiwan
| | - Mingzi M Zhang
- Institute of Molecular and Genomic Medicine, National Health Research Institutes, Zhunan, Miaoli 35053, Taiwan
| | - Lun Kelvin Tsou
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Zhunan, Miaoli 35053, Taiwan
| |
Collapse
|
88
|
Gong G, Shen S, Shen S, Wang R, Zheng T, Xu W, Wu J. YAP/TAZ-mediated nuclear membrane rupture in promoting senescence of skeletal muscle associated with COPD. Respir Res 2025; 26:98. [PMID: 40075503 PMCID: PMC11905641 DOI: 10.1186/s12931-025-03170-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2024] [Accepted: 02/25/2025] [Indexed: 03/14/2025] Open
Abstract
Patients with chronic obstructive pulmonary disease (COPD) often develop complications associated with sarcopenia; however, the underlying mechanisms remain unclear. Through a combination of in vitro and in vivo experiments, as well as bioinformatics analysis, our study identified YAP/TAZ as a key regulator of the aging phenotype in the skeletal muscle of COPD patients. In skeletal muscle affected by cigarette smoke-induced COPD, we observed significant reductions in YAP/TAZ levels, alongside markers indicative of skeletal muscle aging and dysfunction. Notably, overexpression of YAP/TAZ significantly improved these conditions. Our results suggest a novel mechanism whereby the maintenance of YAP/TAZ activity interacts with ACTR2 to preserve nuclear membrane integrity and reduce cytoplasmic dsDNA levels, thereby attenuating STING activation and cellular senescence. Additionally, we found that YAP is involved in the transcriptional regulation of the ACTR2 promoter region. Overall, preserving YAP/TAZ activity may help prevent skeletal muscle aging associated with COPD, representing a new strategy for intervening in COPD-related sarcopenia.
Collapse
Affiliation(s)
- Ge Gong
- Key Laboratory of Geriatrics of Jiangsu Province, Department of Geriatrics, The First Affiliated Hospital with Nanjing Medical University, Nanjing, China
- Department of Geriatrics, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Shuping Shen
- Key Laboratory of Geriatrics of Jiangsu Province, Department of Geriatrics, The First Affiliated Hospital with Nanjing Medical University, Nanjing, China
| | - Shaoran Shen
- Key Laboratory of Geriatrics of Jiangsu Province, Department of Geriatrics, The First Affiliated Hospital with Nanjing Medical University, Nanjing, China
| | - Ran Wang
- Key Laboratory of Geriatrics of Jiangsu Province, Department of Geriatrics, The First Affiliated Hospital with Nanjing Medical University, Nanjing, China
| | - Tianping Zheng
- Key Laboratory of Geriatrics of Jiangsu Province, Department of Geriatrics, The First Affiliated Hospital with Nanjing Medical University, Nanjing, China
| | - Wei Xu
- Key Laboratory of Geriatrics of Jiangsu Province, Department of Geriatrics, The First Affiliated Hospital with Nanjing Medical University, Nanjing, China.
| | - Jianqing Wu
- Key Laboratory of Geriatrics of Jiangsu Province, Department of Geriatrics, The First Affiliated Hospital with Nanjing Medical University, Nanjing, China.
| |
Collapse
|
89
|
Chen Y, Yue S, Yu L, Cao J, Liu Y, Deng A, Lu Y, Yang J, Li H, Du J, Xia J, Li Y, Xia Y. Regulation and Function of the cGAS-STING Pathway: Mechanisms, Post-Translational Modifications, and Therapeutic Potential in Immunotherapy. Drug Des Devel Ther 2025; 19:1721-1739. [PMID: 40098909 PMCID: PMC11911240 DOI: 10.2147/dddt.s501773] [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: 10/22/2024] [Accepted: 02/24/2025] [Indexed: 03/19/2025] Open
Abstract
Autoimmune diseases arise when the immune system attacks healthy tissues, losing tolerance for self-tissues. Normally, the immune system recognizes and defends against pathogens like bacteria and viruses. The cGAS-STING pathway, activated by pattern-recognition receptors (PRRs), plays a key role in autoimmune responses. The cGAS protein senses pathogenic DNA and synthesizes cGAMP, which induces conformational changes in STING, activating kinases IKK and TBK1 and leading to the expression of interferon genes or inflammatory mediators. This pathway is crucial in immunotherapy, activating innate immunity, enhancing antigen presentation, modulating the tumor microenvironment, and integrating into therapeutic strategies. Modulation strategies include small molecule inhibitors, oligonucleotide therapies, protein and antibody therapies, genetic and epigenetic regulation, cytokine and metabolite modulation, and nanoscale delivery systems. Post-translational modifications (PTMs) of the cGAS-STING pathway, such as phosphorylation, acetylation, ubiquitination, methylation, palmitoylation, and glycosylation, fine-tune immune responses by regulating protein activity, stability, localization, and interactions. These modifications are interconnected and collectively influence pathway functionality. We summarize the functions of cGAS-STING and its PTMs in immune and non-immune cells across various diseases, and explore potential clinical applications.
