1
|
Cui Y, Lin H, Ma J, Zhao Y, Li J, Wang Y, Zhuang J, Yang Y. Ischemia-reperfusion injury induces ZBP1-dependent PANoptosis in endothelial cells. Biochim Biophys Acta Mol Basis Dis 2025; 1871:167782. [PMID: 40057207 DOI: 10.1016/j.bbadis.2025.167782] [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: 10/10/2024] [Revised: 02/02/2025] [Accepted: 03/04/2025] [Indexed: 04/15/2025]
Abstract
Endothelial cells play a critical role in the pathophysiology of ischemia-reperfusion injury (IRI). Although previous studies have shown that IRI can activate PANoptosis, the underlying mechanisms remain unclear. Our research investigates how IRI induces PANoptosis in endothelial cells, aiming to identify protective strategies to safeguard these cells from PANoptosis triggered by IRI. We established an in vitro endothelial cell hypoxia/reoxygenation (H/R) treatment model and an in vivo SD rat free flap IRI model. A series of assays, including PI/Hoechst staining, Western blotting, and immunohistochemistry, were conducted to assess PANoptosis-like cell death in endothelial cells. Cell transfection with ZBP1 siRNA and immunoprecipitation were used to explore the involved signaling pathways. Our results showed activation of PANoptosis-like cell death and upregulation of ZBP1 expression following IRI. After knocking down ZBP1 expression, a significant alteration in PANoptosis-like cell death and the assembly of the ZBP1-PANoptosome in endothelial cells was observed, confirming the occurrence of PANoptosis. In conclusion, our research confirms that IRI induces PANoptosome formation, promoting ZBP1-dependent PANoptosis in endothelial cells.
Collapse
Affiliation(s)
- Yue Cui
- Department of Aesthetic Plastic Surgery and Laser Medicine, Beijing Anzhen Hospital Affiliated to Capital Medical University, Beijing 100029, China
| | - Huang Lin
- Department of Aesthetic Plastic Surgery and Laser Medicine, Beijing Anzhen Hospital Affiliated to Capital Medical University, Beijing 100029, China.
| | - Jiaxing Ma
- Department of Aesthetic Plastic Surgery and Laser Medicine, Beijing Anzhen Hospital Affiliated to Capital Medical University, Beijing 100029, China
| | - Yinhua Zhao
- Department of Aesthetic Plastic Surgery and Laser Medicine, Beijing Anzhen Hospital Affiliated to Capital Medical University, Beijing 100029, China
| | - Jiaxi Li
- Department of Aesthetic Plastic Surgery and Laser Medicine, Beijing Anzhen Hospital Affiliated to Capital Medical University, Beijing 100029, China
| | - Yang Wang
- Department of Aesthetic Plastic Surgery and Laser Medicine, Beijing Anzhen Hospital Affiliated to Capital Medical University, Beijing 100029, China
| | - Jingwei Zhuang
- Department of Aesthetic Plastic Surgery and Laser Medicine, Beijing Anzhen Hospital Affiliated to Capital Medical University, Beijing 100029, China
| | - Yu Yang
- Department of Plastic Surgery, Fujian Provincial Hospital, Provincial Clinical College of Fujian Medical University, Fuzhou 350001, China
| |
Collapse
|
2
|
Allushi B, Chlebicz M, Kumar G, Massey K, Labombarde JG, Turner S, Miller RAJ, Williams AP, Quinn A, Kovats S, Axtell RC. Interferon-β treatment reverses the detrimental effect of B-cell depletion therapy on respiratory virus infection. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2025:vkaf085. [PMID: 40334083 DOI: 10.1093/jimmun/vkaf085] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Accepted: 03/25/2025] [Indexed: 05/09/2025]
Abstract
Disease-modifying therapies (DMTs) are critical for managing autoimmunity such as multiple sclerosis (MS), yet concerns exist regarding their impact on viral infections. B-cell depletion (α-CD20) and IFN-β are 2 DMTs with seemingly opposing effects on viral infections. Pre-vaccine COVID-19 data linked B-cell depletion to worse outcomes, while IFN-β is believed to offer protection to viral infection. The mechanisms underlying the interactions between these DMTs and infection have yet to be fully elucidated. Our goal was to determine the modulatory effects of α-CD20 and IFN-β, administered individually or in combination, during acute respiratory viral infections in mice. In our study, B-cell depletion was achieved by administering α-CD20 antibodies 3 times every 5 days, starting 7 days before influenza A virus (IAV) infection. IFN-β was administered on days 1 and 2 p.i. α-CD20 administered alone exacerbated infection outcomes. At day 9 postinfection, mice treated with α-CD20 had elevated viral RNA, accompanied by greater weight loss, impaired viral clearance, heightened myeloid cell infiltration in the lungs, and elevated systemic inflammatory cytokines in the blood. Notably, T-cell responses to IAV were not inhibited by α-CD20. IFN-β monotherapy failed to confer significant protection against viral infection, but when combined with α-CD20, it reversed the exacerbated effects of B-cell depletion by reducing viral load, improving morbidity, limiting neutrophil infiltration, and restoring cytokine homeostasis. These findings suggest IFN-β's capacity to counteract the deleterious impacts of α-CD20 on respiratory viral infections, offering potential treatment strategies for autoimmune diseases during viral outbreaks.
Collapse
Affiliation(s)
- Bujana Allushi
- Arthritis and Clinical Immunology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, United States
- Department of Microbiology and Immunology, Oklahoma University Health Science Center, Oklahoma City, OK, United States
| | - Magdalena Chlebicz
- Arthritis and Clinical Immunology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, United States
| | - Gaurav Kumar
- Arthritis and Clinical Immunology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, United States
| | - Kaylea Massey
- Arthritis and Clinical Immunology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, United States
| | - Jocelyn G Labombarde
- Arthritis and Clinical Immunology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, United States
- Department of Pathology, Oklahoma University Health Science Center, Oklahoma City, OK, United States
| | - Sean Turner
- Arthritis and Clinical Immunology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, United States
| | - Reegan A J Miller
- Arthritis and Clinical Immunology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, United States
- Department of Microbiology and Immunology, Oklahoma University Health Science Center, Oklahoma City, OK, United States
| | - Abigael P Williams
- Arthritis and Clinical Immunology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, United States
- Department of Microbiology and Immunology, Oklahoma University Health Science Center, Oklahoma City, OK, United States
| | - Amia Quinn
- Arthritis and Clinical Immunology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, United States
| | - Susan Kovats
- Arthritis and Clinical Immunology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, United States
- Department of Microbiology and Immunology, Oklahoma University Health Science Center, Oklahoma City, OK, United States
- Department of Pathology, Oklahoma University Health Science Center, Oklahoma City, OK, United States
| | - Robert C Axtell
- Arthritis and Clinical Immunology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, United States
- Department of Microbiology and Immunology, Oklahoma University Health Science Center, Oklahoma City, OK, United States
| |
Collapse
|
3
|
Nadendla EK, Tweedell RE, Kasof G, Kanneganti TD. Caspases: structural and molecular mechanisms and functions in cell death, innate immunity, and disease. Cell Discov 2025; 11:42. [PMID: 40325022 PMCID: PMC12052993 DOI: 10.1038/s41421-025-00791-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Accepted: 03/05/2025] [Indexed: 05/07/2025] Open
Abstract
Caspases are critical regulators of cell death, development, innate immunity, host defense, and disease. Upon detection of pathogens, damage-associated molecular patterns, cytokines, or other homeostatic disruptions, innate immune sensors, such as NLRs, activate caspases to initiate distinct regulated cell death pathways, including non-lytic (apoptosis) and innate immune lytic (pyroptosis and PANoptosis) pathways. These cell death pathways are driven by specific caspases and distinguished by their unique molecular mechanisms, supramolecular complexes, and enzymatic properties. Traditionally, caspases are classified as either apoptotic (caspase-2, -3, -6, -7, -8, -9, and -10) or inflammatory (caspase-1, -4, -5, and -11). However, extensive data from the past decades have shown that apoptotic caspases can also drive lytic inflammatory cell death downstream of innate immune sensing and inflammatory responses, such as in the case of caspase-3, -6, -7, and -8. Therefore, more inclusive classification systems based on function, substrate specificity, or the presence of pro-domains have been proposed to better reflect the multifaceted roles of caspases. In this review, we categorize caspases into CARD-, DED-, and short/no pro-domain-containing groups and examine their critical functions in innate immunity and cell death, along with their structural and molecular mechanisms, including active site/exosite properties and substrates. Additionally, we highlight the emerging roles of caspases in cellular homeostasis and therapeutic targeting. Given the clinical relevance of caspases across multiple diseases, improved understanding of these proteins and their structure-function relationships is critical for developing effective treatment strategies.
Collapse
Affiliation(s)
- Eswar Kumar Nadendla
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Rebecca E Tweedell
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Gary Kasof
- Cell Signaling Technology, Danvers, MA, USA
| | | |
Collapse
|
4
|
Wu J, Guo Z, Wang L, Shen Y, Li X, Zhang Z, Han X, Zhang J, Cai K, Tang C. Porphyromonas gingivalis induces Zbp1-mediated macrophages PANoptosis in periodonitis pathophysiology. Exp Mol Med 2025:10.1038/s12276-025-01443-y. [PMID: 40307566 DOI: 10.1038/s12276-025-01443-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2024] [Revised: 01/18/2025] [Accepted: 02/09/2025] [Indexed: 05/02/2025] Open
Abstract
Periodontitis is an oral immunoinflammatory disease, and macrophages play a crucial role in its pathophysiology. However, macrophage death during antibacterial activities will exacerbate inflammation and tissue damage. Porphyromonas gingivalis is a major constituent of subgingival biofilm plaques in periodontitis, but the effects and precise molecular mechanisms by which it triggers macrophage death remain unknown. Here we found that P. gingivalis infection notably activated multiple death pathways in bone-marrow-derived macrophages, including pyroptosis, apoptosis and necrosis. Furthermore, using RNA sequencing, we identified that P. gingivalis infection markedly increased the expression of Z-DNA binding protein 1 (Zbp1) in bone-marrow-derived macrophages. Initially identified as an interferon-induced tumor-associated protein, Zbp1 serves as an upstream sensor that regulates cell death by activating PANoptosis. Mechanistically, P. gingivalis induced a mitochondrial stress response, prompting the release of mitochondrial DNA. This mitochondrial DNA then interacted with Zbp1, consequently augmenting its downstream PANoptosis signals. In addition, P. gingivalis stimulated macrophage Zbp1 expression through the Tlr2/4-JNK-Stat3/5 pathway, exacerbating macrophage death. Importantly, blocking the biosynthesis of endogenous Zbp1 by pharmacological delivery with microneedles improved the survival of P. gingivalis-infected macrophages and inhibited periodontal tissue destruction. These findings highlight Zbp1 as a potential therapeutic target for P. gingivalis-induced periodontitis.
Collapse
Affiliation(s)
- Jin Wu
- State Key Laboratory Cultivation Base of Research, Prevention and Treatment for Oral Diseases, Nanjing Medical University, Nanjing, China
- Department of Oral Implantology Affiliated Hospital of Stomatology, Nanjing Medical University, Nanjing, China
| | - Zixiang Guo
- State Key Laboratory Cultivation Base of Research, Prevention and Treatment for Oral Diseases, Nanjing Medical University, Nanjing, China
- Department of Oral Implantology Affiliated Hospital of Stomatology, Nanjing Medical University, Nanjing, China
| | - Long Wang
- State Key Laboratory Cultivation Base of Research, Prevention and Treatment for Oral Diseases, Nanjing Medical University, Nanjing, China
- Department of Oral Implantology Affiliated Hospital of Stomatology, Nanjing Medical University, Nanjing, China
| | - Yue Shen
- State Key Laboratory Cultivation Base of Research, Prevention and Treatment for Oral Diseases, Nanjing Medical University, Nanjing, China
- Department of Oral Implantology Affiliated Hospital of Stomatology, Nanjing Medical University, Nanjing, China
| | - Xiang Li
- State Key Laboratory Cultivation Base of Research, Prevention and Treatment for Oral Diseases, Nanjing Medical University, Nanjing, China
- Department of Oral and Maxillofacial Surgery Affiliated Hospital of Stomatology, Nanjing Medical University, Nanjing, China
| | - Zhewei Zhang
- State Key Laboratory Cultivation Base of Research, Prevention and Treatment for Oral Diseases, Nanjing Medical University, Nanjing, China
- Department of Oral Implantology Affiliated Hospital of Stomatology, Nanjing Medical University, Nanjing, China
| | - Xiao Han
- State Key Laboratory Cultivation Base of Research, Prevention and Treatment for Oral Diseases, Nanjing Medical University, Nanjing, China
- Department of Oral Implantology Affiliated Hospital of Stomatology, Nanjing Medical University, Nanjing, China
| | - Jianlan Zhang
- Department of Oral Implantology Affiliated Hospital of Stomatology, Nanjing Medical University, Nanjing, China
| | - Kunzhan Cai
- State Key Laboratory Cultivation Base of Research, Prevention and Treatment for Oral Diseases, Nanjing Medical University, Nanjing, China.
- Department of Oral Implantology Affiliated Hospital of Stomatology, Nanjing Medical University, Nanjing, China.
| | - Chunbo Tang
- State Key Laboratory Cultivation Base of Research, Prevention and Treatment for Oral Diseases, Nanjing Medical University, Nanjing, China.
- Department of Oral Implantology Affiliated Hospital of Stomatology, Nanjing Medical University, Nanjing, China.
| |
Collapse
|
5
|
Liu Z, Chen DH, Lin ZH, Wang ZY, Peng H, Liu RT, Hu ZC, He YH, Wei XJ, Zhang CQ, Feng Y, Tang Q, Zhu ZZ. In-situ Sprayed platelet-derived small extracellular vesicles for the skin flap survival by reducing PANoptosis. Biomaterials 2025; 316:123001. [PMID: 39671720 DOI: 10.1016/j.biomaterials.2024.123001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Revised: 11/02/2024] [Accepted: 12/04/2024] [Indexed: 12/15/2024]
Abstract
Necrosis at the distal end of random skin flaps remains a significant challenge, limiting the clinical application of these flaps in plastic and reconstructive surgery. Inhibiting ischemia/reperfusion (I/R) injury and promoting the formation of neovascular networks are critical preventive strategies. Platelet-derived small extracellular vesicles (PL-sEV) are nanocarriers of growth factors that provide an alternative to clinically used platelet-rich plasma and platelet lysates, offering higher growth factor concentrations and lower immunogenicity. In this study, PANoptosis, a distinct form of inflammatory cell death, was fully characterized in a random skin flap model. Subcutaneous injection of PL-sEV improved ischemic skin flap survival by enhancing blood perfusion and reducing PANoptosis levels. In vitro, PL-sEV inhibited oxygen-glucose deprivation/reoxygenation-induced dysfunction in human umbilical vein endothelial cells. Furthermore, PL-sEV was incorporated into a thermosensitive triblock hydrogel, creating a sprayable delivery system (PLEL@PL-sEV). Mechanistic analysis through RNA sequencing indicated that the protective effects of PL-sEV against PANoptosis likely resulted from its anti-inflammatory properties, particularly via suppression of the NF-κB signaling pathway. This novel hydrogel system demonstrated controlled release of PL-sEV and proved effective in improving skin flap transplantation outcomes.
Collapse
Affiliation(s)
- Zhe Liu
- Department of Orthopedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 600 Yishan Road, Shanghai, 200233, China
| | - De-Heng Chen
- Department of Orthopedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 600 Yishan Road, Shanghai, 200233, China
| | - Zi-Hao Lin
- Department of Orthopedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 600 Yishan Road, Shanghai, 200233, China
| | - Zi-Yi Wang
- Department of Orthopedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 600 Yishan Road, Shanghai, 200233, China
| | - Hao Peng
- Department of Orthopedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 600 Yishan Road, Shanghai, 200233, China
| | - Ruo-Tao Liu
- Department of Orthopedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 600 Yishan Road, Shanghai, 200233, China
| | - Zhi-Chao Hu
- Department of Orthopedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 600 Yishan Road, Shanghai, 200233, China
| | - Yao-Hua He
- Department of Orthopedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 600 Yishan Road, Shanghai, 200233, China
| | - Xiao-Juan Wei
- Institute of Microsurgery on Extremities, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 600 Yishan Road, Shanghai, 200233, China.
| | - Chang-Qing Zhang
- Department of Orthopedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 600 Yishan Road, Shanghai, 200233, China
| | - Yong Feng
- Department of Orthopedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 600 Yishan Road, Shanghai, 200233, China.
| | - Qian Tang
- Department of Orthopedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 600 Yishan Road, Shanghai, 200233, China.
| | - Zhen-Zhong Zhu
- Department of Orthopedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 600 Yishan Road, Shanghai, 200233, China.
| |
Collapse
|
6
|
Kumar SP, Nadendla EK, Malireddi RKS, Haque SA, Mall R, Neuwald AF, Kanneganti TD. Evolutionary and Functional Analysis of Caspase-8 and ASC Interactions to Drive Lytic Cell Death, PANoptosis. Mol Biol Evol 2025; 42:msaf096. [PMID: 40277230 PMCID: PMC12066828 DOI: 10.1093/molbev/msaf096] [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: 12/09/2024] [Revised: 04/09/2025] [Accepted: 04/15/2025] [Indexed: 04/26/2025] Open
Abstract
Caspases are evolutionarily conserved proteins essential for driving cell death in development and host defense. Caspase-8, a key member of the caspase family, is implicated in nonlytic apoptosis, as well as lytic forms of cell death. Recently, caspase-8 has been identified as an integral component of PANoptosomes, multiprotein complexes formed in response to innate immune sensor activation. Several innate immune sensors can nucleate caspase-8-containing PANoptosome complexes to drive inflammatory lytic cell death, PANoptosis. However, how the evolutionarily conserved and diverse functions of caspase-8 drive PANoptosis remains unclear. To address this, we performed evolutionary, sequence, structural, and functional analyses to decode caspase-8's complex-forming abilities and its interaction with the PANoptosome adaptor ASC. Our study distinguished distinct subgroups within the death domain superfamily based on their evolutionary and functional relationships, identified homotypic traits among subfamily members, and captured key events in caspase evolution. We also identified critical residues defining the heterotypic interaction between caspase-8's death effector domain and ASC's pyrin domain, validated through cross-species analyses, dynamic simulations, and in vitro experiments. Overall, our study elucidated recent evolutionary adaptations of caspase-8 that allowed it to interact with ASC, improving our understanding of critical molecular associations in PANoptosome complex formation and the underlying PANoptotic responses in host defense and inflammation. These findings have implications for understanding mammalian immune responses and developing new therapeutic strategies for inflammatory diseases.