Collapse
Affiliation(s)
- Yuhan Chen
- Laboratory Medicine Center, Department of Clinical Laboratory, Zhejiang Provincial People’s Hospital (Affiliated People’s Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, People’s Republic of China
| | - Si Yue
- Laboratory Medicine Center, Department of Clinical Laboratory, Zhejiang Provincial People’s Hospital (Affiliated People’s Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, People’s Republic of China
| | - Lingyan Yu
- Laboratory Medicine Center, Department of Clinical Laboratory, Zhejiang Provincial People’s Hospital (Affiliated People’s Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, People’s Republic of China
| | - Jinghao Cao
- Laboratory Medicine Center, Department of Clinical Laboratory, Zhejiang Provincial People’s Hospital (Affiliated People’s Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, People’s Republic of China
| | - Yingchao Liu
- Laboratory Medicine Center, Department of Clinical Laboratory, Zhejiang Provincial People’s Hospital (Affiliated People’s Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, People’s Republic of China
| | - Aoli Deng
- Laboratory Medicine Center, Department of Clinical Laboratory, Zhejiang Provincial People’s Hospital (Affiliated People’s Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, People’s Republic of China
| | - Yajuan Lu
- Laboratory Medicine Center, Department of Clinical Laboratory, Zhejiang Provincial People’s Hospital (Affiliated People’s Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, People’s Republic of China
| | - Jing Yang
- Laboratory Medicine Center, Department of Clinical Laboratory, Zhejiang Provincial People’s Hospital (Affiliated People’s Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, People’s Republic of China
| | - Huanjuan Li
- Laboratory Medicine Center, Department of Clinical Laboratory, Zhejiang Provincial People’s Hospital (Affiliated People’s Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, People’s Republic of China
| | - Jing Du
- Laboratory Medicine Center, Department of Clinical Laboratory, Zhejiang Provincial People’s Hospital (Affiliated People’s Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, People’s Republic of China
| | - Jun Xia
- Laboratory Medicine Center, Department of Clinical Laboratory, Zhejiang Provincial People’s Hospital (Affiliated People’s Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, People’s Republic of China
| | - Yanchun Li
- Department of Clinical Laboratory, Affiliated Hangzhou First People’s Hospital, School of Medicine, Westlake University, Hangzhou, Zhejiang, People’s Republic of China
| | - Yongming Xia
- Department of Hematology, Yuyao People’s Hospital, Yuyao, Zhejiang, People’s Republic of China
| |
Collapse
|
90
|
Xu X, Wang X, Liao YP, Luo L, Nel AE. Reprogramming the Tolerogenic Immune Response Against Pancreatic Cancer Metastases by Lipid Nanoparticles Delivering a STING Agonist Plus Mutant KRAS mRNA. ACS NANO 2025; 19:8579-8594. [PMID: 40025875 PMCID: PMC11912578 DOI: 10.1021/acsnano.4c14102] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Revised: 02/14/2025] [Accepted: 02/18/2025] [Indexed: 03/04/2025]
Abstract
We demonstrate reprogramming of the tolerogenic immune environment in the liver for mounting an effective immune response against often-fatal pancreatic cancer metastases. This was achieved by engineering a lipid nanoparticle (LNP) to deliver mRNA encoding the KRAS G12D neoantigenic epitope along with cGAMP, a dinucleotide agonist of the stimulator of the interferon genes (STING) pathway, capable of activating a type I interferon response. cGAMP/mKRAS/LNP were synthesized by a microfluidics approach involving nanoprecipitation of mRNA and cGAMP by an ionizable lipid, MC3. Controls included nanoparticles delivering individual components or a wild-type RAS sequence. The dual delivery carrier successfully activated the type I interferon pathway in vitro as well as in vivo, with reprogramming of costimulatory receptor (CD80 and CD86) and MHC-I expression on liver antigen-presenting cells (APC). This allowed the generation of IFN-γ producing cytotoxic T cells, capable of mounting an effective immune response in the metastatic KRAS pancreatic cancer (KPC) mouse model. Noteworthy, intravenous injection of cGAMP/mKRAS/LNP suppressed metastatic growth significantly and prolonged animal survival, both prophylactically and during treatment of established metastases. The protective immune response was mediated by the generation of perforin-releasing CD8+ cytotoxic T cells, engaged in pancreatic cancer cell killing. Importantly, the immune response could also be adoptively transferred by injecting splenocytes (containing memory T cells) from treated into nontreated recipient mice. This study demonstrates that reprogramming the immune-protective niche for metastatic pancreatic cancer can be achieved by the delivery of a STING agonist and mutant KRAS mRNA via ionizable LNPs, offering both prophylactic and therapeutic advantages.
Collapse
Affiliation(s)
- Xiao Xu
- Division
of NanoMedicine, Department of Medicine, University of California, Los Angeles, California 90095, United States
- California
NanoSystems Institute, University of California, Los Angeles, California 90095, United States
| | - Xiang Wang
- Division
of NanoMedicine, Department of Medicine, University of California, Los Angeles, California 90095, United States
- California
NanoSystems Institute, University of California, Los Angeles, California 90095, United States
| | - Yu-Pei Liao
- Division
of NanoMedicine, Department of Medicine, University of California, Los Angeles, California 90095, United States
| | - Lijia Luo
- Division
of NanoMedicine, Department of Medicine, University of California, Los Angeles, California 90095, United States
- California
NanoSystems Institute, University of California, Los Angeles, California 90095, United States
| | - Andre E. Nel
- Division
of NanoMedicine, Department of Medicine, University of California, Los Angeles, California 90095, United States
- California
NanoSystems Institute, University of California, Los Angeles, California 90095, United States
| |
Collapse
|
91
|
Deng J, Wang Z, Wu L, Song Z, Bahlol HS, Li X, Zhao L, Han H. Metal-Phenolic Network Hydrogel Vaccine Platform for Enhanced Humoral Immunity against Lethal Rabies Virus. ACS NANO 2025; 19:9042-9052. [PMID: 40025824 DOI: 10.1021/acsnano.4c17759] [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: 03/04/2025]
Abstract
Rabies, caused by rabies virus (RABV), is a zoonotic disease with a high mortality rate that has attracted global attention with the goal of eradication by 2030. However, rabies can only be prevented by appropriate and multiple vaccinations, which impede widespread vaccination in developing countries due to its high expenditure. Designing single-dose vaccines is a pressing challenge in the prevention of rabies and other infectious diseases. Herein, a metal-phenolic network (MPN)-based hydrogel vaccine (designated as CGMR) was developed to stimulate potent humoral immunity against RABV infection by a single immunization, resulting in 4.3-fold and 1.8-fold enhancements of virus-neutralizing antibody compared with that induced by inactivated RABV and alum adjuvant. The CGMR, cross-linked by phenol-modified chitosan with manganese ion, could prolong residence time by confining the antigen to the network of hydrogel, acting as a "hydrogel antigen depot". It also stimulated the activation of the cyclic guanosine monophosphate-adenosine monophosphate synthase (cGAS)-stimulator of interferon gene (STING) pathway, facilitating dendritic cell maturation and antigen presentation. The vaccine formulation recruited immunocytes and activated the germinal center, enhancing and sustaining humoral immune responses against the virulent RABV challenge. Collectively, this injectable manganese-based hydrogel vaccine provides a universal and ideal avenue for rabies and other infectious diseases.