Collapse
Affiliation(s)
- Sivakumar Prasanth Kumar
- Department of Immunology, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN 38105, USA
| | - Eswar Kumar Nadendla
- Department of Immunology, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN 38105, USA
| | - R K Subbarao Malireddi
- Department of Immunology, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN 38105, USA
| | - Syed Asfarul Haque
- Cryo-Electron Microscopy Center, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN 38105, USA
| | - Raghvendra Mall
- Department of Immunology, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN 38105, USA
| | - Andrew F Neuwald
- Institute for Genome Sciences and Department of Biochemistry and Molecular Biology, University of Maryland School of Medicine, 670 W. Baltimore Street, Baltimore, MD 21201, USA
| | - Thirumala-Devi Kanneganti
- Department of Immunology, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN 38105, USA
| |
Collapse
|
7
|
Jin X, Zhu Y, Xing L, Ding X, Liu Z. PANoptosis: a potential target of atherosclerotic cardiovascular disease. Apoptosis 2025:10.1007/s10495-025-02089-x. [PMID: 40285923 DOI: 10.1007/s10495-025-02089-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/05/2025] [Indexed: 04/29/2025]
Abstract
PANoptosis is a newly discovered cell death pathway triggered by the innate immunizer, which in turn promotes the assembly of the PANoptosome and activates downstream effectors. As a special cell death mode, it is characterized by apoptosis, pyroptosis, and necroptosis at the same time; therefore, it is not feasible to inhibit PANoptosis by suppressing a single cell death pathway. However, active ingredients targeting the PANoptosome can effectively inhibit PANoptosis.Given the importance of cell death in disease, targeting PANoptosis would be an important therapeutic tool. Previous studies have focused more on infectious diseases and cancer, and the role of PANoptosis in the cardiovascular field has not been comprehensively addressed. While ASCVD is the number one killer of cardiovascular diseases, it is important to explore new targets to determine future research directions. Therefore, this review focuses on the assembly of PANoptosome, the molecular mechanism of PANoptosis, and the related mechanisms of PANoptosis leading to ASCVD such as myocardial infarction, ischemic cardiomyopathy and ischemic stroke, in order to provide a new perspective for the prevention and treatment of ASCVD.
Collapse
Affiliation(s)
- Xiao Jin
- Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200062, China
| | - Yanan Zhu
- Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200062, China
| | - Lina Xing
- Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200062, China
| | - Xinyue Ding
- Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200062, China
| | - Zongjun Liu
- Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200062, China.
- , No. 164, Lanxi Road, Putuo District, Shanghai, China.
| |
Collapse
|
8
|
Barthez M, Xue B, Zheng J, Wang Y, Song Z, Mu WC, Wang CL, Guo J, Yang F, Ma Y, Wei X, Ye C, Sims N, Martinez-Sobrido L, Perlman S, Chen D. SIRT2 suppresses aging-associated cGAS activation and protects aged mice from severe COVID-19. Cell Rep 2025; 44:115562. [PMID: 40220296 PMCID: PMC12074670 DOI: 10.1016/j.celrep.2025.115562] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2023] [Revised: 04/11/2024] [Accepted: 03/24/2025] [Indexed: 04/14/2025] Open
Abstract
Aging-associated vulnerability to coronavirus disease 2019 (COVID-19) remains poorly understood. Here, we show that severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2)-infected aged mice lacking SIRT2, a cytosolic NAD+-dependent deacetylase, develop more severe disease and show increased mortality, while treatment with an NAD+ booster, 78c, protects aged mice from lethal infection. Mechanistically, we demonstrate that SIRT2 modulates the acetylation of cyclic GMP-AMP synthase (cGAS), an immune sensor for cytosolic DNA, and suppresses aging-associated cGAS activation and inflammation. Furthermore, we show that SARS-CoV-2 infection-induced inflammation is mediated at least in part by ORF3a, which triggers mtDNA release and cGAS activation. Collectively, our study reveals a molecular basis for aging-associated susceptibility to COVID-19 and suggests therapeutic approaches to protect aged populations from severe SARS-CoV-2 infection.
Collapse
Affiliation(s)
- Marine Barthez
- Department of Nutritional Sciences and Toxicology, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Biyun Xue
- Department of Microbiology and Immunology, University of Iowa, Iowa City, IA, USA
| | - Jian Zheng
- Department of Microbiology and Immunology, Center for Predictive Medicine, University of Louisville, Louisville, KY, USA
| | - Yifei Wang
- Department of Nutritional Sciences and Toxicology, University of California, Berkeley, Berkeley, CA 94720, USA; Metabolic Biology Graduate Program, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Zehan Song
- Department of Nutritional Sciences and Toxicology, University of California, Berkeley, Berkeley, CA 94720, USA; Metabolic Biology Graduate Program, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Wei-Chieh Mu
- Department of Nutritional Sciences and Toxicology, University of California, Berkeley, Berkeley, CA 94720, USA; Endocrinology Graduate Program, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Chih-Ling Wang
- Department of Nutritional Sciences and Toxicology, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Jiayue Guo
- Department of Nutritional Sciences and Toxicology, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Fanghan Yang
- Department of Nutritional Sciences and Toxicology, University of California, Berkeley, Berkeley, CA 94720, USA; Endocrinology Graduate Program, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Yuze Ma
- Department of Nutritional Sciences and Toxicology, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Xuetong Wei
- Department of Nutritional Sciences and Toxicology, University of California, Berkeley, Berkeley, CA 94720, USA; Metabolic Biology Graduate Program, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Chengjin Ye
- Texas Biomedical Research Institute, San Antonio, TX, USA
| | - Nicholas Sims
- Department of Nutritional Sciences and Toxicology, University of California, Berkeley, Berkeley, CA 94720, USA
| | | | - Stanley Perlman
- Department of Microbiology and Immunology, University of Iowa, Iowa City, IA, USA; Department of Pediatrics, University of Iowa, Iowa City, IA, USA.
| | - Danica Chen
- Department of Nutritional Sciences and Toxicology, University of California, Berkeley, Berkeley, CA 94720, USA; Metabolic Biology Graduate Program, University of California, Berkeley, Berkeley, CA 94720, USA; Endocrinology Graduate Program, University of California, Berkeley, Berkeley, CA 94720, USA.
| |
Collapse
|
9
|
Chen W, Jiang Y, Zeng J, Liu D, Feng X, Cheng Y, Lu D, Sun Y, Zhu Q, Zhang X, Wang Q. FDX1 promotes elesclomol-induced PANoptosis in diffuse large B-cell lymphoma via activating IRF3/IFN-β signaling. Oncogene 2025:10.1038/s41388-025-03366-4. [PMID: 40240522 DOI: 10.1038/s41388-025-03366-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2024] [Revised: 03/02/2025] [Accepted: 03/20/2025] [Indexed: 04/18/2025]
Abstract
Diffuse large B-cell lymphoma (DLBCL) remains a major clinical challenge and requires the development of new therapeutic approaches. The identification of cuproptosis, a newly defined form of copper-induced cell death, has provided innovative insights for cancer therapy. Here, we report that loss of the mitochondrial matrix reductase FDX1 in DLBCL cells impairs the antitumor effect of elesclomol (ES), which performs its function by transporting excess copper into cells. Overexpressing (OE) FDX1 significantly sensitized DLBCL cells to ES-induced cell death in vitro and enhanced the anticancer activity of ES in vivo. Furthermore, treatment with ES in FDX1-high expression patient-derived xenograft (PDX) showed a significantly greater inhibitory effect than in FDX1-low expression PDX. Mechanistically, FDX1 promotes the induction of IFN-β-dependent PANoptosis by increasing IRF3 phosphorylation in DLBCL cells upon ES treatment. Consistent with this finding, patient cohort analysis revealed that FDX1 expression correlated positively with enhanced IRF3 phosphorylation. Together, our findings are the first to identify the central role of FDX1 in synergizing with ES to activate IFN-β signaling and induce PANoptosis. This study enables us to re-explore the clinical anticancer potential of ES as a novel therapeutic strategy for DLBCL.
Collapse
Affiliation(s)
- Weifeng Chen
- Qingyuan People's Hospital, The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan, 511518, China
- Affiliated Cancer Hospital/Institute and GMU-GIBH Joint School of Life Sciences of Guangzhou Medical University, State Key Laboratory of Respiratory Disease, Guangzhou, 511495, China
| | - Yuhang Jiang
- Department of Orthopedics, The Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, 518033, China
| | - Jun Zeng
- Qingyuan People's Hospital, The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan, 511518, China
| | - Dandan Liu
- Affiliated Cancer Hospital/Institute and GMU-GIBH Joint School of Life Sciences of Guangzhou Medical University, State Key Laboratory of Respiratory Disease, Guangzhou, 511495, China
| | - Xiaoting Feng
- Affiliated Cancer Hospital/Institute and GMU-GIBH Joint School of Life Sciences of Guangzhou Medical University, State Key Laboratory of Respiratory Disease, Guangzhou, 511495, China
| | - Yiqiu Cheng
- Qingyuan People's Hospital, The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan, 511518, China
| | - Di Lu
- Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510623, China
| | - Yangbai Sun
- Department of Musculoskeletal Oncology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China.
| | - Qinyuan Zhu
- Department of Dermatology, Huashan Hospital, Fudan University, Shanghai, 311599, China.
| | - Xiaoren Zhang
- Affiliated Cancer Hospital/Institute and GMU-GIBH Joint School of Life Sciences of Guangzhou Medical University, State Key Laboratory of Respiratory Disease, Guangzhou, 511495, China.
| | - Qi Wang
- Qingyuan People's Hospital, The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan, 511518, China.
| |
Collapse
|
10
|
An H, Shao C, He Y, Zhou H, Wang T, Xu G, Yang J, Wan H. Calycosin Inhibit PANoptosis and Alleviate Brain Damage: A Bioinformatics and Experimental Verification Approach. ACS Chem Neurosci 2025; 16:1550-1564. [PMID: 40156525 DOI: 10.1021/acschemneuro.5c00072] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/01/2025] Open
Abstract
PANoptosis is a newly identified form of cell death that encompasses pyroptosis, apoptosis, and necroptosis. Numerous studies have highlighted the significance of PANoptosis in brain ischemia-reperfusion (I/R) injury. Calycosin, a natural product with diverse biological activities, has demonstrated a significant reduction in neuronal death caused by ischemic brain injury by modulating multiple cell death pathways. In order to investigate the potential mechanisms underlying the neuroprotective role of calycosin in alleviating PANoptosis-induced damage in ischemic stroke therapy, we used mouse hippocampal neuronal cell line HT22 to stimulate ischemia in vitro through Oxygen and Glucose Deprivation/Reperfusion (OGD/R) and established molecular docking to assess the binding affinity of Calycosin with key targets and molecular dynamics simulations (MDS) to study the stability of the ligand-protein complex. The results demonstrate that Calycosin could improve the cell growth of HT22, leading to enhanced cell viability, reduced lactate dehydrogenase leakage, and decreased cell apoptosis after OGD/R. It also regulated the expression of PANoptosis-related genes such as NLRP3, GSDMD, MLKL, and RIPK1 and increased the Bcl-2/Bax ratio, effectively reducing cellular damage and providing protection. Molecular docking and MDS simulations demonstrated strong binding activity and stability between Calycosin and PANoptosis-related targets. Furthermore, Calycosin successfully passed the drug similarity (DS) evaluation and exhibited favorable absorption, distribution, metabolism, excretion, and toxicity (ADMET) properties and biological activity. In conclusion, Calycosin could alleviate ischemic stroke by inhibiting PANoptosis, reducing neuronal inflammation and apoptosis, and improving damage caused by the OGD/R. Thus, it could serve as a potential therapy for ischemic stroke.
Collapse
Affiliation(s)
- Huiyan An
- School of Life Science, Zhejiang Chinese Medical University, Hangzhou 310053, Zhejiang, China
| | - Chongyu Shao
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, Zhejiang, China
| | - Yu He
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, Zhejiang, China
| | - Huifen Zhou
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, Zhejiang, China
| | - Ting Wang
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, Zhejiang, China
| | - Guanfeng Xu
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, Zhejiang, China
| | - Jiehong Yang
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, Zhejiang, China
| | - Haitong Wan
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, Zhejiang, China
- School of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou 450046, Henan, China
| |
Collapse
|
11
|
Li J, Jia YC, Lu J, Zhang H, Wang Z, Xie X, Cao F, Li F. Inhibition of Zbp1-PANoptosome-mediated PANoptosis effectively attenuates acute pancreatitis. Cell Death Discov 2025; 11:180. [PMID: 40240343 PMCID: PMC12003674 DOI: 10.1038/s41420-025-02451-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2024] [Revised: 03/21/2025] [Accepted: 03/27/2025] [Indexed: 04/18/2025] Open
Abstract
Early acute pancreatitis is an acute inflammatory disease that involves multiple modes of cell death, including apoptosis, necrotic apoptosis, and pyroptosis in its disease process. PANoptosis, a type of cell death that includes pyroptosis, apoptosis, and necroptosis, has had an important role in a variety of infectious and inflammatory diseases in recent years. To judge the relationship between PANoptosis and AP, we first analyzed the data from pancreatic transcriptome data by bioinformatics techniques, and we found the enrichment of PANoptosis pathway in AP. Next, we screened the genes and identified differentially expressed genes (DEGs) associated with AP and PANoptosis. Finally, we found that Zbp1 may have a major role in the process of PANoptosis. For this purpose, we constructed AP models in mice and in vitro cell line 266-6 and intervened by inhibiting Zbp1. The final results showed that the PANoptosis in mice was significantly suppressed after inhibition of Zbp1. In conclusion, inflammatory injury in AP can be significantly improved by inhibiting Zbp1- PANoptosome-mediated PANoptosis.
Collapse
Affiliation(s)
- Jie Li
- Department of General Surgery, Xuanwu Hospital, Capital Medical University, Beijing, China
- Clinical Center for Acute Pancreatitis, Capital Medical University, Beijing, China
| | - Yu-Chen Jia
- Department of General Surgery, Xuanwu Hospital, Capital Medical University, Beijing, China
- Clinical Center for Acute Pancreatitis, Capital Medical University, Beijing, China
| | - Jiongdi Lu
- Department of General Surgery, Xuanwu Hospital, Capital Medical University, Beijing, China
- Clinical Center for Acute Pancreatitis, Capital Medical University, Beijing, China
| | - Haoyu Zhang
- Department of General Surgery, Xuanwu Hospital, Capital Medical University, Beijing, China
- Clinical Center for Acute Pancreatitis, Capital Medical University, Beijing, China
| | - Zheng Wang
- Department of General Surgery, Xuanwu Hospital, Capital Medical University, Beijing, China
- Clinical Center for Acute Pancreatitis, Capital Medical University, Beijing, China
| | - Xiaozhou Xie
- Department of General Surgery, Xuanwu Hospital, Capital Medical University, Beijing, China
- Clinical Center for Acute Pancreatitis, Capital Medical University, Beijing, China
| | - Feng Cao
- Department of General Surgery, Xuanwu Hospital, Capital Medical University, Beijing, China
- Clinical Center for Acute Pancreatitis, Capital Medical University, Beijing, China
| | - Fei Li
- Department of General Surgery, Xuanwu Hospital, Capital Medical University, Beijing, China.
- Clinical Center for Acute Pancreatitis, Capital Medical University, Beijing, China.
| |
Collapse
|
12
|
Wu J, Huang H, Xu W, Cui B, Sun P, Hao X, Jiang S, Hou X, Qi X, Wei Z, Cheng Y, Zheng Y, Liu K, He J. Inflammation-driven biomimetic nano-polyphenol drug delivery system alleviates severe acute pancreatitis by inhibiting macrophage PANoptosis and pancreatic enzymes oversecretion. J Adv Res 2025:S2090-1232(25)00225-5. [PMID: 40210149 DOI: 10.1016/j.jare.2025.04.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2024] [Revised: 03/22/2025] [Accepted: 04/02/2025] [Indexed: 04/12/2025] Open
Abstract
INTRODUCTION Severe acute pancreatitis (SAP) is a critical inflammatory disease with high morbidity and mortality. Current treatments focused on symptomatic relief but failed to prevent inflammation progression in cellular level. OBJECTIVES In order to develop an SAP-targeting drug delivery system to alleviate SAP in cellular level and illustrate its mechanism, we explored the use of proanthocyanidin (PYD) and pentoxifylline (PTX) loaded into macrophage membrane-coated self-assembly nanoparticles (FePTX@CM NPs) for targeted SAP treatment. The combination application of these two drugs was innovative in SAP aid. METHODS We developed the NPs by self-assembly strategy and cell membrane coating. Its particle size and zeta potential were measured by dynamic light scatter (DLS). The morphology of the NPs was observed by transmission electron microscopy (TEM). And the encapsulation efficiency was evaluated by nano-flow cytometry. The total protein profile was determined via Coomassie brilliant blue. We explore the mechanism of our NPs against SAP in cellular and animal levels. Bioinformatics approaches, TEM, immunofluorescent assay and co-immunoprecipitation were performed to comprehensively explain the specific anti-SAP mechanism of FePTX@CM NPs. RESULTS After inflammation-driven targeting, PYD in the NPs inhibited pancreatic amylase and lipase release by suppressing mitochondrial reactive oxygen species (mtROS)/Golgi stress, while PTX prevented SAP-associated macrophage PANoptosis by inhibiting Zbp1 signal pathway. The protection effect of these biomimetic NPs worked from different aspects to alleviate SAP symptoms and inflammation progression in relative cells. CONCLUSION The FePTX@CM NPs demonstrated effective pancreas targeting, reduced systemic inflammation especially pro-inflammatory cell recruitment and activation, and minimized tissue damage in SAP mouse models, offering a promising therapeutic strategy for clinical SAP management.
Collapse
Affiliation(s)
- Junyong Wu
- Department of Pharmacy, The Second Xiangya Hospital Central South University Changsha 410011, China; Institute of Clinical Pharmacy Central South University Changsha 410011, China
| | - Hai Huang
- Department of Pharmacy, The Second Xiangya Hospital Central South University Changsha 410011, China; Institute of Clinical Pharmacy Central South University Changsha 410011, China
| | - Wenjie Xu
- Department of Pharmacy, The Second Xiangya Hospital Central South University Changsha 410011, China; Institute of Clinical Pharmacy Central South University Changsha 410011, China
| | - Beibei Cui
- Department of General Surgery, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China
| | - Pengcheng Sun
- Department of Pharmacy, The Second Xiangya Hospital Central South University Changsha 410011, China; Institute of Clinical Pharmacy Central South University Changsha 410011, China
| | - XinYan Hao
- Department of Pharmacy, The Second Xiangya Hospital Central South University Changsha 410011, China; Institute of Clinical Pharmacy Central South University Changsha 410011, China
| | - Shihe Jiang
- Department of General Surgery, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China
| | - Xuyang Hou
- Department of General Surgery, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China
| | - Xiaoyan Qi
- Department of General Surgery, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China
| | - Zuxing Wei
- Department of General Surgery, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China
| | - Yimiao Cheng
- Department of General Surgery, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China
| | - Yanwen Zheng
- Department of General Surgery, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China
| | - Kuijie Liu
- Department of General Surgery, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China.
| | - Jun He
- Department of General Surgery, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China.
| |
Collapse
|
13
|
Lee C, Kim JE, Cha YE, Moon JH, Kim ER, Chang DK, Kim YH, Hong SN. IFN-γ-Induced intestinal epithelial cell-type-specific programmed cell death: PANoptosis and its modulation in Crohn's disease. Front Immunol 2025; 16:1523984. [PMID: 40230837 PMCID: PMC11994596 DOI: 10.3389/fimmu.2025.1523984] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2024] [Accepted: 03/04/2025] [Indexed: 04/16/2025] Open
Abstract
Background Crohn's disease (CD) is a chronic inflammatory bowel disease (IBD) and is considered a Th1-mediated disease, supported by the over-expression of interferon-gamma (IFN-γ) in the intestinal lamina propria. IFN-γ has a pleiotropic effect on the intestinal epithelial cells (IECs), suggesting that IFN-γ-induced responses may differ between epithelial cell types. Methods We established human small intestinal organoids (enteroids) derived from non-IBD controls and CD patients. Using human enteroids, the major response of IECs induced by IFN-γ was evaluated, focusing on the IFN-γ-induced programmed cell death (PCD) pathway. Identified IFN-γ-induced responses were validated in surgically resected intestinal samples and publicly available single-cell RNA-sequencing datasets. Results IFN-γ stimulated programmed cell death (PCD) of IECs in both control and CD enteroids in a dose-dependent manner. Pyroptosis, apoptosis. and necroptosis were activated in enteroids, suggesting that PANoptosis was the main process of IFN-γ-induced PCD in IECs. The response to IFN-γ depends on the cell type of the IECs. IFN-γ induced depletion of enterocytes with upregulation of PANoptosis-associated genes, while leading to expansion of goblet cells without significant change in PANoptosis-associated gene expression. Individual PCD inhibitors were insufficient to block IFN-γ-induced cytotoxicity, whereas the selective JAK1 inhibitor (upadacitinib) effectively blocked IFN-γ-induced cytotoxicity and PANoptosis. Furthermore, PANoptosis was significantly activated in surgically resected tissues and in publicly available single-cell RNA-sequencing datasets of intestinal tissues from patients with CD. Conclusion IFN-γ induces PANoptosis in enterocytes, which can be treated with a selective JAK1 inhibitor in patients with CD.
Collapse
Affiliation(s)
- Chansu Lee
- Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
- Stem Cell and Regenerative Medicine Center, Samsung Medical Center, Seoul, Republic of Korea
| | - Ji Eun Kim
- Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Yeo-Eun Cha
- Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
- Stem Cell and Regenerative Medicine Center, Samsung Medical Center, Seoul, Republic of Korea
| | - Ji Hwan Moon
- Samsung Genome Institute, Samsung Medical Center, Seoul, Republic of Korea
| | - Eun Ran Kim
- Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Dong Kyung Chang
- Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Young-Ho Kim
- Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Sung Noh Hong
- Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
- Stem Cell and Regenerative Medicine Center, Samsung Medical Center, Seoul, Republic of Korea
| |
Collapse
|
14
|
Xiao J, Wang L, Zhang B, Hou A. Cell death in acute lung injury: caspase-regulated apoptosis, pyroptosis, necroptosis, and PANoptosis. Front Pharmacol 2025; 16:1559659. [PMID: 40191423 PMCID: PMC11968751 DOI: 10.3389/fphar.2025.1559659] [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: 01/13/2025] [Accepted: 03/03/2025] [Indexed: 04/09/2025] Open
Abstract
There has been abundant research on the variety of programmed cell death pathways. Apoptosis, pyroptosis, and necroptosis under the action of the caspase family are essential for the innate immune response. Caspases are classified into inflammatory caspase-1/4/5/11, apoptotic caspase-3/6/7, and caspase-2/8/9/10. Although necroptosis is not caspase-dependent to transmit cell death signals, it can cross-link with pyroptosis and apoptosis signals under the regulation of caspase-8. An increasing number of studies have reiterated the involvement of the caspase family in acute lung injuries caused by bacterial and viral infections, blood transfusion, and ventilation, which is influenced by noxious stimuli that activate or inhibit caspase engagement pathways, leading to subsequent lung injury. This article reviews the role of caspases implicated in diverse programmed cell death mechanisms in acute lung injury and the status of research on relevant inhibitors against essential target proteins of the described cell death mechanisms. The findings of this review may help in delineating novel therapeutic targets for acute lung injury.
Collapse
Affiliation(s)
| | | | | | - Ana Hou
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang, China
| |
Collapse
|
15
|
Chen Z, Behrendt R, Wild L, Schlee M, Bode C. Cytosolic nucleic acid sensing as driver of critical illness: mechanisms and advances in therapy. Signal Transduct Target Ther 2025; 10:90. [PMID: 40102400 PMCID: PMC11920230 DOI: 10.1038/s41392-025-02174-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Revised: 01/14/2025] [Accepted: 02/11/2025] [Indexed: 03/20/2025] Open
Abstract
Nucleic acids from both self- and non-self-sources act as vital danger signals that trigger immune responses. Critical illnesses such as acute respiratory distress syndrome, sepsis, trauma and ischemia lead to the aberrant cytosolic accumulation and massive release of nucleic acids that are detected by antiviral innate immune receptors in the endosome or cytosol. Activation of receptors for deoxyribonucleic acids and ribonucleic acids triggers inflammation, a major contributor to morbidity and mortality in critically ill patients. In the past decade, there has been growing recognition of the therapeutic potential of targeting nucleic acid sensing in critical care. This review summarizes current knowledge of nucleic acid sensing in acute respiratory distress syndrome, sepsis, trauma and ischemia. Given the extensive research on nucleic acid sensing in common pathological conditions like cancer, autoimmune disorders, metabolic disorders and aging, we provide a comprehensive summary of nucleic acid sensing beyond critical illness to offer insights that may inform its role in critical conditions. Additionally, we discuss potential therapeutic strategies that specifically target nucleic acid sensing. By examining nucleic acid sources, sensor activation and function, as well as the impact of regulating these pathways across various acute diseases, we highlight the driving role of nucleic acid sensing in critical illness.
Collapse
Affiliation(s)
- Zhaorong Chen
- Department of Anesthesiology and Intensive Care Medicine, University Hospital Bonn, 53127, Bonn, Germany
| | - Rayk Behrendt
- Institute of Clinical Chemistry and Clinical Pharmacology, University Hospital Bonn, 53127, Bonn, Germany
| | - Lennart Wild
- Department of Anesthesiology and Intensive Care Medicine, University Hospital Bonn, 53127, Bonn, Germany
| | - Martin Schlee
- Institute of Clinical Chemistry and Clinical Pharmacology, University Hospital Bonn, 53127, Bonn, Germany
| | - Christian Bode
- Department of Anesthesiology and Intensive Care Medicine, University Hospital Bonn, 53127, Bonn, Germany.
| |
Collapse
|
16
|
Williams EP, Xue Y, Vogel P, Yang D, Ponce-Flores A, Li X, Ogorek TJ, Saini M, Iulek J, Ruiz FX, Arnold E, Golden JE, Meibohm B, Jonsson CB. The antiviral BDGR-49 provides protection from lethal, neurotropic Venezuelan equine encephalitis virus intranasal infection in mice. J Virol 2025; 99:e0167924. [PMID: 39936916 PMCID: PMC11916738 DOI: 10.1128/jvi.01679-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2024] [Accepted: 12/13/2024] [Indexed: 02/13/2025] Open
Abstract
Venezuelan, western, and eastern equine encephalitis virus (VEEV, WEEV, and EEEV) cause a febrile illness that may result in fatal neurological disease in humans and equines. Human infections are typically from mosquito bites, although cases from respiratory exposure in laboratory accidents have been documented. In addition to natural mosquito-borne infection, the potential biothreat inherent in the ability to disseminate these viruses via the respiratory route has driven the development of antiviral drugs for this route of exposure. To address this gap, we tested the prophylactic administration of a novel brain-penetrant, antiviral, BDGR-49, against a lethal intranasal challenge of VEEV, WEEV, or EEEV in BALB/c mouse model. BDGR-49 conferred 100% protection with 6 mg kg-1 twice per day for 6 days for VEEV, but not EEEV or WEEV. By 8 days post-infection (dpi), infectious virus, viral RNA, and viral antigen in the brain of BDGR-49-treated mice were significantly reduced. Brains of VEEV TrD-infected, BDGR-49-treated mice showed a significant reduction in the expression of genes associated with inflammation (IFNB1, TNF, IL6, and CCL5) and cell death (CASP4, GSDMD, PYCARD, and ZBP1). At dpi 14, histopathology showed that neuronal lesions and inflammatory cell infiltrates were essentially absent, and viral antigen was not detected in the brains of VEEV TrD-infected, BDGR-49-treated mice. In summary, although BDGR-49 treatment showed significant promise for the treatment of mice exposed intranasally to VEEV, the more rapid and efficient entry of EEEV and WEEV by this route into the central nervous system will require additional optimization of the dosing regimen.IMPORTANCEProphylactic and therapeutic treatment of viruses that cause encephalitis requires fast-acting drugs that rapidly penetrate the blood-brain barrier. Currently, clinicians have only a limited set of antivirals for the treatment of neurotropic infections such as herpesviruses or HIV-1, and none for alphaviruses, and treatment outcomes remain poor. New medical countermeasures will address the gap in treatment of viral encephalitis such as those caused by the neurotropic alphaviruses and others.
Collapse
Affiliation(s)
- Evan P. Williams
- Department of Microbiology, Immunology and Biochemistry, College of Medicine, The University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | - Yi Xue
- Department of Microbiology, Immunology and Biochemistry, College of Medicine, The University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | - Peter Vogel
- Animal Resources Center and Veterinary Pathology Core, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - Dong Yang
- Regional Biocontainment Laboratory, The University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | - Alejandro Ponce-Flores
- Department of Microbiology, Immunology and Biochemistry, College of Medicine, The University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | - Xiaoyu Li
- Division of Pharmaceutical Sciences, School of Pharmacy, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Tyler J. Ogorek
- Department of Chemistry, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Manisha Saini
- Center for Advanced Biotechnology and Medicine, Rutgers The State University of New Jersey, Piscataway, New Jersey, USA
- Department of Chemistry and Chemical Biology, Rutgers The State University of New Jersey, Piscataway, New Jersey, USA
| | - Jorge Iulek
- Department of Chemistry, State University of Ponta Grossa, Ponta Grossa, Brazil
| | - Francesc Xavier Ruiz
- Center for Advanced Biotechnology and Medicine, Rutgers The State University of New Jersey, Piscataway, New Jersey, USA
- Department of Chemistry and Chemical Biology, Rutgers The State University of New Jersey, Piscataway, New Jersey, USA
| | - Eddy Arnold
- Center for Advanced Biotechnology and Medicine, Rutgers The State University of New Jersey, Piscataway, New Jersey, USA
- Department of Chemistry and Chemical Biology, Rutgers The State University of New Jersey, Piscataway, New Jersey, USA
| | - Jennifer E. Golden
- Division of Pharmaceutical Sciences, School of Pharmacy, University of Wisconsin-Madison, Madison, Wisconsin, USA
- Department of Chemistry, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Bernd Meibohm
- Department of Pharmaceutical Sciences, College of Pharmacy, The University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | - Colleen B. Jonsson
- Department of Microbiology, Immunology and Biochemistry, College of Medicine, The University of Tennessee Health Science Center, Memphis, Tennessee, USA
- Regional Biocontainment Laboratory, The University of Tennessee Health Science Center, Memphis, Tennessee, USA
- Department of Pharmaceutical Sciences, College of Pharmacy, The University of Tennessee Health Science Center, Memphis, Tennessee, USA
- Institute for the Study of Host-Pathogen Systems, The University of Tennessee Health Science Center, Memphis, Tennessee, USA
| |
Collapse
|
17
|
Qin Q, Chen W, King CD, Kumar SP, Vogel P, Tweedell RE, Kanneganti TD. The critical role of the ZBP1-NINJ1 axis and IRF1/IRF9 in ethanol-induced cell death, PANoptosis, and alcohol-associated liver disease. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.12.642836. [PMID: 40161842 PMCID: PMC11952398 DOI: 10.1101/2025.03.12.642836] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
Innate immunity provides the critical first line of defense against infection and sterile triggers. Cell death is a key component of the innate immune response to clear pathogens, but excessive or aberrant cell death can induce inflammation, cytokine storm, and pathology, making it a central molecular mechanism in inflammatory diseases. Alcohol-associated liver disease (ALD) is one such inflammatory disease, but the specific innate immune mechanisms driving pathology in this context remain unclear. Here, by leveraging RNAseq and tissue expression in clinical samples, we identified increased expression of the innate immune sensor Z-DNA binding protein (ZBP1) in patients with ALD. We discovered that ZBP1 expression correlated with ALD progression in patients, and that ethanol induced ZBP1-dependent lytic cell death, PANoptosis, in immune (macrophages, monocytes, Kupffer cells) and non-immune cells (hepatocytes). Mechanistically, the interferon regulatory factors (IRFs) IRF9 and IRF1 upregulated ZBP1 expression, allowing ZBP1 to sense Z-NAs through its Zα2 domain and drive PANoptosis signaling, cell membrane rupture through NINJ1, and DAMP release. Furthermore, the expressions of ZBP1 and NINJ1 were upregulated in both liver and serum samples from patients with ALD. In mouse models of chronic and acute ALD, ZBP1-deficient mice were significantly protected from disease pathology and liver damage. Overall, our findings establish the critical role of the ZBP1-NINJ1 axis regulated by IRFs in driving inflammatory cell death, PANoptosis, in liver cells, suggesting that targeting these molecules will have therapeutic potential in ALD and other inflammatory conditions.