Collapse
Affiliation(s)
- Jiamin Deng
- The National Key Laboratory of Agricultural Microbiology, College of Life Science and Technology, College of Chemistry, Huazhong Agricultural University, Wuhan 430070, Hubei, China
| | - Zongmei Wang
- The National Key Laboratory of Agricultural Microbiology, The Hubei Hongshan Laboratory, The Key Laboratory of Preventive Veterinary Medicine of Hubei Province, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, Hubei, China
| | - Liqin Wu
- The National Key Laboratory of Agricultural Microbiology, The Hubei Hongshan Laboratory, The Key Laboratory of Preventive Veterinary Medicine of Hubei Province, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, Hubei, China
| | - Zhiyong Song
- The National Key Laboratory of Agricultural Microbiology, College of Science, Huazhong Agricultural University, Wuhan 430070, Hubei, China
| | - Hagar Shendy Bahlol
- The National Key Laboratory of Agricultural Microbiology, College of Life Science and Technology, College of Chemistry, Huazhong Agricultural University, Wuhan 430070, Hubei, China
- Department of Biochemistry, Faculty of Agriculture, Benha University, Moshtohor, Toukh 13736, Egypt
| | - Xun Li
- The National Key Laboratory of Agricultural Microbiology, College of Science, Huazhong Agricultural University, Wuhan 430070, Hubei, China
| | - Ling Zhao
- The National Key Laboratory of Agricultural Microbiology, The Hubei Hongshan Laboratory, The Key Laboratory of Preventive Veterinary Medicine of Hubei Province, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, Hubei, China
| | - Heyou Han
- The National Key Laboratory of Agricultural Microbiology, College of Life Science and Technology, College of Chemistry, Huazhong Agricultural University, Wuhan 430070, Hubei, China
| |
Collapse
|
92
|
Sun S, Qian S, Wang R, Zhao M, Li R, Gu W, Zhao M, Qian C, Liu L, Tang X, Li Y, Shi H, Pan Y, Xiao H, Yang K, Hu C, Huang Y, Wei L, Zhang Y, Ji J, Chen Y, Liu H. Targeting GOLPH3L improves glioblastoma radiotherapy by regulating STING-NLRP3-mediated tumor immune microenvironment reprogramming. Sci Transl Med 2025; 17:eado0020. [PMID: 40043140 DOI: 10.1126/scitranslmed.ado0020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Revised: 08/04/2024] [Accepted: 02/12/2025] [Indexed: 04/18/2025]
Abstract
Radiotherapy (RT) has been the standard-of-care treatment for patients with glioblastoma (GBM); however, the clinical effectiveness is hindered by therapeutic resistance. Here, we demonstrated that the tumor immune microenvironment (TIME) exhibited immunosuppressive properties and high expression of Golgi phosphoprotein 3 like (GOLPH3L) in RT-resistant GBM. Our study showed that GOLPH3L interacted with stimulator of interferon genes (STING) at the aspartic acid residue 184 in Golgi after RT, leading to coat protein complex II-mediated retrograde transport of STING from Golgi to endoplasmic reticulum. This suppressed the STING-NOD-like receptor thermal protein domain associated protein 3 (NLRP3)-mediated pyroptosis, resulting in suppressive TIME, driving GBM resistance to RT. Genetic GOLPH3L ablation in RT-resistant GBM cells augmented antitumor immunity and overcame tumor resistance to RT. Moreover, we have identified a small molecular inhibitor of GOLPH3L, vitamin B5 calcium (VB5), which improved the therapeutic efficacy of RT and immune checkpoint blockade by inducing a robust antitumor immune response in mouse models. Clinically, patients with GBM treated with VB5 exhibited improved responses to RT. Thus, reprogramming the TIME by targeting GOLPH3L may offer a potential opportunity to improve RT in GBM.
Collapse
Affiliation(s)
- Shuo Sun
- Department of Neurosurgery, First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Shiyu Qian
- Department of Pharmacy, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing 210008, China
| | - Ran Wang
- Department of Neurosurgery, Affiliated Brain Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Mengya Zhao
- Department of Immunology, Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi People's Hospital, Wuxi Medical Center, Nanjing Medical University, Nanjing 211166, China
- Research Center of Surgery, Nanjing BenQ Medical Center, Affiliated BenQ Hospital of Nanjing Medical University, Department of Immunology, Nanjing Medical University, Nanjing 211166, China
| | - Ran Li
- Department of Immunology, Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi People's Hospital, Wuxi Medical Center, Nanjing Medical University, Nanjing 211166, China
| | - Wei Gu
- Department of Neurosurgery, First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Mengjie Zhao
- Department of Neurosurgery, Affiliated Brain Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Chunfa Qian
- Department of Neurosurgery, Affiliated Brain Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Liang Liu
- Department of Neurosurgery, Affiliated Brain Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Xianglong Tang
- Department of Neurosurgery, Affiliated Brain Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Yangyang Li
- Department of Neurosurgery, Affiliated Brain Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Hui Shi
- Department of Neurosurgery, First Hospital of Lianyungang, Lianyungang 222000, China
| | - Yunsong Pan
- Department of Neurosurgery, First Hospital of Lianyungang, Lianyungang 222000, China
| | - Hong Xiao
- Department of Neurosurgery, Affiliated Brain Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Kun Yang
- Department of Neurosurgery, Affiliated Brain Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Chupeng Hu
- Department of Immunology, Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi People's Hospital, Wuxi Medical Center, Nanjing Medical University, Nanjing 211166, China
| | - Yedi Huang
- Department of Immunology, Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi People's Hospital, Wuxi Medical Center, Nanjing Medical University, Nanjing 211166, China
| | - Liangnian Wei
- Department of Immunology, Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi People's Hospital, Wuxi Medical Center, Nanjing Medical University, Nanjing 211166, China
| | - Yuhan Zhang
- Department of Immunology, Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi People's Hospital, Wuxi Medical Center, Nanjing Medical University, Nanjing 211166, China
| | - Jing Ji
- Department of Neurosurgery, First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
- Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing 211166, China
| | - Yun Chen
- Department of Immunology, Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi People's Hospital, Wuxi Medical Center, Nanjing Medical University, Nanjing 211166, China
- Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing 211166, China
| | - Hongyi Liu
- Department of Neurosurgery, Affiliated Brain Hospital of Nanjing Medical University, Nanjing 210029, China
| |
Collapse
|
93
|
Ran X, Wu BX, Vidhyasagar V, Song L, Zhang X, Ladak RJ, Teng M, Ba-Alawi W, Philip V, He HH, Sonenberg N, Lok BH. PARP inhibitor radiosensitization enhances anti-PD-L1 immunotherapy through stabilizing chemokine mRNA in small cell lung cancer. Nat Commun 2025; 16:2166. [PMID: 40038278 DOI: 10.1038/s41467-025-57257-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Accepted: 02/17/2025] [Indexed: 03/06/2025] Open
Abstract
Immunotherapy (IO) is an effective treatment for various cancers; however, the benefits are modest for small cell lung cancer (SCLC). The poor response of SCLC to anti-PD-1/PD-L1 IO is due in part to the lack of cytotoxic T cells because of limited chemokine expression from SCLC tumors. Immunogenic radiosensitizers that enhance chemokine expression may be a promising strategy forward. Here, we show that the PARP inhibitors (PARPi), including olaparib, talazoparib and veliparib, in combination with radiotherapy (RT) enhance the immune activation and anti-tumor efficacy in SCLC cell lines, patient-derived xenograft (PDX) and syngeneic mouse models. The effect is further enhanced by continued delivery of adjuvant PARPi. The combination treatment (PARPi with RT) activates the cGAS-STING pathway and increases the mRNA levels of the T cell chemo-attractants CCL5 and CXCL10. In addition to upregulation of transcription, the combination treatment increases chemokine CXCL10 protein levels via stabilization of CXCL10 mRNA in an EIF4E2-dependent manner. The incorporation of anti-PD-L1 IO into the PARPi with RT combination therapy further improves the anti-tumor efficacy by increasing T cell infiltration and function. This study thus provides a proof of principle for the combination of PARP inhibitors, RT and anti-PD-L1 IO as a treatment strategy for SCLC.