Collapse
Affiliation(s)
- Qiang Qin
- Department of Immunology, St. Jude Children's Research Hospital; Memphis, TN 38105, USA
| | - Wen Chen
- Department of Immunology, St. Jude Children's Research Hospital; Memphis, TN 38105, USA
| | - Clay D. King
- University of Kansas Medical Center, The University of Kansas; Kansas City, KS 66045, USA
| | | | - Peter Vogel
- Animal Resources Center and the Veterinary Pathology Core, St. Jude Children’s Research Hospital; Memphis, TN 38105, USA
| | - Rebecca E. Tweedell
- Department of Immunology, St. Jude Children's Research Hospital; Memphis, TN 38105, USA
| | | |
Collapse
|
18
|
Evdokimova M, Feng S, Caobi A, Moreira FR, Jones D, Alysandratos KD, Tully ES, Kotton DN, Boyd DF, Banach BS, Kirchdoerfer RN, Saeed M, Baker SC. Coronavirus endoribonuclease antagonizes ZBP1-mediated necroptosis and delays multiple cell death pathways. Proc Natl Acad Sci U S A 2025; 122:e2419620122. [PMID: 40035769 PMCID: PMC11912388 DOI: 10.1073/pnas.2419620122] [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: 09/30/2024] [Accepted: 01/13/2025] [Indexed: 03/06/2025] Open
Abstract
Identifying conserved mechanisms used by viruses to delay host innate responses can reveal potential targets for antiviral therapeutics. Here, we investigated coronavirus nonstructural protein 15 (nsp15), which encodes a highly conserved endoribonuclease (EndoU). EndoU functions as an immune antagonist by limiting the accumulation of viral replication intermediates that would otherwise be sensed by the host. Despite being a promising antiviral target, it has been difficult to develop small-molecule inhibitors that target the EndoU active site. We generated nsp15 mutants of the coronaviruses severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and mouse hepatitis virus (MHV)-A59 and identified conserved residues within the amino-terminal domain that are required for EndoU activity. Loss of EndoU activity caused the activation of host sensors, which limited viral replication in interferon-responsive cells and attenuated disease in MHV-infected mice. Using transcriptional profiling, we found that MHV EndoU mutant viruses upregulate multiple host sensors, including Z-form nucleic acid-binding protein 1 (ZBP1). We found that nsp15 mutants induced early, robust ZBP1-mediated necroptosis. EndoU mutant viruses also induced ZBP1-independent apoptosis and pyroptosis pathways, causing early, robust cell death that limits virus replication and pathogenesis. Overall, we document the importance of the amino-terminal domain for EndoU function. We also highlight the importance of nsp15/EndoU activity for evading host sensors, delaying cell death, and promoting pathogenesis.
Collapse
Affiliation(s)
- Monika Evdokimova
- Department of Microbiology and Immunology, Stritch School of Medicine, Loyola University Chicago, Maywood, IL60153
| | - Shuchen Feng
- Department of Microbiology and Immunology, Stritch School of Medicine, Loyola University Chicago, Maywood, IL60153
| | - Allen Caobi
- Department of Biochemistry and Cell Biology, Boston University Chobanian and Avedisian School of Medicine, Boston University, Boston, MA02118
- National Emerging Infectious Diseases Laboratories, Boston University, Boston, MA02118
| | - Fernando R. Moreira
- Department of Microbiology and Immunology, Stritch School of Medicine, Loyola University Chicago, Maywood, IL60153
| | - Dakota Jones
- Center for Regenerative Medicine, Boston University and Boston Medical Center, Boston, MA02118
- The Pulmonary Center and Department of Medicine, Boston University Chobanian and Avedisian School of Medicine, Boston, MA02118
| | - Konstantinos-Dionysios Alysandratos
- Center for Regenerative Medicine, Boston University and Boston Medical Center, Boston, MA02118
- The Pulmonary Center and Department of Medicine, Boston University Chobanian and Avedisian School of Medicine, Boston, MA02118
| | - Ena S. Tully
- Department of Biochemistry, College of Agricultural and Life Sciences, University of Wisconsin-Madison, Madison, WI53706
| | - Darrell N. Kotton
- Center for Regenerative Medicine, Boston University and Boston Medical Center, Boston, MA02118
- The Pulmonary Center and Department of Medicine, Boston University Chobanian and Avedisian School of Medicine, Boston, MA02118
| | - David F. Boyd
- Department of Molecular, Cell, and Developmental Biology, University of California, Santa Cruz, CA95064
| | - Bridget S. Banach
- Department of Pathology, Delnor Hospital-Northwestern Medicine, Geneva, IL60134
| | - Robert N. Kirchdoerfer
- Department of Biochemistry, College of Agricultural and Life Sciences, University of Wisconsin-Madison, Madison, WI53706
| | - Mohsan Saeed
- Department of Biochemistry and Cell Biology, Boston University Chobanian and Avedisian School of Medicine, Boston University, Boston, MA02118
- National Emerging Infectious Diseases Laboratories, Boston University, Boston, MA02118
| | - Susan C. Baker
- Department of Microbiology and Immunology, Stritch School of Medicine, Loyola University Chicago, Maywood, IL60153
| |
Collapse
|
19
|
Zhang Y, Wang L, Zeng J, Shen W. Research advances in polyphenols from Chinese herbal medicine for the prevention and treatment of chronic obstructive pulmonary disease: a review. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025:10.1007/s00210-025-03945-y. [PMID: 40035820 DOI: 10.1007/s00210-025-03945-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/26/2024] [Accepted: 02/17/2025] [Indexed: 03/06/2025]
Abstract
Chronic obstructive pulmonary disease (COPD) is a global health problem due to its high death and morbidity worldwide, which is characterized by an incompletely reversible limitation in airflow that is not fully reversible. Unfortunately, Western medical treatments are unable to reverse the progressive decline in lung function. Importantly, polyphenolic compounds isolated from Chinese herbal medicine exhibited therapeutic/interventional effects on COPD in preclinical studies. This review systematically analyzed the pathogenesis of COPD, such as inflammation, oxidative stress, protease/antiprotease imbalance, aging, cell death, and dysbiosis of gut microbiota. Moreover, this review summarized the regulatory mechanisms of natural polyphenolic compounds for the treatment of COPD. Several studies have demonstrated that natural polyphenolic compounds have therapeutic effects on COPD by regulating various biological processes, such as anti-inflammatory, reduction of oxidative damage, anti-cell death, and inhibition of airway hyperglycemia. Mechanistically, this review found that the promising effects of natural polyphenolic compounds on COPD were mainly achieved through modulating the NF-κB and MAPK inflammatory pathways, the Nrf2 oxidative stress pathway, and the SIRT1/PGC-1α lung injury pathway. Furthermore, this review analyzed the efficacy and safety of natural polyphenolic compounds for the treatment of COPD in clinical trials, and discussed their challenges and future development directions. In conclusion, this review combined the latest literature to illustrate the various pathogenesis and interrelationships of COPD in the form of graphs, texts, and tables, and sorted out the functional role and mechanisms of natural polyphenols in treating COPD, with a view to providing new ideas and plans for the in-depth research on COPD and the systemic treatment of COPD with Chinese herbal medicine.
Collapse
Affiliation(s)
- Yang Zhang
- Department of General Practice Medicine, The Second Affiliated Hospital of Kunming Medical University, No. 374 Dianmian Avenue, Wuhua District, Kunming, 650101, China
| | - Lijuan Wang
- Department of Nuclear Medicine, The Second Affiliated Hospital of Kunming Medical University, Kunming, 650101, Yunnan, China
| | - Jinyi Zeng
- Department of General Practice Medicine, The Second Affiliated Hospital of Kunming Medical University, No. 374 Dianmian Avenue, Wuhua District, Kunming, 650101, China
| | - Wen Shen
- Department of General Practice Medicine, The Second Affiliated Hospital of Kunming Medical University, No. 374 Dianmian Avenue, Wuhua District, Kunming, 650101, China.
| |
Collapse
|
20
|
Tang H, Wen J, Wang L, Yang Q, Qin T, Ren Y, Zhao Y, Li C, Li J, Cao H, Xu J, Yang Q. Vagus nerve stimulation inhibits PANoptosis and promotes neurofunctional recovery of cerebral ischemic stroke in a Sirt1-dependent manner. Neurochem Int 2025; 184:105950. [PMID: 39971240 DOI: 10.1016/j.neuint.2025.105950] [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: 11/26/2024] [Revised: 01/25/2025] [Accepted: 02/16/2025] [Indexed: 02/21/2025]
Abstract
Vagus nerve stimulation (VNS) can promote neurofunctional recovery following cerebral ischemic stroke (CIS), but the underlying mechanism remains unclear. PANoptosis, a novel form of inflammatory programmed cell death, may play a role in the progression of CIS. Our previous studies have indicated that Sirt1 exerts neuroprotection against CIS by modulating various programmed cell death pathways. It needs to be clarified whether and how VNS regulates PANoptosis through Sirt1, thereby affecting the recovery of CIS. This study aims to clarify the role of VNS in modulating neuronal PANoptosis following CIS, and elucidate its underlying mechanisms. Models of middle cerebral artery occlusion/reperfusion (MCAO/R) in rats and oxygen-glucose deprivation/reoxygenation (OGD/R) in primary neurons were established to assess the occurrence of neuronal PANoptosis following CIS. Circulating Sirt1 levels were measured in two independent cohorts of acute ischemic stroke (AIS) patients. VNS was administered to activate Sirt1, and its effects on PANoptosis and neurological recovery were evaluated. We found that neuronal PANoptosis was induced following CIS, which was reversed via VNS intervention. Sirt1 levels in serum of AIS patients were significantly increased, and positively correlated with infarct volume and National Institutes of Health Stroke Scale scores. In contrast, Sirt1 was downregulated in brain tissue from rodent models and AIS patients. This discrepancy in expression levels can be attributed to the increased generation of Sirt1 by peripheral macrophages. VNS upregulated Sirt1 expression, while the Sirt1 inhibitor EX527 negated the effects of VNS on PANoptosis, infarct volume, and neurofunctional recovery. These findings indicate that VNS may inhibit PANoptosis and promote neurofunctional recovery following CIS in a Sirt1-dependent manner, which may be a new potential target for stroke therapy. Sirt1 may also serve as a blood biomarker for patient stratification with independent prognostic value in AIS patients.
Collapse
Affiliation(s)
- Hao Tang
- Department of Neurology, The Frist Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Jun Wen
- Department of Neurology, The Frist Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Ling Wang
- Department of Neurology, The Frist Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Qinghuan Yang
- Department of Neurology, The Frist Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Ting Qin
- Department of Respiratory Disease, Daping Hospital, Army Medical University, Chongqing, China
| | - Yu Ren
- Department of Neurology, The Frist Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yong Zhao
- Department of Neurology, The Frist Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Changqing Li
- Department of Neurology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Jiani Li
- Department of Neurology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Hui Cao
- Department of Neurosurgery, The Third Hospital of Mianyang, Sichuan, China
| | - Jianfeng Xu
- Department of Neurosurgery, The Third Hospital of Mianyang, Sichuan, China
| | - Qin Yang
- Department of Neurology, The Frist Affiliated Hospital of Chongqing Medical University, Chongqing, China.
| |
Collapse
|
21
|
de Oliveira Silva Pinto M, de Paula Pereira L, de Mendonça Angelo ALP, Xavier MAP, de Magalhães Vieira Machado A, Russo RC. Dissecting the COVID-19 Immune Response: Unraveling the Pathways of Innate Sensing and Response to SARS-CoV-2 Structural Proteins. J Mol Recognit 2025; 38:e70002. [PMID: 39905998 DOI: 10.1002/jmr.70002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2024] [Revised: 01/15/2025] [Accepted: 01/22/2025] [Indexed: 02/06/2025]
Abstract
Severe acute respiratory syndrome coronavirus (SARS-CoV), the virus responsible for COVID-19, interacts with the host immune system through complex mechanisms that significantly influence disease outcomes, affecting both innate and adaptive immunity. These interactions are crucial in determining the disease's severity and the host's ability to clear the virus. Given the virus's substantial socioeconomic impact, high morbidity and mortality rates, and public health importance, understanding these mechanisms is essential. This article examines the diverse innate immune responses triggered by SARS-CoV-2's structural proteins, including the spike (S), membrane (M), envelope (E), and nucleocapsid (N) proteins, along with nonstructural proteins (NSPs) and open reading frames. These proteins play pivotal roles in immune modulation, facilitating viral replication, evading immune detection, and contributing to severe inflammatory responses such as cytokine storms and acute respiratory distress syndrome (ARDS). The virus employs strategies like suppressing type I interferon production and disrupting key antiviral pathways, including MAVS, OAS-RNase-L, and PKR. This study also explores the immune pathways that govern the activation and suppression of immune responses throughout COVID-19. By analyzing immune sensing receptors and the responses initiated upon recognizing SARS-CoV-2 structural proteins, this review elucidates the complex pathways associated with the innate immune response in COVID-19. Understanding these mechanisms offers valuable insights for therapeutic interventions and informs public health strategies, contributing to a deeper understanding of COVID-19 immunopathogenesis.
Collapse
Affiliation(s)
- Matheus de Oliveira Silva Pinto
- Laboratory of Pulmonary Immunology and Mechanics, Department of Physiology and Biophysics, Institute of Biological Sciences, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, Minas Gerais, Brazil
- Viral Disease Immunology Group, Fundação Osvaldo Cruz, Instituto René Rachou, Belo Horizonte, Minas Gerais, Brazil
| | - Leonardo de Paula Pereira
- Laboratory of Pulmonary Immunology and Mechanics, Department of Physiology and Biophysics, Institute of Biological Sciences, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, Minas Gerais, Brazil
- Viral Disease Immunology Group, Fundação Osvaldo Cruz, Instituto René Rachou, Belo Horizonte, Minas Gerais, Brazil
| | | | | | | | - Remo Castro Russo
- Laboratory of Pulmonary Immunology and Mechanics, Department of Physiology and Biophysics, Institute of Biological Sciences, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, Minas Gerais, Brazil
| |
Collapse
|
22
|
Centofanti E, Oyler-Yaniv A, Oyler-Yaniv J. Deep learning-based image classification reveals heterogeneous execution of cell death fates during viral infection. Mol Biol Cell 2025; 36:ar29. [PMID: 39841552 PMCID: PMC11974948 DOI: 10.1091/mbc.e24-10-0438] [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: 10/02/2024] [Revised: 01/09/2025] [Accepted: 01/15/2025] [Indexed: 01/24/2025] Open
Abstract
Cell fate decisions, such as proliferation, differentiation, and death, are driven by complex molecular interactions and signaling cascades. While significant progress has been made in understanding the molecular determinants of these processes, historically, cell fate transitions were identified through light microscopy that focused on changes in cell morphology and function. Modern techniques have shifted toward probing molecular effectors to quantify these transitions, offering more precise quantification and mechanistic understanding. However, challenges remain in cases where the molecular signals are ambiguous, complicating the assignment of cell fate. During viral infection, programmed cell death (PCD) pathways, including apoptosis, necroptosis, and pyroptosis, exhibit complex signaling and molecular cross-talk. This can lead to simultaneous activation of multiple PCD pathways, which confounds assignment of cell fate based on molecular information alone. To address this challenge, we employed deep learning-based image classification of dying cells to analyze PCD in single herpes simplex virus-1 (HSV-1)-infected cells. Our approach reveals that despite heterogeneous activation of signaling, individual cells adopt predominantly prototypical death morphologies. Nevertheless, PCD is executed heterogeneously within a uniform population of virus-infected cells and varies over time. These findings demonstrate that image-based phenotyping can provide valuable insights into cell fate decisions, complementing molecular assays.
Collapse
Affiliation(s)
- Edoardo Centofanti
- The Department of Systems Biology at Harvard Medical School, Boston, MA 02115
| | - Alon Oyler-Yaniv
- The Department of Systems Biology at Harvard Medical School, Boston, MA 02115
| | | |
Collapse
|
23
|
Soe YM, Sim SL, Kumari S. Innate Immune Sensors and Cell Death-Frontiers Coordinating Homeostasis, Immunity, and Inflammation in Skin. Viruses 2025; 17:241. [PMID: 40006996 PMCID: PMC11861910 DOI: 10.3390/v17020241] [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: 12/13/2024] [Revised: 02/05/2025] [Accepted: 02/06/2025] [Indexed: 02/27/2025] Open
Abstract
The skin provides a life-sustaining interface between the body and the external environment. A dynamic communication among immune and non-immune cells in the skin is essential to ensure body homeostasis. Dysregulated cellular communication can lead to the manifestation of inflammatory skin conditions. In this review, we will focus on the following two key frontiers in the skin: innate immune sensors and cell death, as well as their cellular crosstalk in the context of skin homeostasis and inflammation. This review will highlight the recent advancements and mechanisms of how these pathways integrate signals and orchestrate skin immunity, focusing on inflammatory skin diseases and skin infections in mice and humans.
Collapse
Affiliation(s)
| | | | - Snehlata Kumari
- Frazer Institute, The University of Queensland, Dermatology Research Centre, Woolloongabba, Brisbane, QLD 4102, Australia; (Y.M.S.); (S.L.S.)
| |
Collapse
|
24
|
Li F, Zhang Q, Rong Y, Xiang S, Wang J. TAT-N24 enhances retinal ganglion cell survival by suppressing ZBP1-PANoptosome-mediated PANoptosis in an acute glaucoma mouse model. Exp Eye Res 2025; 251:110244. [PMID: 39832635 DOI: 10.1016/j.exer.2025.110244] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Revised: 12/18/2024] [Accepted: 01/17/2025] [Indexed: 01/22/2025]
Abstract
The abrupt and substantial elevation of intraocular pressure (IOP) in acute glaucoma induces retinal ischemia/reperfusion (I/R) injury, resulting in progressive retinal ganglion cell (RGC) death and irreversible visual impairment. PANoptosis, a form of regulated cell death consisting of pyroptosis, apoptosis and necroptosis, is reported to be involved in high IOP-induced RGC death. However, the precise mechanisms of RGC death remain unclear, and neuroinflammation is considered to play a vital role. TAT-N24, a synthetic inhibitor targeting the p55 regulatory subunit of phosphatidylinositol 3-kinase (p55PIK) signaling, demonstrates anti-inflammatory effect in uveitis and may have certain neuroprotective effects. Therefore, we investigated whether TAT-N24 could shield RGCs from immunoinflammatory damage in an acute glaucoma mouse model and explored the potential mechanism associated with PANoptosis. A mouse model of acute ocular hypertension (AOH) was established. Intravitreal injection of TAT-N24 was conducted to evaluate its impact on RGC death. The expression levels of key components in PANoptosis were analyzed using RT-qPCR and Western blotting. Immunohistochemistry and immunofluorescence staining on eyeball sections were employed to assess the expression of p55PIK, Brn3a, and ionized calcium binding adaptor molecule 1 (Iba1). Retinal structure was examined by H&E staining, while cell apoptosis was evaluated by TdT-mediated dUTP nick end labeling (TUNEL). The results showed that intravitreal injection of TAT-N24 effectively alleviated RGC death and retinal damage induced by AOH injury. The key components in PANoptosis were markedly upregulated after AOH injury, while these components were significantly inhibited after TAT-N24 treatment. Moreover, the expression levels of Z-DNA-binding protein 1 (ZBP1)-PANoptosome (ZBP1, RIPK1, RIPK3, and Caspase-8), NLR family pyrin domain-containing protein 3 (NLRP3), and NLR family CARD domain-containing protein 4 (NLRC4) inflammasomes were notably elevated after AOH injury, which was significantly suppressed by TAT-N24. In conclusion, PANoptosis was involved in AOH-induced RGC death and retinal damage. TAT-N24 exhibited an anti-PANoptotic effect, protecting RGCs by inhibiting ZBP1-PANoptosome as well as NLRP3 and NLRC4 inflammasomes after AOH injury.
Collapse
Affiliation(s)
- Fei Li
- Department of Ophthalmology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Qiuxiang Zhang
- Department of Ophthalmology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Yan Rong
- Department of Ophthalmology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Sifei Xiang
- Department of Ophthalmology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Junming Wang
- Department of Ophthalmology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China.
| |
Collapse
|
25
|
Amusan OT, Wang S, Yin C, Koehler HS, Li Y, Tenev T, Wilson R, Bellenie B, Zhang T, Wang J, Liu C, Seong K, Poorbaghi SL, Yates J, Shen Y, Upton JW, Meier P, Balachandran S, Guo H. RIPK1 is required for ZBP1-driven necroptosis in human cells. PLoS Biol 2025; 23:e3002845. [PMID: 39982916 PMCID: PMC11844899 DOI: 10.1371/journal.pbio.3002845] [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: 08/29/2024] [Accepted: 01/06/2025] [Indexed: 02/23/2025] Open
Abstract
Necroptosis initiated by the host sensor Z-NA binding protein 1 (ZBP1) is essential for host defense against a growing number of viruses, including herpes simplex virus 1 (HSV-1). Studies with HSV-1 and other necroptogenic stimuli in murine settings have suggested that ZBP1 triggers necroptosis by directly complexing with the kinase RIPK3. Whether this is also the case in human cells, or whether additional co-factors are needed for ZBP1-mediated necroptosis, is unclear. Here, we show that ZBP1-induced necroptosis in human cells requires RIPK1. We have found that RIPK1 is essential for forming a stable and functional ZBP1-RIPK3 complex in human cells, but is dispensable for the formation of the equivalent murine complex. The receptor-interacting protein (RIP) homology interaction motif (RHIM) in RIPK3 is responsible for this difference between the 2 species, because replacing the RHIM in human RIPK3 with the RHIM from murine RIPK3 is sufficient to overcome the requirement for RIPK1 in human cells. These observations describe a critical mechanistic difference between mice and humans in how ZBP1 engages in necroptosis, with important implications for treating human diseases.
Collapse
Affiliation(s)
- Oluwamuyiwa T. Amusan
- Department of Microbiology and Immunology, Louisiana State University Health Sciences Center-Shreveport, Shreveport, Louisiana, United States of America
| | - Shuqi Wang
- Department of Microbiology and Immunology, Louisiana State University Health Sciences Center-Shreveport, Shreveport, Louisiana, United States of America
| | - Chaoran Yin
- Blood Cell Development and Function, Fox Chase Cancer Center, Philadelphia, Pennsylvania, United States of America
| | - Heather S. Koehler
- School of Molecular Biosciences, Washington State University, Pullman, Washington State, United States of America
| | - Yixun Li
- Department of Microbiology and Immunology, Louisiana State University Health Sciences Center-Shreveport, Shreveport, Louisiana, United States of America
| | - Tencho Tenev
- The Breast Cancer Now Toby Robins Research Centre, Institute of Cancer Research, London, United Kingdom
| | - Rebecca Wilson
- The Breast Cancer Now Toby Robins Research Centre, Institute of Cancer Research, London, United Kingdom
| | - Benjamin Bellenie
- Centre for Cancer Drug Discovery at the Institute of Cancer Research, London, United Kingdom
| | - Ting Zhang
- Blood Cell Development and Function, Fox Chase Cancer Center, Philadelphia, Pennsylvania, United States of America
| | - Jian Wang
- Department of Microbiology and Immunology, Louisiana State University Health Sciences Center-Shreveport, Shreveport, Louisiana, United States of America
- Center for Applied Immunology and Pathological Processes, Louisiana State University Health Sciences Center-Shreveport, Shreveport, Louisiana, United States of America
| | - Chang Liu
- Department of Biophysics and Biophysical Chemistry, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - Kim Seong
- Department of Microbiology and Immunology, Louisiana State University Health Sciences Center-Shreveport, Shreveport, Louisiana, United States of America
| | - Seyedeh L. Poorbaghi
- Department of Microbiology and Immunology, Louisiana State University Health Sciences Center-Shreveport, Shreveport, Louisiana, United States of America
| | - Joseph Yates
- Department of Microbiology and Immunology, Louisiana State University Health Sciences Center-Shreveport, Shreveport, Louisiana, United States of America
| | - Yuchen Shen
- Department of Microbiology and Immunology, Louisiana State University Health Sciences Center-Shreveport, Shreveport, Louisiana, United States of America
| | - Jason W. Upton
- Department of Biological Sciences, Auburn University, Auburn, Alabama, United States of America
| | - Pascal Meier
- The Breast Cancer Now Toby Robins Research Centre, Institute of Cancer Research, London, United Kingdom
| | - Siddharth Balachandran
- Blood Cell Development and Function, Fox Chase Cancer Center, Philadelphia, Pennsylvania, United States of America
| | - Hongyan Guo
- Department of Microbiology and Immunology, Louisiana State University Health Sciences Center-Shreveport, Shreveport, Louisiana, United States of America
| |
Collapse
|
26
|
Cai Y, Xiao H, Zhou Q, Lin J, Liang X, Xu W, Cao Y, Zhang X, Wang H. Comprehensive Analyses of PANoptosome with Potential Implications in Cancer Prognosis and Immunotherapy. Biochem Genet 2025; 63:331-353. [PMID: 38436818 PMCID: PMC11832696 DOI: 10.1007/s10528-024-10687-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2023] [Accepted: 01/04/2024] [Indexed: 03/05/2024]
Abstract
Cell death resistance significantly contributes to poor therapeutic outcomes in various cancers. PANoptosis, a unique inflammatory programmed cell death (PCD) pathway activated by specific triggers and regulated by the PANoptosome, possesses key features of apoptosis, pyroptosis, and necroptosis, but these cannot be accounted for by any of the three PCD pathways alone. While existing studies on PANoptosis have predominantly centered on infectious and inflammatory diseases, its role in cancer malignancy has been understudied. In this comprehensive investigation, we conducted pan-cancer analyses of PANoptosome component genes across 33 cancer types. We characterized the genetic, epigenetic, and transcriptomic landscapes, and introduced a PANoptosome-related potential index (PANo-RPI) for evaluating the intrinsic PANoptosome assembly potential in cancers. Our findings unveil PANo-RPI as a prognostic factor in numerous cancers, including KIRC, LGG, and PAAD. Crucially, we established a significant correlation between PANo-RPI and tumor immune responses, as well as the infiltration of diverse lymphoid and myeloid cell subsets across nearly all cancer types. Moreover, a high PANo-RPI was consistently associated with improved immunotherapy response and efficacy, as evidenced by re-analysis of multiple immunotherapy cohorts. In conclusion, our study suggests that targeting PANoptosome components and modulating PANoptosis may hold tremendous therapeutic potential in the context of cancer.
Collapse
Affiliation(s)
- Yonghua Cai
- Department of Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong, People's Republic of China
| | - Heng Xiao
- Southern Medical School, No. 1023, South Shatai Road, Baiyun District, Guangzhou, 510515, Guangdong, China
| | - Qixiong Zhou
- Department of Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong, People's Republic of China
| | - Jie Lin
- Department of Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong, People's Republic of China
| | - Xianqiu Liang
- Department of Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong, People's Republic of China
| | - Wei Xu
- Department of Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong, People's Republic of China
| | - Yongfu Cao
- Department of Neurosurgery, Key Laboratory of Biological Targeting Diagnosis, Therapy and Rehabilitation of Guangdong Higher Education Institutes, The Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, China.
| | - Xian Zhang
- Department of Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong, People's Republic of China.
| | - Hai Wang
- Department of Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong, People's Republic of China.
| |
Collapse
|
27
|
Meng C, Wang Y, Zheng T, Rong Z, Lv Z, Wu C, Zhou X, Mao W. A novel approach to the prevention and management of chemotherapy-induced cardiotoxicity: PANoptosis. Chem Biol Interact 2025; 407:111379. [PMID: 39788474 DOI: 10.1016/j.cbi.2025.111379] [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: 11/11/2024] [Revised: 12/31/2024] [Accepted: 01/07/2025] [Indexed: 01/12/2025]
Abstract
As a fundamental component of antitumor therapy, chemotherapy-induced cardiotoxicity (CIC) has emerged as a leading cause of long-term mortality in patients with malignant tumors. Unfortunately, there are currently no effective therapeutic preventive or treatment strategies, and the underlying pathophysiological mechanisms of CIC remain inadequately understood. A growing number of studies have shown that different mechanisms of cell death, such as apoptosis, pyroptosis, and necroptosis, are essential for facilitating the cardiotoxic effects of chemotherapy. The PANoptosis mode represents a highly synchronized and dynamically balanced programmed cell death (PCD) process that integrates the principal molecular characteristics of necroptosis, apoptosis, and pyroptosis. Recent research has revealed a significant correlation between PANoptosis and the apoptosis of tumor cells. Chemotherapy drugs can activate PANoptosis, which is involved in the development of cardiovascular diseases. These findings suggest that PANoptosis marks the point where the effectiveness of chemotherapy against tumors overlaps with the onset and development of cardiovascular diseases. Furthermore, previous studies have demonstrated that CIC can simultaneously induce pyrodeath, apoptosis, and necrotic apoptosis. Therefore, PANoptosis may represent a potential mechanism and target for the prevention of CIC. This study explored the interactions among the three main mechanisms of PCD, pyroptosis, apoptosis, and necroptosis in CICs and analyzed the relevant literature on PANoptosis and CICs. The purpose of this work is to serve as a reference for future investigations on the role of PANoptosis in the development and mitigation of cardiotoxicity associated with chemotherapy.
Collapse
Affiliation(s)
- Chenchen Meng
- The First School of Clinical Medicine, Zhejiang Chinese Medical University, 310053, Hangzhou, China
| | - Yali Wang
- Department of Cardiology, Zhejiang Hospital (Affiliated Zhejiang Hospital, Zhejiang University School of Medicine), Hangzhou, Zhejiang, 310007, China
| | - Tiantian Zheng
- The First School of Clinical Medicine, Zhejiang Chinese Medical University, 310053, Hangzhou, China
| | - Zheng Rong
- The First School of Clinical Medicine, Zhejiang Chinese Medical University, 310053, Hangzhou, China
| | - Zhengtian Lv
- The First School of Clinical Medicine, Zhejiang Chinese Medical University, 310053, Hangzhou, China
| | - Chenxia Wu
- The First School of Clinical Medicine, Zhejiang Chinese Medical University, 310053, Hangzhou, China; Department of Cardiology, Zhejiang Hospital (Affiliated Zhejiang Hospital, Zhejiang University School of Medicine), Hangzhou, Zhejiang, 310007, China
| | - Xinbin Zhou
- Department of Cardiology, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), 310006, Hangzhou, Zhejiang, China.
| | - Wei Mao
- Department of Cardiology, Zhejiang Hospital (Affiliated Zhejiang Hospital, Zhejiang University School of Medicine), Hangzhou, Zhejiang, 310007, China; Zhejiang Key Laboratory of Integrative Chinese and Western Medicine for Diagnosis and Treatment of Circulatory Diseases, Zhejiang Hospital (Affiliated Zhejiang Hospital, Zhejiang University School of Medicine), Hangzhou, Zhejiang, 310007, China; Zhejiang Engineering Research Center for Precise Diagnosis and Innovative Traditional Chinese Medicine for Cardiovascular Diseases, Zhejiang Hospital (Affiliated Zhejiang Hospital, Zhejiang University School of Medicine), Hangzhou, Zhejiang, 310007, China.
| |
Collapse
|
28
|
Tat VY, Drelich AK, Huang P, Khanipov K, Hsu JC, Widen SG, Tseng CTK, Golovko G. Characterizing temporal and global host innate immune responses against SARS-CoV-1 and -2 infection in pathologically relevant human lung epithelial cells. PLoS One 2025; 20:e0317921. [PMID: 39874350 PMCID: PMC11774383 DOI: 10.1371/journal.pone.0317921] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Accepted: 01/07/2025] [Indexed: 01/30/2025] Open
Abstract
Severe acute respiratory syndrome coronavirus-1 (SARS-CoV-1) and -2 (SARS-CoV-2) are beta-coronaviruses (β-CoVs) that have caused significant morbidity and mortality worldwide. Therefore, a better understanding of host responses to β-CoVs would provide insights into the pathogenesis of these viruses to identify potential targets for medical countermeasures. In this study, our objective is to use a systems biology approach to explore the magnitude and scope of innate immune responses triggered by SARS-CoV-1 and -2 infection over time in pathologically relevant human lung epithelial cells (Calu-3/2B4 cells). Total RNA extracted at 12, 24, and 48 hours after β-CoVs or mock infection of Calu-3/2B4 cells were subjected to RNA sequencing and functional enrichment analysis to select genes whose expressions were significantly modulated post-infection. The results demonstrate that SARS-CoV-1 and -2 stimulate similar yet distinct innate antiviral signaling pathways in pathologically relevant human lung epithelial cells. Furthermore, we found that many genes related to the viral life cycle, interferons, and interferon-stimulated genes (ISGs) were upregulated at multiple time points. Based on their profound modulation upon infection by SARS-CoV-1, SARS-CoV-2, and Omicron BA.1, four ISGs, i.e., bone marrow stromal cell antigen 2 (BST2), Z-DNA Binding Protein 1 (ZBP1), C-X-C Motif Chemokine Ligand 11 (CXCL11), and Interferon Induced Transmembrane Protein 1 (IFITM1), were identified as potential drug targets against β-CoVs. Our findings suggest that these genes affect both pathogens directly and indirectly through the innate immune response, making them potential targets for host-directed antivirals. Altogether, our results demonstrate that SARS-CoV-1 and SARS-CoV-2 infection induce differential effects on host innate immune responses.