Collapse
Affiliation(s)
- Xiaozhuo Ran
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Bell Xi Wu
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
- Department of Medical Biophysics, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | | | - Lifang Song
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Xu Zhang
- Rosalind and Morris Goodman Cancer Institute, McGill University, Montreal, QC, Canada
- Department of Biochemistry, McGill University, Montreal, QC, Canada
| | - Reese Jalal Ladak
- Rosalind and Morris Goodman Cancer Institute, McGill University, Montreal, QC, Canada
- Department of Biochemistry, McGill University, Montreal, QC, Canada
| | - Mona Teng
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
- Department of Medical Biophysics, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - Wail Ba-Alawi
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Vivek Philip
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Housheng H He
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
- Department of Medical Biophysics, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - Nahum Sonenberg
- Rosalind and Morris Goodman Cancer Institute, McGill University, Montreal, QC, Canada
- Department of Biochemistry, McGill University, Montreal, QC, Canada
| | - Benjamin H Lok
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada.
- Department of Medical Biophysics, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada.
- Institute of Medical Science, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada.
- Department of Radiation Oncology, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
94
|
Ge X, Zhu X, Liu W, Li M, Zhang Z, Zou M, Deng M, Cui H, Chen Z, Wang L, Hu X, Ju R, Tang X, Ding X, Gong L. cGAMP promotes inner blood-retinal barrier breakdown through P2RX7-mediated transportation into microglia. J Neuroinflammation 2025; 22:58. [PMID: 40025497 PMCID: PMC11871612 DOI: 10.1186/s12974-025-03391-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2024] [Accepted: 02/20/2025] [Indexed: 03/04/2025] Open
Abstract
BACKGROUND Impairment of the inner blood-retinal barrier (iBRB) leads to various blinding diseases including diabetic retinopathy (DR). The cGAS-STING pathway has emerged as a driving force of cardiovascular destruction, but its impact on the neurovascular system is unclear. Here, we show that cGAMP, the endogenous STING agonist, causes iBRB breakdown and retinal degeneration thorough P2RX7-mediated transport into microglia. METHODS Extracellular cGAMP and STING pathway were determined in tissue samples from patients with proliferative DR (PDR) and db/db diabetic mice. Histological, molecular, bioinformatic and behavioral analysis accessed effects of cGAMP on iBRB. Single-cell RNA sequencing identified the primary retinal cell type responsive to cGAMP. Specific inhibitors and P2RX7-deficienct mice were used to evaluate P2RX7' role as a cGAMP transporter. The therapeutic effects of P2RX7 inhibitor were tested in db/db mice. RESULTS cGAMP was detected in the aqueous humor of patients with PDR and elevated in the vitreous humor with STING activation in db/db mouse retinas. cGAMP administration led to STING-dependent iBRB breakdown and neuron degeneration. Microglia were the primary cells responding to cGAMP, essential for cGAMP-induced iBRB breakdown and visual impairment. The ATP-gated P2RX7 transporter was required for cGAMP import and STING activation in retinal microglia. Contrary to previous thought that mouse P2RX7 nonselectively transports cGAMP only at extremely high ATP concentrations, human P2RX7 directly binds to cGAMP and activates STING under physiological conditions. Clinically, cGAMP-induced microglial signature was recapitulated in fibrovascular membranes from patients with PDR, with P2RX7 being predominantly expressed in microglia. Inhibiting P2RX7 reduced cGAMP-STING activation, protected iBRB and improved neuron survival in diabetic mouse retinas. CONCLUSIONS Our study reveals a mechanism for cGAMP-mediated iBRB breakdown and suggests that targeting microglia and P2RX7 may mitigate the deleterious effects of STING activation in retinal diseases linked to iBRB impairment.
Collapse
Affiliation(s)
- Xiangyu Ge
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Sun Yat-Sen University, Guangzhou, 510060, China
| | - Xingfei Zhu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Sun Yat-Sen University, Guangzhou, 510060, China
| | - Wei Liu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Sun Yat-Sen University, Guangzhou, 510060, China
| | - Mingsen Li
- Interdisciplinary Eye Research Institute (EYE-X Institute), Anhui Provincial Key Laboratory of Immunology in Chronic Diseases, Bengbu Medical University, Bengbu, China
| | - Zhaotian Zhang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Sun Yat-Sen University, Guangzhou, 510060, China
| | - Ming Zou
- Health Science Center, Peking University International Cancer Institute, Peking University, Beijing, China
| | - Mi Deng
- Health Science Center, Peking University International Cancer Institute, Peking University, Beijing, China
- Peking University Cancer Hospital and Institute, Peking University, Beijing, China
| | - Haifeng Cui
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Sun Yat-Sen University, Guangzhou, 510060, China
| | - Ziqing Chen
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Sun Yat-Sen University, Guangzhou, 510060, China
| | - Li Wang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Sun Yat-Sen University, Guangzhou, 510060, China
| | - Xuebin Hu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Sun Yat-Sen University, Guangzhou, 510060, China
| | - Rong Ju
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Sun Yat-Sen University, Guangzhou, 510060, China
| | - Xiangcheng Tang
- Shenzhen Eye Hospital, Shenzhen Eye Medical Center, Southern Medical University, Shenzhen, 518040, Guangdong, China.