Collapse
Affiliation(s)
- Vivian Y. Tat
- Department of Pathology, The University of Texas Medical Branch, Galveston, Texas, United States of America
| | - Aleksandra K. Drelich
- Department of Microbiology & Immunology, The University of Texas Medical Branch, Galveston, Texas, United States of America
| | - Pinghan Huang
- Department of Microbiology & Immunology, The University of Texas Medical Branch, Galveston, Texas, United States of America
| | - Kamil Khanipov
- Department of Pharmacology & Toxicology, The University of Texas Medical Branch, Galveston, Texas, United States of America
| | - Jason C. Hsu
- Department of Biochemistry & Molecular Biology, The University of Texas Medical Branch, Galveston, Texas, United States of America
| | - Steven G. Widen
- Department of Biochemistry & Molecular Biology, The University of Texas Medical Branch, Galveston, Texas, United States of America
| | - Chien-Te Kent Tseng
- Department of Pathology, The University of Texas Medical Branch, Galveston, Texas, United States of America
- Department of Microbiology & Immunology, The University of Texas Medical Branch, Galveston, Texas, United States of America
- Department of Biochemistry & Molecular Biology, The University of Texas Medical Branch, Galveston, Texas, United States of America
| | - George Golovko
- Department of Pharmacology & Toxicology, The University of Texas Medical Branch, Galveston, Texas, United States of America
| |
Collapse
|
29
|
Hou G, Chen Y, Lei H, Lu S, Cheng L. Nanomaterials-Induced PANoptosis: A Promising Anti-Tumor Strategy. Angew Chem Int Ed Engl 2025; 64:e202419649. [PMID: 39560000 DOI: 10.1002/anie.202419649] [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: 10/11/2024] [Revised: 11/19/2024] [Accepted: 11/19/2024] [Indexed: 11/20/2024]
Abstract
Malignant tumors pose a significant threat to global public health. Promoting programmed cell death in cancer cells has become a critical strategy for cancer treatment. PANoptosis, a newly discovered form of regulated cell death, integrates key molecular components of pyroptosis, apoptosis, and necroptosis, activating these three death pathways simultaneously to achieve synergistic multi-mechanistic killing. PANoptosis significantly inhibits cancer cell growth and resistance and activates strong anti-tumor immune response, making tumor-specific induction of PANoptosis a potential cancer therapeutic strategy. Currently, cancer treatment research related to PANoptosis is focused mainly on the development of small molecules and cytokines. However, these approaches still face limitations in terms of metabolic stability and tumor specificity. The unique physicochemical properties and biological activities of nanomaterials hold significant promise for optimizing PANoptosis induction strategies. This review summarizes the concept and mechanisms of PANoptosis, highlights the latest applications of nanoagents in PANoptosis-based anti-cancer therapy, and discusses the challenges and future directions for clinical translation. It is hoped that this review will inspire further exploration and development of PANoptosis-based cancer treatments, providing new perspectives for researchers in the field.
Collapse
Affiliation(s)
- Guanghui Hou
- Institute of Functional Nano & Soft Materials (FUNSOM) Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices, Soochow University, Suzhou, 215123, China
| | - Youdong Chen
- Institute of Functional Nano & Soft Materials (FUNSOM) Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices, Soochow University, Suzhou, 215123, China
| | - Huali Lei
- Institute of Functional Nano & Soft Materials (FUNSOM) Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices, Soochow University, Suzhou, 215123, China
| | - Shunyi Lu
- 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
|
30
|
Bi X, Li M, Guo Y, Hu M, Chen Y, Lian N, Chen S, Li M, Gu H, Chen X. ZBP1-mediated PANoptosis is a crucial lethal form in diverse keratinocyte death modalities in UVB-induced skin injury. Cell Death Dis 2025; 16:44. [PMID: 39863598 PMCID: PMC11762280 DOI: 10.1038/s41419-025-07351-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Revised: 12/20/2024] [Accepted: 01/14/2025] [Indexed: 01/27/2025]
Abstract
UVB irradiation induces diverse modalities of regulatory cell death in keratinocytes. Recently, the pattern of coexistence of pyroptosis, apoptosis, and necroptosis has been termed PANoptosis; however, whether PANoptosis occurs in keratinocytes in UVB-induced skin injury remains unclear. We observed that the key molecules of GSDMD-mediated pyroptosis, apoptosis, and necroptosis, which are N-terminal GSDMD, cleaved caspase-3/PARP, and phosphorylated MLKL, respectively, were elevated in keratinocytes of UVB-challenged mice and human skin tissue. Through keratinocyte-specific gene knockout or using corresponding inhibitors, we found that individual inhibition of GSDMD-mediated pyroptosis, caspase-3-mediated apoptosis, or MLKL-mediated necroptosis did not reduce the overall level of keratinocyte death after UVB exposure, and that the other two pathways maintained the activation. However, when the PANoptosome sensor ZBP1 was knocked out, keratinocyte death was reduced and epidermal thickening was alleviated in UVB-challenged mice. In conclusion, our study demonstrated that UVB irradiation induces ZBP1-mediated PANoptosis in keratinocytes, which is a crucial lethal form in diverse keratinocyte death modalities in UVB-induced skin injury. The above findings provide a new insight on the complexity of regulated cell death modalities in keratinocytes exposed to UV irradiation.
Collapse
Affiliation(s)
- Xuechan Bi
- Jiangsu Key Laboratory of Molecular Biology for Skin Diseases and STIs, Hospital for Skin Diseases, Institute of Dermatology, Chinese Academy of Medical Sciences & Peking Union Medical College, Nanjing, 210042, Jiangsu, China
| | - Min Li
- Jiangsu Key Laboratory of Molecular Biology for Skin Diseases and STIs, Hospital for Skin Diseases, Institute of Dermatology, Chinese Academy of Medical Sciences & Peking Union Medical College, Nanjing, 210042, Jiangsu, China
| | - Yiming Guo
- State Key Laboratory of Natural Medicines, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, 211198, Jiangsu, China
| | - Mengyao Hu
- School of Public Health, Nanjing Medical University, Nanjing, 211166, Jiangsu, China
| | - Yujie Chen
- Jiangsu Key Laboratory of Molecular Biology for Skin Diseases and STIs, Hospital for Skin Diseases, Institute of Dermatology, Chinese Academy of Medical Sciences & Peking Union Medical College, Nanjing, 210042, Jiangsu, China
| | - Ni Lian
- Jiangsu Key Laboratory of Molecular Biology for Skin Diseases and STIs, Hospital for Skin Diseases, Institute of Dermatology, Chinese Academy of Medical Sciences & Peking Union Medical College, Nanjing, 210042, Jiangsu, China
| | - Sihan Chen
- Jiangsu Key Laboratory of Molecular Biology for Skin Diseases and STIs, Hospital for Skin Diseases, Institute of Dermatology, Chinese Academy of Medical Sciences & Peking Union Medical College, Nanjing, 210042, Jiangsu, China
| | - Min Li
- Jiangsu Key Laboratory of Molecular Biology for Skin Diseases and STIs, Hospital for Skin Diseases, Institute of Dermatology, Chinese Academy of Medical Sciences & Peking Union Medical College, Nanjing, 210042, Jiangsu, China
| | - Heng Gu
- Jiangsu Key Laboratory of Molecular Biology for Skin Diseases and STIs, Hospital for Skin Diseases, Institute of Dermatology, Chinese Academy of Medical Sciences & Peking Union Medical College, Nanjing, 210042, Jiangsu, China.
| | - Xu Chen
- Jiangsu Key Laboratory of Molecular Biology for Skin Diseases and STIs, Hospital for Skin Diseases, Institute of Dermatology, Chinese Academy of Medical Sciences & Peking Union Medical College, Nanjing, 210042, Jiangsu, China.
| |
Collapse
|
31
|
Jiang J, Huang LS. Special Issue "Molecular Advances and Perspectives of Lung Disease". Int J Mol Sci 2025; 26:946. [PMID: 39940714 PMCID: PMC11816814 DOI: 10.3390/ijms26030946] [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: 12/09/2024] [Accepted: 01/09/2025] [Indexed: 02/16/2025] Open
Abstract
Respiratory diseases represent a significant global public health challenge, contributing to high mortality and morbidity rates worldwide [...].
Collapse
Affiliation(s)
- Jiacheng Jiang
- Shanghai Frontiers Science Center of Drug Target Identification and Delivery, School of Pharmaceutical Sciences, Shanghai Jiao Tong University, Shanghai 200240, China;
- National Key Laboratory of Innovative Immunotherapy, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Long Shuang Huang
- Shanghai Frontiers Science Center of Drug Target Identification and Delivery, School of Pharmaceutical Sciences, Shanghai Jiao Tong University, Shanghai 200240, China;
- National Key Laboratory of Innovative Immunotherapy, Shanghai Jiao Tong University, Shanghai 200240, China
| |
Collapse
|
32
|
Stegeman SK, Kourko O, Amsden H, Pellizzari Delano IE, Mamatis JE, Roth M, Colpitts CC, Gee K. RNA Viruses, Toll-Like Receptors, and Cytokines: The Perfect Storm? J Innate Immun 2025; 17:126-153. [PMID: 39820070 PMCID: PMC11845175 DOI: 10.1159/000543608] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2024] [Accepted: 01/13/2025] [Indexed: 01/19/2025] Open
Abstract
BACKGROUND The interactions between viruses and the host immune response are nuanced and intricate. The cytokine response arguably plays a central role in dictating the outcome of virus infection, balancing inflammation, and healing, which is crucial to resolving infection without destructive immunopathologies. SUMMARY Early innate immune responses are key to the generation of a beneficial or detrimental immune response. These initial responses are regulated by a plethora of surface bound, endosomal, and cytoplasmic innate immune receptors known as pattern recognition receptors. Of these, the Toll-like receptors (TLRs) play an important role in the induction of cytokines during virus infection. Recognizing pathogen-associated molecular patterns (PAMPs) such as viral proteins and/or nucleotide sequences, the TLRs act as sentinels for the initiation and propagation of immune responses. KEY MESSAGES TLRs are important receptors for initiating the innate response to single-stranded RNA (ssRNA) viruses like influenza A virus (IAV), severe acute respiratory syndrome coronavirus-1 (SARS-CoV-1), SARS-CoV-2, Middle East respiratory syndrome coronavirus, dengue virus, and Ebola virus. Infection with these viruses is also associated with aberrant expression of proinflammatory cytokines that contribute to a harmful cytokine storm response. Herein we discuss the connections between these ssRNA viruses, cytokine storm, and the roles of TLRs. BACKGROUND The interactions between viruses and the host immune response are nuanced and intricate. The cytokine response arguably plays a central role in dictating the outcome of virus infection, balancing inflammation, and healing, which is crucial to resolving infection without destructive immunopathologies. SUMMARY Early innate immune responses are key to the generation of a beneficial or detrimental immune response. These initial responses are regulated by a plethora of surface bound, endosomal, and cytoplasmic innate immune receptors known as pattern recognition receptors. Of these, the Toll-like receptors (TLRs) play an important role in the induction of cytokines during virus infection. Recognizing pathogen-associated molecular patterns (PAMPs) such as viral proteins and/or nucleotide sequences, the TLRs act as sentinels for the initiation and propagation of immune responses. KEY MESSAGES TLRs are important receptors for initiating the innate response to single-stranded RNA (ssRNA) viruses like influenza A virus (IAV), severe acute respiratory syndrome coronavirus-1 (SARS-CoV-1), SARS-CoV-2, Middle East respiratory syndrome coronavirus, dengue virus, and Ebola virus. Infection with these viruses is also associated with aberrant expression of proinflammatory cytokines that contribute to a harmful cytokine storm response. Herein we discuss the connections between these ssRNA viruses, cytokine storm, and the roles of TLRs.
Collapse
Affiliation(s)
- Sophia K Stegeman
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, Canada
| | - Olena Kourko
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, Canada
| | - Heather Amsden
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, Canada
| | | | - John E Mamatis
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, Canada
| | - Madison Roth
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, Canada
| | - Che C Colpitts
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, Canada
| | - Katrina Gee
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, Canada
| |
Collapse
|
33
|
Yang K, Wang Y, Jian Y, Wang B, Du H, Xia Y, Bi J, Guo M, Li Z, Wang N. CASP5 associated with PANoptosis promotes tumorigenesis and progression of clear cell renal cell carcinoma. Cancer Cell Int 2025; 25:8. [PMID: 39780201 PMCID: PMC11716502 DOI: 10.1186/s12935-024-03630-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2024] [Accepted: 12/29/2024] [Indexed: 01/11/2025] Open
Abstract
Clear cell renal cell carcinoma (ccRCC) is a globally severe cancer with an unfavorable prognosis. PANoptosis, a form of cell death regulated by PANoptosomes, plays a role in numerous cancer types. However, the specific roles of genes associated with PANoptosis in the development and advancement of ccRCC remain unclear. Our study developed a risk model utilizing three PANoptosis-associated genes (Caspase 4 (CASP4), TLR3, and CASP5). This model demonstrated a high degree of precision in predicting the prognosis for patients with ccRCC. ccRCC patients in the high-risk group had the strongest immune cell activity, experiencing immune evasion, and might potentially derive advantages from treatment involving combined immune checkpoint inhibitors. CASP5 was highly expressed in ccRCC tissues by RT-qPCR, western blotting, and immunofluorescence. Stable CASP5 knockdown cell lines were constructed by lentivirus in vitro transfection technique. Reducing CASP5 level suppressed the growth, migration, and invasion of ccRCC cells, while encouraging cell apoptosis. In addition, the results of in vivo tumorigenesis experiments showed that down-regulating CASP5 expression inhibited the tumorigenic ability of 786-O cells. Together, the innovative risk model using PANoptosis-associated genes effectively forecasts the tumor microenvironment and survival rates for ccRCC, offering a novel approach to the early, precise diagnosis of ccRCC and the advancement of personalized treatment strategies.
Collapse
Affiliation(s)
- Kangkang Yang
- Institute for Genome Engineered Animal Models of Human Diseases, National Center of Genetically Engineered Animal Models for International Research, Dalian Medical University, 9 West Section Lvshun South Road, Dalian, 116044, China
| | - Yushuang Wang
- Department of Clinical Laboratory, Central Hospital of Dalian University of Technology, 826 Southwest Road, Dalian, 116033, China
| | - Yuli Jian
- Liaoning Provincial Core Lab of Glycobiology and Glycoengineering, College of Basic Medical Sciences, Dalian Medical University, 9 West Section Lvshun South Road, Dalian, 116044, China
| | - Bo Wang
- Department of Clinical Laboratory, Central Hospital of Dalian University of Technology, 826 Southwest Road, Dalian, 116033, China
| | - Hao Du
- Department of Urology, Central Hospital of Dalian University of Technology, 826 Southwest Road, Dalian, 116033, China
| | - Yuqing Xia
- Pharmaceutical sciences, Massachusetts College of Pharmacy and Health Science University, 179 Longwood Avenue, Boston, Massachusetts, 02115, USA
| | - Jianlei Bi
- Department of Obstetrics and Gynecology, The Second Hospital of Dalian Medical University, 467 Zhongshan Road, Dalian, 116023, China
| | - Meihua Guo
- Institute for Genome Engineered Animal Models of Human Diseases, National Center of Genetically Engineered Animal Models for International Research, Dalian Medical University, 9 West Section Lvshun South Road, Dalian, 116044, China
| | - Zhi Li
- Department of Clinical Laboratory, Central Hospital of Dalian University of Technology, 826 Southwest Road, Dalian, 116033, China.
| | - Ning Wang
- Institute for Genome Engineered Animal Models of Human Diseases, National Center of Genetically Engineered Animal Models for International Research, Dalian Medical University, 9 West Section Lvshun South Road, Dalian, 116044, China.
| |
Collapse
|
34
|
Song K, Wu Y, Tan S. Caspases in PANoptosis. Curr Res Transl Med 2025; 73:103502. [PMID: 39985853 DOI: 10.1016/j.retram.2025.103502] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Revised: 02/13/2025] [Accepted: 02/19/2025] [Indexed: 02/24/2025]
Abstract
Recent studies prove that the three well-established cell death pathways-pyroptosis, apoptosis, and necroptosis-are not isolated but rather engage in extensive crosstalk. PANoptosis, a newly identified pathway of inflammatory regulated cell death (RCD), integrates characteristics of apoptosis, pyroptosis, and necroptosis. Caspases are a family of conserved cysteine proteases that play critical roles in pyroptosis, apoptosis, and necroptosis. Similarly, caspases also play a role in PANoptosis. In this paper, we review the molecular mechanisms of these three RCDs and the crosstalk between them. We also delineate the discovery of PANoptosis and its association with disease. Furthermore, we discuss the caspase function in PANoptosis, mainly focusing on caspase-6 and caspase-8 molecules. This review describes the key molecules, especially caspases, in the context of PANoptosis research, aiming to provide a foundation for targeted interventions in PANoptosis-associated diseases.
Collapse
Affiliation(s)
- Kaiyuan Song
- Department of Pathophysiology, Xiangya School of Basic Medicine Science, Central South University, Changsha, PR China; Sepsis Translational Medicine Key Laboratory of Hunan Province, Central South University, Changsha, PR China; National Medicine Functional Experimental Teaching Center, Central South University, Changsha, PR China
| | - Yongbin Wu
- Department of Pathophysiology, Xiangya School of Basic Medicine Science, Central South University, Changsha, PR China; Sepsis Translational Medicine Key Laboratory of Hunan Province, Central South University, Changsha, PR China; National Medicine Functional Experimental Teaching Center, Central South University, Changsha, PR China
| | - Sipin Tan
- Department of Pathophysiology, Xiangya School of Basic Medicine Science, Central South University, Changsha, PR China; Sepsis Translational Medicine Key Laboratory of Hunan Province, Central South University, Changsha, PR China; National Medicine Functional Experimental Teaching Center, Central South University, Changsha, PR China.
| |
Collapse
|
35
|
Mishra S, Dey AA, Kesavardhana S. Z-Nucleic Acid Sensing and Activation of ZBP1 in Cellular Physiology and Disease Pathogenesis. Immunol Rev 2025; 329:e13437. [PMID: 39748135 DOI: 10.1111/imr.13437] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2024] [Revised: 12/13/2024] [Accepted: 12/14/2024] [Indexed: 01/04/2025]
Abstract
Z-nucleic acid binding protein 1 (ZBP1) is an innate immune sensor recognizing nucleic acids in Z-conformation. Upon Z-nucleic acid sensing, ZBP1 triggers innate immune activation, inflammation, and programmed cell death during viral infections, mice development, and inflammation-associated diseases. The Zα domains of ZBP1 sense Z-nucleic acids and promote RIP-homotypic interaction motif (RHIM)-dependent signaling complex assembly to mount cell death and inflammation. The studies on ZBP1 spurred an understanding of the role of Z-form RNA and DNA in cellular and physiological functions. In particular, short viral genomic segments, endogenous retroviral elements, and 3'UTR regions are likely sources of Z-RNAs that orchestrate ZBP1 functions. Recent seminal studies identify an intriguing association of ZBP1 with adenosine deaminase acting on RNA-1 (ADAR1), and cyclic GMP-AMP synthase (cGAS) in regulating aberrant nucleic acid sensing, chronic inflammation, and cancer. Thus, ZBP1 is an attractive target to aid the development of specific therapeutic regimes for disease biology. Here, we discuss the role of ZBP1 in Z-RNA sensing, activation of programmed cell death, and inflammation. Also, we discuss how ZBP1 coordinates intracellular perturbations in homeostasis, and Z-nucleic acid formation to regulate chronic diseases and cancer.
Collapse
Affiliation(s)
- Sanchita Mishra
- Department of Biochemistry, Division of Biological Sciences, Indian Institute of Science, Bengaluru, Karnataka, India
| | - Ayushi Amin Dey
- Department of Biochemistry, Division of Biological Sciences, Indian Institute of Science, Bengaluru, Karnataka, India
| | - Sannula Kesavardhana
- Department of Biochemistry, Division of Biological Sciences, Indian Institute of Science, Bengaluru, Karnataka, India
| |
Collapse
|
36
|
Oh J, Kim H, Lee J, Kim S, Shin S, Kim YE, Park S, Lee S. Korean Red ginseng enhances ZBP1-mediated cell death to suppress viral protein expression in host defense against Influenza A virus. J Microbiol 2025; 63:e.2409007. [PMID: 39895072 DOI: 10.71150/jm.2409007] [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: 09/06/2024] [Accepted: 11/22/2024] [Indexed: 02/04/2025]
Abstract
Korean Red ginseng has emerged as a potent candidate in the fight against various viral infections, demonstrating significant efficacy both in vitro and in vivo, particularly against influenza A viruses. Despite substantial evidence of its antiviral properties, the detailed molecular mechanisms through which it reduces viral lethality remain insufficiently understood. Our investigations have highlighted the superior effectiveness of Korean Red ginseng against influenza viruses, outperforming its effects on numerous other viral strains. We aim to uncover the specific mechanisms by which Korean Red ginseng exerts its antiviral effects, focusing on influenza A viruses. Our prior studies have identified the role of Z-DNA-binding protein 1 (ZBP1), a signaling complex involved in inducing programmed cell death in response to influenza virus infection. Given the critical role of ZBP1 as a sensor for viral nucleic acid, we hypothesize that Korean Red ginseng may modulate the ZBP1-derived cell death pathway. This interaction is anticipated to enhance cell death while concurrently suppressing viral protein expression, offering novel insights into the antiviral mechanism of Korean Red ginseng against influenza A viruses.
Collapse
Affiliation(s)
- Jueun Oh
- Department of Biological Science, Ulsan National Institute of Science and Technology (UNIST), Ulsan 44919, Republic of Korea
| | - Hayeon Kim
- Department of Biological Science, Ulsan National Institute of Science and Technology (UNIST), Ulsan 44919, Republic of Korea
| | - Jihye Lee
- Department of Biological Science, Ulsan National Institute of Science and Technology (UNIST), Ulsan 44919, Republic of Korea
| | - Suhyun Kim
- Department of Biological Science, Ulsan National Institute of Science and Technology (UNIST), Ulsan 44919, Republic of Korea
| | - Seyun Shin
- Department of Biological Science, Ulsan National Institute of Science and Technology (UNIST), Ulsan 44919, Republic of Korea
| | - Young-Eui Kim
- Division of Acute Viral Disease, Center for Emerging Virus Research, National Institute of Infectious Diseases, Korea National Institute of Health, Cheongju 28159, Republic of Korea
| | - Sehee Park
- Division of Acute Viral Disease, Center for Emerging Virus Research, National Institute of Infectious Diseases, Korea National Institute of Health, Cheongju 28159, Republic of Korea
| | - SangJoon Lee
- Department of Biological Science, Ulsan National Institute of Science and Technology (UNIST), Ulsan 44919, Republic of Korea
- Graduate School of Health Science and Technology, Ulsan National Institute of Science and Technology (UNIST), Ulsan 44919, Republic of Korea
| |
Collapse
|
37
|
Mishra S, Jain D, Dey AA, Nagaraja S, Srivastava M, Khatun O, Balamurugan K, Anand M, Ashok AK, Tripathi S, Ganji M, Kesavardhana S. Bat RNA viruses employ viral RHIMs orchestrating species-specific cell death programs linked to Z-RNA sensing and ZBP1-RIPK3 signaling. iScience 2024; 27:111444. [PMID: 39697597 PMCID: PMC11652944 DOI: 10.1016/j.isci.2024.111444] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2023] [Revised: 01/06/2024] [Accepted: 11/18/2024] [Indexed: 12/20/2024] Open
Abstract
RHIM is a protein motif facilitating the assembly of large signaling complexes triggering regulated cell death. A few DNA viruses employ viral RHIMs mimicking host RHIMs and counteract cell death by interacting with host RHIM-proteins to alleviate antiviral defenses. Whether RNA viruses operate such viral RHIMs remains unknown. Here, we identified viral RHIMs in Nsp13 of SARS-CoV-2 and other bat RNA viruses, providing the basis for bats as the hosts for their evolution. Nsp13 promoted viral RHIM and RNA-binding channel-dependent cell death. However, Nsp13 viral RHIM is more critical for human cell death than in bat-derived Tb1 Lu cells, suggesting species-specific regulation. Nsp13 showed RHIM-dependent interactions with ZBP1 and RIPK3, forming large complexes and promoting ZBP1-RIPK3 signaling-mediated cell death. Intriguingly, the SARS-CoV-2 genome consisted of Z-RNA-forming segments promoting Nsp13-dependent cell death. Our findings reveal the functional viral RHIMs of bat-originated RNA viruses regulating host cell death associated with ZBP1-RIPK3 signaling, indicating possible mechanisms of cellular damage and cytokine storm in bat-originated RNA virus infections.
Collapse
Affiliation(s)
- Sanchita Mishra
- Department of Biochemistry, Division of Biological Sciences, Indian Institute of Science, Bengaluru, Karnataka 560012, India
| | - Disha Jain
- Department of Biochemistry, Division of Biological Sciences, Indian Institute of Science, Bengaluru, Karnataka 560012, India
| | - Ayushi Amin Dey
- Department of Biochemistry, Division of Biological Sciences, Indian Institute of Science, Bengaluru, Karnataka 560012, India
| | - Sahana Nagaraja
- Department of Biochemistry, Division of Biological Sciences, Indian Institute of Science, Bengaluru, Karnataka 560012, India
| | - Mansi Srivastava
- Department of Biochemistry, Division of Biological Sciences, Indian Institute of Science, Bengaluru, Karnataka 560012, India
| | - Oyahida Khatun
- Department of Microbiology and Cell Biology, Division of Biological Sciences, Indian Institute of Science, Bengaluru 560012, India
- Centre for Infectious Disease Research, Indian Institute of Science, Bengaluru 560012, India
| | - Keerthana Balamurugan
- Department of Biochemistry, Division of Biological Sciences, Indian Institute of Science, Bengaluru, Karnataka 560012, India
| | - Micky Anand
- Department of Biochemistry, Division of Biological Sciences, Indian Institute of Science, Bengaluru, Karnataka 560012, India
| | - Avinash Karkada Ashok
- Department of Biochemistry, Division of Biological Sciences, Indian Institute of Science, Bengaluru, Karnataka 560012, India
| | - Shashank Tripathi
- Department of Microbiology and Cell Biology, Division of Biological Sciences, Indian Institute of Science, Bengaluru 560012, India
- Centre for Infectious Disease Research, Indian Institute of Science, Bengaluru 560012, India
| | - Mahipal Ganji
- Department of Biochemistry, Division of Biological Sciences, Indian Institute of Science, Bengaluru, Karnataka 560012, India
| | - Sannula Kesavardhana
- Department of Biochemistry, Division of Biological Sciences, Indian Institute of Science, Bengaluru, Karnataka 560012, India
| |
Collapse
|
38
|
Ma Z, Liu D, Cao W, Guo L, Liu K, Bai J, Li X, Jiang P, Liu X. Suppression of ZBP1-mediated NLRP3 inflammasome by the tegument protein VP22 facilitates pseudorabies virus infection. mBio 2024; 15:e0194524. [PMID: 39475237 PMCID: PMC11633114 DOI: 10.1128/mbio.01945-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Accepted: 10/07/2024] [Indexed: 12/12/2024] Open
Abstract
The interaction between Z-DNA binding protein 1 (ZBP1) and the NLR family pyrin domain-containing 3 (NLRP3) inflammasome has been uncovered in several viral infections. However, the role of this molecular pathway during infection with the alpha-herpesvirus pseudorabies virus (PRV) remains largely elusive. Here, we report that during PRV infection, ZBP1-mediated NLRP3 inflammasome activation is inhibited by the viral tegument protein VP22, thereby facilitating viral infection. Through a combination of RNA sequencing and genetic studies, we demonstrate that PRV VP22 functions as a virus-encoded virulence factor by evading the inhibitory effects of ZBP1 on virus infection. Importantly, the replication and pathogenicity of a recombinant PRV lacking VP22 are significantly increased in ZBP1-deficient cells and mice. Mechanistically, PRV VP22 interacts with ZBP1, impeding the recruitment of receptor-interacting protein kinase 3 and Caspase-8, thereby inhibiting NLRP3 activation. Furthermore, we show that the N-terminal 1-50 amino acid domain of VP22 dominantly destabilizes ZBP1-mediated function. Taken together, these findings identify a functional link between PRV infection and ZBP1-mediated NLRP3 inflammatory response, providing novel insights into the pathogenesis of PRV and other herpesviruses. IMPORTANCE Z-DNA binding protein 1 (ZBP1) functions as a pivotal innate immune sensor that regulates inflammatory cell death during viral infections. However, its role in pseudorabies virus (PRV) infection remains unknown. Here, we demonstrate that ZBP1 serves as a restrictive factor by triggering the activation of the NLR family pyrin domain-containing 3 inflammasome, a process counteracted by PRV-encoded protein VP22. Furthermore, VP22 interferes with the interaction between ZBP1 and receptor-interacting protein kinase 3/Caspase-8, particularly through its N-terminal 1-50 amino acids. Importantly, deficiency in ZBP1 enhances the replication and virulence of recombinant viruses lacking VP22 or its N-terminal 1-50 amino acids. These findings reveal how PRV escapes ZBP1-mediated inflammatory responses during infection, potentially informing the rational design of therapeutic interventions.
Collapse
Affiliation(s)
- Zicheng Ma
- Key Laboratory of Animal Diseases Diagnostic and Immunology, Ministry of Agriculture, MOE International Joint Collaborative Research Laboratory for Animal Health & Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Depeng Liu
- Key Laboratory of Animal Diseases Diagnostic and Immunology, Ministry of Agriculture, MOE International Joint Collaborative Research Laboratory for Animal Health & Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Wandi Cao
- Key Laboratory of Animal Diseases Diagnostic and Immunology, Ministry of Agriculture, MOE International Joint Collaborative Research Laboratory for Animal Health & Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Lei Guo
- Key Laboratory of Animal Diseases Diagnostic and Immunology, Ministry of Agriculture, MOE International Joint Collaborative Research Laboratory for Animal Health & Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Kesen Liu
- Key Laboratory of Animal Diseases Diagnostic and Immunology, Ministry of Agriculture, MOE International Joint Collaborative Research Laboratory for Animal Health & Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Juan Bai
- Key Laboratory of Animal Diseases Diagnostic and Immunology, Ministry of Agriculture, MOE International Joint Collaborative Research Laboratory for Animal Health & Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, China
| | - Xingyi Li
- School of Computer Science, Northwestern Polytechnical University, Xi'an, Shanxi, China
| | - Ping Jiang
- Key Laboratory of Animal Diseases Diagnostic and Immunology, Ministry of Agriculture, MOE International Joint Collaborative Research Laboratory for Animal Health & Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, China
| | - Xing Liu
- Key Laboratory of Animal Diseases Diagnostic and Immunology, Ministry of Agriculture, MOE International Joint Collaborative Research Laboratory for Animal Health & Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, China
| |
Collapse
|
39
|
Topper MJ, Guarnieri JW, Haltom JA, Chadburn A, Cope H, Frere J, An J, Borczuk A, Sinha S, Kim J, Park J, Butler D, Meydan C, Foox J, Bram Y, Richard SA, Epsi NJ, Agan B, Chenoweth JG, Simons MP, Tribble D, Burgess T, Dalgard C, Heise MT, Moorman NJ, Baxter VK, Madden EA, Taft-Benz SA, Anderson EJ, Sanders WA, Dickmander RJ, Beigel K, Widjaja GA, Janssen KA, Lie T, Murdock DG, Angelin A, Soto Albrecht YE, Olali AZ, Cen Z, Dybas J, Priebe W, Emmett MR, Best SM, Kelsey Johnson M, Trovao NS, Clark KB, Zaksas V, Meller R, Grabham P, Schisler JC, Moraes-Vieira PM, Pollett S, Mason CE, Syrkin Wurtele E, Taylor D, Schwartz RE, Beheshti A, Wallace DC, Baylin SB. Lethal COVID-19 associates with RAAS-induced inflammation for multiple organ damage including mediastinal lymph nodes. Proc Natl Acad Sci U S A 2024; 121:e2401968121. [PMID: 39602262 PMCID: PMC11626201 DOI: 10.1073/pnas.2401968121] [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: 02/15/2024] [Accepted: 10/07/2024] [Indexed: 11/29/2024] Open
Abstract
Lethal COVID-19 outcomes are attributed to classic cytokine storm. We revisit this using RNA sequencing of nasopharyngeal and 40 autopsy samples from patients dying of SARS-CoV-2. Subsets of the 100 top-upregulated genes in nasal swabs are upregulated in the heart, lung, kidney, and liver, but not mediastinal lymph nodes. Twenty-two of these are "noncanonical" immune genes, which we link to components of the renin-angiotensin-activation-system that manifest as increased fibrin deposition, leaky vessels, thrombotic tendency, PANoptosis, and mitochondrial dysfunction. Immunohistochemistry of mediastinal lymph nodes reveals altered architecture, excess collagen deposition, and pathogenic fibroblast infiltration. Many of the above findings are paralleled in animal models of SARS-CoV-2 infection and human peripheral blood mononuclear and whole blood samples from individuals with early and later SARS-CoV-2 variants. We then redefine cytokine storm in lethal COVID-19 as driven by upstream immune gene and mitochondrial signaling producing downstream RAAS (renin-angiotensin-aldosterone system) overactivation and organ damage, including compromised mediastinal lymph node function.