| | - Xiaoyan Ding
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Sun Yat-Sen University, Guangzhou, 510060, China.
| | - Lili Gong
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Sun Yat-Sen University, Guangzhou, 510060, China.
| |
Collapse
|
95
|
Cao Z, Zhang Y, Jia H, Sun X, Feng Y, Wu H, Xu B, Wei Z. Immune checkpoint inhibitors mediate myocarditis by promoting macrophage polarization via cGAS/STING pathway. Cytokine 2025; 187:156873. [PMID: 39884185 DOI: 10.1016/j.cyto.2025.156873] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2024] [Revised: 12/16/2024] [Accepted: 01/10/2025] [Indexed: 02/01/2025]
Abstract
BACKGROUND Immune checkpoint inhibitors has opened up new avenues for cancer treatment, but serious cardiac injury has emerged in their use. A large number of data have shown that abnormal activation of cytosolic DNA-sensing cyclic GMP-AMP synthase-interferon gene activator pathway is closely related to cardiovascular inflammation and autoimmune diseases. However, the pathophysiological function of the cGAS-STING cascade in myocarditis induced by Immune checkpoint inhibitors is unclear. METHODS In order to establish a Immune checkpoint inhibitors-associated myocarditis model, BALB/c mice were injected with mouse cardiac troponin I peptide and anti-mouse programmed death 1 antibody. Echocardiography and HE staining were then performed to assess cardiac function and inflammation. Macrophages and damaged DNA in mouse heart tissue were detected by immunofluorescence. The mitochondrial damage of macrophages was observed by electron microscope. In vitro experiments, RAW264.7 was used to detect macrophage polarization after anti-PD-1 antibody induction and STING inhibition by qPCR and flow cytometry. Mitochondrial damage was detected by immunofluorescence, and activation of the cGAS-STING signaling pathway was evaluated by protein imprinting analysis. RESULTS In the Immune checkpoint inhibitors-associated myocarditis model, DNA damage was found to activate the cGAS-STING pathway and macrophages were polarized to M1 type. In vitro experiments, anti-PD-1 antibody activate the cGAS-STING pathway through the release of damaged DNA from macrophage mitochondrial damage, causing macrophage polarization into a pro-inflammatory phenotype leading to autoimmune myocarditis. CONCLUSION Our results suggested that the cGAS-STING pathway played a key role in myocarditis caused by immune checkpoint inhibitors. It provided a new possibility for Immune checkpoint inhibitors to be widely used in clinic.
Collapse
Affiliation(s)
- Zhenzhu Cao
- Department of Cardiology, Nanjing Drum Tower Hospital, Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, 358 Zhongshan Road, 210008 Nanjing, China
| | - Yu Zhang
- Department of Cardiology, Nanjing Drum Tower Hospital, Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, 358 Zhongshan Road, 210008 Nanjing, China
| | - Huihui Jia
- Department of Cardiology, Nanjing Drum Tower Hospital, Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, 358 Zhongshan Road, 210008 Nanjing, China
| | - Xuan Sun
- Department of Cardiology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, 358 Zhongshan Road, 210008 Nanjing, China
| | - Yuting Feng
- Department of Cardiology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, 358 Zhongshan Road, 210008 Nanjing, China
| | - Han Wu
- Department of Cardiology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, 358 Zhongshan Road, 210008 Nanjing, China.
| | - Biao Xu
- Department of Cardiology, Nanjing Drum Tower Hospital, Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, 358 Zhongshan Road, 210008 Nanjing, China.
| | - Zhonghai Wei
- Department of Cardiology, Nanjing Drum Tower Hospital, Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, 358 Zhongshan Road, 210008 Nanjing, China; Department of Cardiology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, 358 Zhongshan Road, 210008 Nanjing, China.
| |
Collapse
|
96
|
Lu S, Yang Y, Song Z, Cao J, Han Z, Chen L, He Y, Wang J, Teng Y, Zhang Z, Zou J, Ge J, Yang H, Cheng L. Dual functional nanoplatforms potentiate osteosarcoma immunotherapy via microenvironment modulation. Natl Sci Rev 2025; 12:nwaf002. [PMID: 39936146 PMCID: PMC11812574 DOI: 10.1093/nsr/nwaf002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Revised: 12/13/2024] [Accepted: 01/06/2025] [Indexed: 02/13/2025] Open
Abstract
Osteosarcoma (OS), a highly aggressive bone tumor, presents significant challenges in terms of effective treatment. We identified that cellular autophagy was impaired within OS by comparing clinical OS samples through bioinformatic analyses and further validated the inhibition of mitochondrial autophagy in OS at the transcriptomic level. Based on this finding, we investigated the therapeutic potential of a dual functional metal nanoplatform (MnSx) to facilitate a transition from the protective effect of low-level autophagy in OS to the killing effect of high-level autophagy in OS. MnSx facilitated intracellular H2S generation via endocytosis, leading to the S-sulfhydration of ubiquitin-specific peptidase 8 (USP8) and subsequent promotion of mitochondrial autophagy in vitro. Additionally, MnSx activated the cyclic guanosine monophosphate-adenosine monophosphate synthase (cGAS)-stimulator of interferon genes (STING) pathway, further enhancing the cellular autophagic response and accelerating tumor cell death. Moreover, it was demonstrated in vivo that MnSx, on the one hand, mediated the activation of tumor autophagy by USP8 via intracellular H2S, while Mn2+ promoted the maturation of dendritic cells, activated cytotoxic T lymphocytes and contributed to tumor eradication. Such tumor killing could be suppressed by the autophagy inhibitor chloroquine. Importantly, synergistic combination therapy with immune checkpoint inhibitors showed promise for achieving complete remission of OS. This study highlights the potential of MnSx as a dual-functional therapeutic platform for OS treatment and offers novel directions for future research in this field.
Collapse
Affiliation(s)
- Shunyi Lu
- Department of Orthopedic Surgery, The First Affiliated Hospital of Soochow University, Suzhou 215123, China
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices, Soochow University, Suzhou 215123, China
| | - Yuqi Yang
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices, Soochow University, Suzhou 215123, China
| | - Zhuorun Song
- Department of Orthopedic Surgery, The First Affiliated Hospital of Soochow University, Suzhou 215123, China
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices, Soochow University, Suzhou 215123, China
| | - Jie Cao
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices, Soochow University, Suzhou 215123, China
| | - Zhihui Han
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices, Soochow University, Suzhou 215123, China
| | - Linfu Chen
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices, Soochow University, Suzhou 215123, China
| | - Yunfei He
- Soochow University Institues for Translational Medicine, The First Affiliated Hospital of Soochow University, Suzhou 215123, China
| | - Jiayi Wang
- Department of Orthopedic Surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, China
| | - Yun Teng
- Department of Orthopedic Surgery, The First Affiliated Hospital of Soochow University, Suzhou 215123, China
| | - Zengli Zhang
- Department of Environmental Health School of Public Health, Soochow University, Suzhou 215123, China
| | - Jun Zou
- Department of Orthopedic Surgery, The First Affiliated Hospital of Soochow University, Suzhou 215123, China
| | - Jun Ge
- Department of Orthopedic Surgery, The First Affiliated Hospital of Soochow University, Suzhou 215123, China
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices, Soochow University, Suzhou 215123, China
| | - Huilin Yang
- Department of Orthopedic Surgery, The First Affiliated Hospital of Soochow University, Suzhou 215123, China
| | - Liang Cheng
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices, Soochow University, Suzhou 215123, China
| |
Collapse
|
97
|
Zhou Z, Guo F, Zhang J, Liao L, Jiang M, Huang Y, Liu Y, Lei L, Tao Z, Yu CY, Wei H. Facile integration of a binary nano-prodrug with αPD-L1 as a translatable technology for potent immunotherapy of TNBC. Acta Biomater 2025; 194:373-384. [PMID: 39870152 DOI: 10.1016/j.actbio.2025.01.038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2024] [Revised: 01/20/2025] [Accepted: 01/23/2025] [Indexed: 01/29/2025]
Abstract
Immune checkpoint blockers (ICBs)-based immunotherapy is a favorable approach for efficient triple-negative breast cancer (TNBC) treatment. However, the therapeutic efficacy of ICBs is greatly compromised by immunosuppressive tumor microenvironments (TMEs) and low expression levels of programmed cell death ligand-1 (PD-L1). Herein, we constructed an amphiphilic prodrug by linking a hydrophobic STING agonist, MSA-2 and a hydrophilic chemotherapeutic drug, gemcitabine (GEM) via an ester bond, which can self-assemble into GEM-MSA-2 (G-M) nanoparticles (NPs) with a tumor growth inhibition (TGI) value of 87.1 % in a murine 4T1 transplantation tumor model. Notably, the immunogenic cell death (ICD)-triggering effect of GEM together with the cyclic GMP-AMP synthase-stimulator of interferon genes (cGAS-STING) pathway activation properties of MSA-2 enables efficient infiltration of non-exhausting T cells and repolarization of macrophages from M2 to M1 types in the tumor microenvironment for transforming a cold tumor to a hot one. Most importantly, G-M NPs treatment increases the PD-L1 expression levels, thus providing a unique opportunity for further integration with anti-PD-L1 monoclonal antibody (αPD-L1) for eliciting stronger immunity that ultimately leads to a TGI value of 98.0 % in the primary tumor and significantly protects against distal and disseminated tumor rechallenge. Overall, this study presents a minimalist nano-prodrug combined with αPD-L1 as a simple yet robust translatable nanotechnology for potent chemo-immunotherapy of TNBC. STATEMENT OF SIGNIFICANCE: Enhancing the therapeutic efficacy of αPD-L1 for tumor immunotherapy via a translatable technology remains a challenge. We report herein facile integration of a binary nano-prodrug with αPD-L1 for potent immunotherapy of TNBC. An amphiphilic prodrug is constructed by linking a hydrophobic STING agonist, MSA-2 and a hydrophilic chemotherapeutic drug, gemcitabine (GEM) via an ester bond. The resulting self-assembled GEM-MSA-2 (G-M) nanoparticles (NPs) show a tumor growth inhibition (TGI) value of 87.1 % in a murine 4T1 transplantation tumor model. Besides the induced immunogenic cell death (ICD) and activated cGAS-STING pathway, G-M NPs increase the PD-L1 expression levels, providing a unique opportunity for further integration with αPD-L1 to elicit stronger immunity that ultimately leads to a TGI value of 98.0 % in the primary tumor and significantly protects against distal and disseminated tumor rechallenge.
Collapse
Affiliation(s)
- Zongtao Zhou
- Hengyang Medical School, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, School of Pharmaceutical Science, MOE Key Lab of Rare Pediatric Disease, University of South China, Hengyang 421001, China
| | - Fangru Guo
- Hengyang Medical School, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, School of Pharmaceutical Science, MOE Key Lab of Rare Pediatric Disease, University of South China, Hengyang 421001, China
| | - Jinyan Zhang
- Hengyang Medical School, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, School of Pharmaceutical Science, MOE Key Lab of Rare Pediatric Disease, University of South China, Hengyang 421001, China
| | - Luanfeng Liao
- Hengyang Medical School, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, School of Pharmaceutical Science, MOE Key Lab of Rare Pediatric Disease, University of South China, Hengyang 421001, China
| | - Mingchao Jiang
- Hengyang Medical School, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, School of Pharmaceutical Science, MOE Key Lab of Rare Pediatric Disease, University of South China, Hengyang 421001, China
| | - Yun Huang
- Hengyang Medical School, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, School of Pharmaceutical Science, MOE Key Lab of Rare Pediatric Disease, University of South China, Hengyang 421001, China
| | - Ying Liu
- Hengyang Medical School, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, School of Pharmaceutical Science, MOE Key Lab of Rare Pediatric Disease, University of South China, Hengyang 421001, China
| | - Longtianyang Lei
- Hengyang Medical School, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, School of Pharmaceutical Science, MOE Key Lab of Rare Pediatric Disease, University of South China, Hengyang 421001, China
| | - Zhenghao Tao
- Hengyang Medical School, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, School of Pharmaceutical Science, MOE Key Lab of Rare Pediatric Disease, University of South China, Hengyang 421001, China
| | - Cui-Yun Yu
- Hengyang Medical School, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, School of Pharmaceutical Science, MOE Key Lab of Rare Pediatric Disease, University of South China, Hengyang 421001, China; Affiliated Hospital of Hunan Academy of Chinese Medicine, Hunan Academy of Chinese Medicine, Changsha 410013, China.
| | - Hua Wei
- Hengyang Medical School, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, School of Pharmaceutical Science, MOE Key Lab of Rare Pediatric Disease, University of South China, Hengyang 421001, China.
| |
Collapse
|
98
|
Zhang X, Chen Y, Liu X, Li G, Zhang S, Zhang Q, Cui Z, Qin M, Simon HU, Terzić J, Kocic G, Polić B, Yin C, Li X, Zheng T, Liu B, Zhu Y. STING in cancer immunoediting: Modeling tumor-immune dynamics throughout cancer development. Cancer Lett 2025; 612:217410. [PMID: 39826670 DOI: 10.1016/j.canlet.2024.217410] [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: 09/09/2024] [Revised: 12/16/2024] [Accepted: 12/21/2024] [Indexed: 01/22/2025]
Abstract
Cancer immunoediting is a dynamic process of tumor-immune system interaction that plays a critical role in cancer development and progression. Recent studies have highlighted the importance of innate signaling pathways possessed by both cancer cells and immune cells in this process. The STING molecule, a pivotal innate immune signaling molecule, mediates DNA-triggered immune responses in both cancer cells and immune cells, modulating the anti-tumor immune response and shaping the efficacy of immunotherapy. Emerging evidence has shown that the activation of STING signaling has dual opposing effects in cancer progression, simultaneously provoking and restricting anti-tumor immunity, and participating in every phase of cancer immunoediting, including immune elimination, equilibrium, and escape. In this review, we elucidate the roles of STING in the process of cancer immunoediting and discuss the dichotomous effects of STING agonists in the cancer immunotherapy response or resistance. A profound understanding of the sophisticated roles of STING signaling pathway in cancer immunoediting would potentially inspire the development of novel cancer therapeutic approaches and overcome the undesirable protumor effects of STING activation.
Collapse
Affiliation(s)
- Xiao Zhang
- Department of Oral and Maxillofacial Surgery, The First Affiliated Hospital of Harbin Medical University, 23 Youzheng Street, Nangang District, Harbin 150001, People's Republic of China; Department of Pathology, Harbin Medical University, Harbin, 150081, People's Republic of China
| | - Yan Chen
- Department of Pathology, Harbin Medical University, Harbin, 150081, People's Republic of China
| | - Xi Liu
- Department of Cardiology, ordos central hospital, Ordos, People's Republic of China
| | - Guoli Li
- Department of Colorectal and Anal Surgery, Chifeng Municipal Hospital, Chifeng Clinical Medical School of Inner Mongolia Medical University, Chifeng, People's Republic of China
| | - Shuo Zhang
- Department of Oral and Maxillofacial Surgery, The First Affiliated Hospital of Harbin Medical University, 23 Youzheng Street, Nangang District, Harbin 150001, People's Republic of China
| | - Qi Zhang
- Department of Pathology, Harbin Medical University, Harbin, 150081, People's Republic of China
| | - Zihan Cui
- Department of Pathology, Harbin Medical University, Harbin, 150081, People's Republic of China
| | - Minglu Qin
- Department of Pathology, Harbin Medical University, Harbin, 150081, People's Republic of China
| | - Hans-Uwe Simon
- Institute of Pharmacology, University of Bern, 3010 Bern, Switzerland; Institute of Biochemistry, Brandenburg Medical School, Neuruppin, 16816, Germany
| | - Janoš Terzić
- Laboratory for Cancer Research, University of Split School of Medicine, Split, Croatia
| | - Gordana Kocic
- Department of Biochemistry, Faculty of Medicine, University of Nis, 18000 Nis, Serbia
| | - Bojan Polić
- University of Rijeka Faculty of Medicine, Croatia
| | - Chengliang Yin
- Faculty of Medicine, Macau University of Science and Technology, 999078, Macao.
| | - Xiaobo Li
- Department of Pathology, Harbin Medical University, Harbin, 150081, People's Republic of China.
| | - Tongsen Zheng
- Department of Gastrointestinal Medical Oncology, Harbin Medical University Cancer Hospital, No.150 Haping Road, Nangang District, Harbin, Heilongjiang, People's Republic of China.
| | - Bing Liu
- Department of Oral and Maxillofacial Surgery, The First Affiliated Hospital of Harbin Medical University, 23 Youzheng Street, Nangang District, Harbin 150001, People's Republic of China; School of Stomatology, Harbin Medical University, Harbin, 150001, People's Republic of China.
| | - Yuanyuan Zhu
- Department of Oral and Maxillofacial Surgery, The First Affiliated Hospital of Harbin Medical University, 23 Youzheng Street, Nangang District, Harbin 150001, People's Republic of China; Department of Pathology, Harbin Medical University, Harbin, 150081, People's Republic of China.
| |
Collapse
|
99
|
Messaoud‐Nacer Y, Culerier E, Rose S, Maillet I, Boussad R, Veront C, Savigny F, Malissen B, Radzikowska U, Sokolowska M, da Silva GVL, Edwards MR, Jackson DJ, Johnston SL, Ryffel B, Quesniaux VF, Togbe D. STING-dependent induction of neutrophilic asthma exacerbation in response to house dust mite. Allergy 2025; 80:715-737. [PMID: 39466641 PMCID: PMC11891437 DOI: 10.1111/all.16369] [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: 10/13/2023] [Revised: 07/22/2024] [Accepted: 09/01/2024] [Indexed: 10/30/2024]
Abstract
BACKGROUND Severe refractory, neutrophilic asthma remains an unsolved clinical problem. STING agonists induce a neutrophilic response in the airways, suggesting that STING activation may contribute to the triggering of neutrophilic exacerbations. We aim to determine whether STING-induced neutrophilic lung inflammation mimics severe asthma. METHODS We developed new models of neutrophilic lung inflammation induced by house dust mite (HDM) plus STING agonists diamidobenzimidazole (diABZI) or cGAMP in wild-type, and conditional-STING-deficient mice. We measured DNA damage, cell death, NETs, cGAS/STING pathway activation by immunoblots, N1/N2 balance by flow cytometry, lung function by plethysmography, and Th1/Th2 cytokines by multiplex. We evaluated diABZI effects on human airway epithelial cells from healthy or patients with asthma, and validated the results by transcriptomic analyses of rhinovirus infected healthy controls vs patients with asthma. RESULTS DiABZI administration during HDM challenge increased airway hyperresponsiveness, neutrophil recruitment with prominent NOS2+ARG1- type 1 neutrophils, protein extravasation, cell death by PANoptosis, NETs formation, extracellular dsDNA release, DNA sensors activation, IFNγ, IL-6 and CXCL10 release. Functionally, STING agonists exacerbated airway hyperresponsiveness. DiABZI caused DNA and epithelial barrier damage, STING pathway activation in human airway epithelial cells exposed to HDM, in line with DNA-sensing and PANoptosis pathways upregulation and tight-junction downregulation induced by rhinovirus challenge in patients with asthma. CONCLUSIONS Our study identifies that triggering STING in the context of asthma induces cell death by PANoptosis, fueling the flame of inflammation through a mixed Th1/Th2 immune response recapitulating the features of severe asthma with a prognostic signature of type 1 neutrophils.
Collapse
Affiliation(s)
- Yasmine Messaoud‐Nacer
- Experimental and Molecular Immunology and NeurogeneticsINEM UMR7355 University of Orleans and CNRSOrleansFrance
| | - Elodie Culerier
- Experimental and Molecular Immunology and NeurogeneticsINEM UMR7355 University of Orleans and CNRSOrleansFrance
| | - Stéphanie Rose
- Experimental and Molecular Immunology and NeurogeneticsINEM UMR7355 University of Orleans and CNRSOrleansFrance
| | - Isabelle Maillet
- Experimental and Molecular Immunology and NeurogeneticsINEM UMR7355 University of Orleans and CNRSOrleansFrance
| | - Rania Boussad
- Experimental and Molecular Immunology and NeurogeneticsINEM UMR7355 University of Orleans and CNRSOrleansFrance
| | - Chloé Veront
- Experimental and Molecular Immunology and NeurogeneticsINEM UMR7355 University of Orleans and CNRSOrleansFrance
| | - Florence Savigny
- Experimental and Molecular Immunology and NeurogeneticsINEM UMR7355 University of Orleans and CNRSOrleansFrance
| | - Bernard Malissen
- Centre d'Immunophénomique (CIPHE)Aix Marseille Université, INSERM, CNRSMarseilleFrance
| | - Urszula Radzikowska
- Swiss Institute of Allergy and Asthma Research (SIAF)University of ZurichDavosSwitzerland
- Christine Kühne—Center for Allergy Research and Education (CK‐CARE)Herman‐ Burchard‐Strasse 1DavosSwitzerland
| | - Milena Sokolowska
- Swiss Institute of Allergy and Asthma Research (SIAF)University of ZurichDavosSwitzerland
- Christine Kühne—Center for Allergy Research and Education (CK‐CARE)Herman‐ Burchard‐Strasse 1DavosSwitzerland
| | | | - Michael R. Edwards
- National Heart and Lung InstituteImperial College LondontLondonUK
- Asthma UK Centre in Allergic Mechanism of AsthmaLondonUK
- Imperial College Healthcare NHS TrustLondonUK
| | - David J. Jackson
- Guy’s Severe Asthma Centre, Guy’s & St Thomas’ NHS TrustLondonUK
- School of Immunology & Microbial Sciences, King’s College LondonLondonUK
| | - Sebastian L. Johnston
- National Heart and Lung InstituteImperial College LondontLondonUK
- Asthma UK Centre in Allergic Mechanism of AsthmaLondonUK
- Imperial College Healthcare NHS TrustLondonUK
| | - Bernhard Ryffel
- Experimental and Molecular Immunology and NeurogeneticsINEM UMR7355 University of Orleans and CNRSOrleansFrance
| | - Valerie F. Quesniaux
- Experimental and Molecular Immunology and NeurogeneticsINEM UMR7355 University of Orleans and CNRSOrleansFrance
| | - Dieudonnée Togbe
- Experimental and Molecular Immunology and NeurogeneticsINEM UMR7355 University of Orleans and CNRSOrleansFrance
| |
Collapse
|
100
|
Lu M, Wu J, Gao Q, Jin R, An C, Ma T. To cleave or not and how? The DNA exonucleases and endonucleases in immunity. Genes Dis 2025; 12:101219. [PMID: 39759116 PMCID: PMC11697192 DOI: 10.1016/j.gendis.2024.101219] [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: 07/19/2023] [Revised: 11/02/2023] [Accepted: 12/29/2023] [Indexed: 01/07/2025] Open
Abstract
DNA exonucleases and endonucleases are key executors of the genome during many physiological processes. They generate double-stranded DNA by cleaving damaged endogenous or exogenous DNA, triggering the activation of the innate immune pathways such as cGAS-STING-IFN, and enabling the body to produce anti-viral or anti-tumor immune responses. This is of great significance for maintaining the stability of the genome and improving the therapeutic efficacy of tumors. In addition, genomic instability caused by exonuclease mutations contributes to the development of various autoimmune diseases. This review summarizes the DNA exonucleases and endonucleases which have critical functions in immunity and associated diseases.
Collapse
Affiliation(s)
- Mingjun Lu
- Cancer Research Center, Beijing Chest Hospital, Capital Medical University/Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing 101149, China
| | - Jinghong Wu
- Cancer Research Center, Beijing Chest Hospital, Capital Medical University/Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing 101149, China
| | - Qing Gao
- Cancer Research Center, Beijing Chest Hospital, Capital Medical University/Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing 101149, China
| | - Renjing Jin
- Cancer Research Center, Beijing Chest Hospital, Capital Medical University/Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing 101149, China
| | - Changming An
- Department of Head and Neck Surgery, Cancer Hospital, National Cancer Center, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100021, China
| | - Teng Ma
- Cancer Research Center, Beijing Chest Hospital, Capital Medical University/Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing 101149, China
| |
Collapse
|