Collapse
Affiliation(s)
- Michael J. Topper
- COVID-19 International Research Team, Medford, MA02155
- Department of Oncology, The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD21287
| | - Joseph W. Guarnieri
- COVID-19 International Research Team, Medford, MA02155
- The Children’s Hospital of Philadelphia, Philadelphia, PA19104
- Center for Mitochondrial and Epigenomic Medicine, The Children’s Hospital of Philadelphia, Philadelphia, PA19104
| | - Jeffrey A. Haltom
- COVID-19 International Research Team, Medford, MA02155
- The Children’s Hospital of Philadelphia, Philadelphia, PA19104
- Center for Mitochondrial and Epigenomic Medicine, The Children’s Hospital of Philadelphia, Philadelphia, PA19104
| | - Amy Chadburn
- Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY10065
| | - Henry Cope
- School of Medicine, University of Nottingham, DerbyDE22 3DT, United Kingdom
| | - Justin Frere
- Icahn School of Medicine, Mount Sinai, New York, NY10023
| | - Julia An
- COVID-19 International Research Team, Medford, MA02155
- Department of Oncology, The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD21287
| | | | | | | | | | | | - Cem Meydan
- Weill Cornell Medicine, New York, NY10065
| | | | - Yaron Bram
- Weill Cornell Medicine, New York, NY10065
| | - Stephanie A. Richard
- Infectious Disease Clinical Research Program, Department of Preventive Medicine and Biostatistics, Uniformed Services University, Bethesda, MD20814
- Henry M. Jackson Foundation for the Advancement of Military Medicine Inc., Bethesda, MD20817
| | - Nusrat J. Epsi
- Infectious Disease Clinical Research Program, Department of Preventive Medicine and Biostatistics, Uniformed Services University, Bethesda, MD20814
- Henry M. Jackson Foundation for the Advancement of Military Medicine Inc., Bethesda, MD20817
| | - Brian Agan
- Infectious Disease Clinical Research Program, Department of Preventive Medicine and Biostatistics, Uniformed Services University, Bethesda, MD20814
- Henry M. Jackson Foundation for the Advancement of Military Medicine Inc., Bethesda, MD20817
| | - Josh G. Chenoweth
- Henry M. Jackson Foundation for the Advancement of Military Medicine Inc., Bethesda, MD20817
| | - Mark P. Simons
- Infectious Disease Clinical Research Program, Department of Preventive Medicine and Biostatistics, Uniformed Services University, Bethesda, MD20814
| | - David Tribble
- Infectious Disease Clinical Research Program, Department of Preventive Medicine and Biostatistics, Uniformed Services University, Bethesda, MD20814
| | - Timothy Burgess
- Infectious Disease Clinical Research Program, Department of Preventive Medicine and Biostatistics, Uniformed Services University, Bethesda, MD20814
| | - Clifton Dalgard
- Department of Anatomy, Physiology & Genetics, Uniformed Services University, Bethesda, MD20814
| | | | | | | | | | | | | | | | | | - Katherine Beigel
- COVID-19 International Research Team, Medford, MA02155
- The Children’s Hospital of Philadelphia, Philadelphia, PA19104
- Department of Biomedical and Health, The Children’s Hospital of Philadelphia, Philadelphia, PA19104
| | - Gabrielle A. Widjaja
- COVID-19 International Research Team, Medford, MA02155
- The Children’s Hospital of Philadelphia, Philadelphia, PA19104
- Center for Mitochondrial and Epigenomic Medicine, The Children’s Hospital of Philadelphia, Philadelphia, PA19104
| | - Kevin A. Janssen
- COVID-19 International Research Team, Medford, MA02155
- The Children’s Hospital of Philadelphia, Philadelphia, PA19104
- Center for Mitochondrial and Epigenomic Medicine, The Children’s Hospital of Philadelphia, Philadelphia, PA19104
| | - Timothy Lie
- COVID-19 International Research Team, Medford, MA02155
- The Children’s Hospital of Philadelphia, Philadelphia, PA19104
- Center for Mitochondrial and Epigenomic Medicine, The Children’s Hospital of Philadelphia, Philadelphia, PA19104
| | - Deborah G. Murdock
- COVID-19 International Research Team, Medford, MA02155
- The Children’s Hospital of Philadelphia, Philadelphia, PA19104
- Center for Mitochondrial and Epigenomic Medicine, The Children’s Hospital of Philadelphia, Philadelphia, PA19104
| | - Alessia Angelin
- COVID-19 International Research Team, Medford, MA02155
- The Children’s Hospital of Philadelphia, Philadelphia, PA19104
- Center for Mitochondrial and Epigenomic Medicine, The Children’s Hospital of Philadelphia, Philadelphia, PA19104
| | - Yentli E. Soto Albrecht
- COVID-19 International Research Team, Medford, MA02155
- The Children’s Hospital of Philadelphia, Philadelphia, PA19104
- Center for Mitochondrial and Epigenomic Medicine, The Children’s Hospital of Philadelphia, Philadelphia, PA19104
- The University of Pennsylvania, Philadelphia, PA19104
| | - Arnold Z. Olali
- COVID-19 International Research Team, Medford, MA02155
- The Children’s Hospital of Philadelphia, Philadelphia, PA19104
- Center for Mitochondrial and Epigenomic Medicine, The Children’s Hospital of Philadelphia, Philadelphia, PA19104
| | - Zimu Cen
- COVID-19 International Research Team, Medford, MA02155
- The Children’s Hospital of Philadelphia, Philadelphia, PA19104
- Center for Mitochondrial and Epigenomic Medicine, The Children’s Hospital of Philadelphia, Philadelphia, PA19104
| | - Joseph Dybas
- COVID-19 International Research Team, Medford, MA02155
- The Children’s Hospital of Philadelphia, Philadelphia, PA19104
| | - Waldemar Priebe
- COVID-19 International Research Team, Medford, MA02155
- University of Texas Monroe Dunaway Anderson Cancer Center, Houston, TX77030
| | - Mark R. Emmett
- COVID-19 International Research Team, Medford, MA02155
- University of Texas Medical Branch, Galveston, TX77555
| | - Sonja M. Best
- COVID-19 International Research Team, Medford, MA02155
- Innate Immunity and Pathogenesis Section, Laboratory of Neurological Infections and Immunity, National Institute of Allergy and Infectious Diseases, NIH, Rocky Mountain Laboratories, Hamilton, MT59840
| | - Maya Kelsey Johnson
- COVID-19 International Research Team, Medford, MA02155
- Department of Oncology, The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD21287
| | - Nidia S. Trovao
- COVID-19 International Research Team, Medford, MA02155
- Fogarty International Center, NIH, Bethesda, MD20892
| | - Kevin B. Clark
- COVID-19 International Research Team, Medford, MA02155
- Cures Within Reach, Chicago, IL60602
- Champions Service, Computational Sciences Support Network, Multi-Tier Assistance, Training, and Computational Help Track, NSF's Advanced Cyberinfrastructure Coordination Ecosystem: Services and Support, Carnegie-Mellon University, Pittsburgh, PA15213
| | - Victoria Zaksas
- COVID-19 International Research Team, Medford, MA02155
- Center for Translational Data Science, University of Chicago, Chicago, IL60615
- Clever Research Lab, Springfield, IL62704
| | - Robert Meller
- COVID-19 International Research Team, Medford, MA02155
- Morehouse School of Medicine, Atlanta, GA30310
| | - Peter Grabham
- COVID-19 International Research Team, Medford, MA02155
- Center for Radiological Research, College of Physicians and Surgeons, Columbia University, New York, NY19103
| | - Jonathan C. Schisler
- COVID-19 International Research Team, Medford, MA02155
- University of North Carolina, Chapel Hill, NC27599
| | - Pedro M. Moraes-Vieira
- COVID-19 International Research Team, Medford, MA02155
- Laboratory of Immunometabolism, Department of Genetics, Evolution, Microbiology and Immunology, Institute of Biology, University of Campinas, Campinas, São Paulo, Brazil13083-862
| | - Simon Pollett
- Infectious Disease Clinical Research Program, Department of Preventive Medicine and Biostatistics, Uniformed Services University, Bethesda, MD20814
- Henry M. Jackson Foundation for the Advancement of Military Medicine Inc., Bethesda, MD20817
| | - Christopher E. Mason
- COVID-19 International Research Team, Medford, MA02155
- Weill Cornell Medicine, New York, NY10065
- New York Genome Center, New York, NY10013
| | - Eve Syrkin Wurtele
- COVID-19 International Research Team, Medford, MA02155
- Center for Metabolic Biology, Bioinformatics and Computational Biology, and Genetics Development, and Cell Biology, Iowa State University, Ames, IA50011
- Center for Bioinformatics and Computational Biology Iowa State University, Ames, IA50011
- Center for Genetics Development, and Cell Biology Iowa State University, Ames, IA50011
| | - Deanne Taylor
- COVID-19 International Research Team, Medford, MA02155
- The Children’s Hospital of Philadelphia, Philadelphia, PA19104
- Center for Mitochondrial and Epigenomic Medicine, The Children’s Hospital of Philadelphia, Philadelphia, PA19104
- Department of Biomedical and Health, The Children’s Hospital of Philadelphia, Philadelphia, PA19104
- Department of Pediatrics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA19104
| | - Robert E. Schwartz
- COVID-19 International Research Team, Medford, MA02155
- Weill Cornell Medicine, New York, NY10065
| | - Afshin Beheshti
- COVID-19 International Research Team, Medford, MA02155
- Stanley Center for Psychiatric Research, Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA02142
- Blue Marble Space Institute of Science, Seattle, WA98104
- McGowan Institute for Regenerative Medicine and Center for Space Biomedicine, Department of Surgery, University of Pittsburgh, Pittsburgh, PA15219
| | - Douglas C. Wallace
- COVID-19 International Research Team, Medford, MA02155
- The Children’s Hospital of Philadelphia, Philadelphia, PA19104
- Center for Mitochondrial and Epigenomic Medicine, The Children’s Hospital of Philadelphia, Philadelphia, PA19104
- Division of Human Genetics, Department of Pediatrics, University of Pennsylvania, Philadelphia, PA19104
| | - Stephen B. Baylin
- COVID-19 International Research Team, Medford, MA02155
- Department of Oncology, The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD21287
- Van Andel Institute, Grand Rapids, MI49503
| |
Collapse
|
40
|
Hu XM, Zheng S, Zhang Q, Wan X, Li J, Mao R, Yang R, Xiong K. PANoptosis signaling enables broad immune response in psoriasis: From pathogenesis to new therapeutic strategies. Comput Struct Biotechnol J 2024; 23:64-76. [PMID: 38125299 PMCID: PMC10730955 DOI: 10.1016/j.csbj.2023.11.049] [Citation(s) in RCA: 21] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Revised: 11/24/2023] [Accepted: 11/24/2023] [Indexed: 12/23/2023] Open
Abstract
BACKGROUND Accumulating evidence suggests that regulated cell death, such as pyroptosis, apoptosis, and necroptosis, is deeply involved in the pathogenesis of psoriasis. As a newly recognized form of systematic cell death, PANoptosis is involved in a variety of inflammatory disorders through amplifying inflammatory and immune cascades, but its role in psoriasis remains elusive. OBJECTIVES To reveal the role of PANoptosis in psoriasis for a potential therapeutic strategy. METHODS Multitranscriptomic analysis and experimental validation were used to identify PANoptosis signaling in psoriasis. RNA-seq and scRNA-seq analyses were performed to establish a PANoptosis-mediated immune response in psoriasis, which revealed hub genes through WGCNA and predicted disulfiram as a potential drug. The effect and mechanism of disulfiram were verified in imiquimod (IMQ)-induced psoriasis. RESULTS Here, we found a highlighted PANoptosis signature in psoriasis patients through multitranscriptomic analysis and experimental validation. Based on this, two distinct PANoptosis patterns (non/high) were identified, which were the options for clinical classification. The high-PANoptosis-related group had a higher response rate to immune cell infiltration (such as M1 macrophages and keratinocytes). Subsequently, WGCNA showed the hub genes (e.g., S100A12, CYCS, NOD2, STAT1, HSPA4, AIM2, MAPK7), which were significantly associated with clinical phenotype, PANoptosis signature, and identified immune response in psoriasis. Finally, we explored disulfiram (DSF) as a candidate drug for psoriasis through network pharmacology, which ameliorated IMQ-mediated psoriatic symptoms through antipyroptosis-mediated inflammation and enhanced apoptotic progression. By analyzing the specific ligand-receptor interaction pairs within and between cell lineages, we speculated that DSF might exert its effects by targeting keratinocytes directly or targeting M1 macrophages to downregulate the proliferation of keratinocytes. CONCLUSIONS PANoptosis with its mediated immune cell infiltration provides a roadmap for research on the pathogenesis and therapeutic strategies of psoriasis.
Collapse
Affiliation(s)
- Xi-min Hu
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha 410008, China
- Department of Anatomy and Neurobiology, School of Basic Medical Science, Central South University, Changsha 410013, China
| | - Shengyuan Zheng
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Qi Zhang
- Department of Anatomy and Neurobiology, School of Basic Medical Science, Central South University, Changsha 410013, China
| | - Xinxing Wan
- Department of Endocrinology, Third Xiangya Hospital, Central South University, Changsha 410013, China
| | - Ji Li
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha 410008, China
- Hunan Key Laboratory of Aging Biology, Xiangya Hospital, Central South University, Changsha 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Rui Mao
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Ronghua Yang
- Department of Burn and Plastic Surgery, Guangzhou First People's Hospital, South China University of Technology, Guangzhou 510000, China
| | - Kun Xiong
- Department of Anatomy and Neurobiology, School of Basic Medical Science, Central South University, Changsha 410013, China
- Hunan Key Laboratory of Ophthalmology, Xiangya Hospital, Central South University, Changsha 410008, China
- Key Laboratory of Emergency and Trauma, Ministry of Education, College of Emergency and Trauma, Hainan Medical University, Haikou 571199, China
| |
Collapse
|
41
|
Shi R, Liang R, Wang F, Wang L, Zidai W, Zhang J, Min L, Du X, Sun S, Xiao C, Li C, Liang X, Chen AF, Yang W. Identification and experimental validation of PYCARD as a crucial PANoptosis-related gene for immune response and inflammation in COPD. Apoptosis 2024; 29:2091-2107. [PMID: 38652339 DOI: 10.1007/s10495-024-01961-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/15/2024] [Indexed: 04/25/2024]
Abstract
Chronic inflammatory and immune responses play key roles in the development and progression of chronic obstructive pulmonary disease (COPD). PANoptosis, as a unique inflammatory cell death modality, is involved in the pathogenesis of many inflammatory diseases. We aim to identify critical PANoptosis-related biomarkers and explore their potential effects on respiratory tract diseases and immune infiltration landscapes in COPD. Total microarray data consisting of peripheral blood and lung tissue datasets associated with COPD were obtained from the GEO database. PANoptosis-associated genes in COPD were identified by intersecting differentially expressed genes (DEGs) with genes involved in pyroptosis, apoptosis, and necroptosis after normalizing and removing the batch effect. Furthermore, GO, KEGG, PPI network, WGCNA, LASSO-COX, and ROC curves analysis were conducted to screen and verify hub genes, and the correlation between PYCARD and infiltrated immune cells was analyzed. The effect of PYCARD on respiratory tract diseases and the potential small-molecule agents for the treatment of COPD were identified. PYCARD expression was verified in the lung tissue of CS/LPS-induced COPD mice. PYCARD was a critical PANoptosis-related gene in all COPD patients. PYCARD was positively related to NOD-like receptor signaling pathway and promoted immune cell infiltration. Moreover, PYCARD was significantly activated in COPD mice mainly by targeting PANoptosis. PANoptosis-related gene PYCARD is a potential biomarker for COPD diagnosis and treatment.
Collapse
Affiliation(s)
- Rui Shi
- Yunnan Key Laboratory of Pharmacology for Natural Products, Kunming Medical University, Kunming, China
| | - Renwen Liang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Fang Wang
- Department of Emergency, The First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Lueli Wang
- Yunnan Key Laboratory of Pharmacology for Natural Products, Kunming Medical University, Kunming, China
| | - Wuyi Zidai
- Yunnan Key Laboratory of Pharmacology for Natural Products, Kunming Medical University, Kunming, China
| | - Jie Zhang
- Yunnan Key Laboratory of Pharmacology for Natural Products, Kunming Medical University, Kunming, China
| | - Luo Min
- Yunnan Key Laboratory of Pharmacology for Natural Products, Kunming Medical University, Kunming, China
| | - Xiaohua Du
- Department of Respiratory and Critical Care Medicine, First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Shibo Sun
- Department of Pulmonary and Critical Care Medicine, First Affiliated Hospital, Kunming Medical University, Kunming, China
| | - Chuang Xiao
- Yunnan Key Laboratory of Pharmacology for Natural Products, Kunming Medical University, Kunming, China
| | - Chaozhong Li
- Department of Emergency, The First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Xuewu Liang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China.
| | - Alex F Chen
- Institute for Developmental and Regenerative Cardiovascular Medicine, Xinhua Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai, China.
| | - Weimin Yang
- Yunnan Key Laboratory of Pharmacology for Natural Products, Kunming Medical University, Kunming, China.
| |
Collapse
|
42
|
Ye Z, Deng M, Yang Y, Song Y, Weng L, Qi W, Ding P, Huang Y, Yu C, Wang Y, Wu Y, Zhang Y, Yuan S, Nie W, Zhang L, Zeng C. Epithelial mitochondrial fission-mediated PANoptosis is crucial for ulcerative colitis and its inhibition by saquinavir through Drp1. Pharmacol Res 2024; 210:107538. [PMID: 39643069 DOI: 10.1016/j.phrs.2024.107538] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Revised: 12/01/2024] [Accepted: 12/03/2024] [Indexed: 12/09/2024]
Abstract
Ulcerative colitis (UC) is characterized by increased cell death in intestinal epithelial cell (IEC), which compromises gut barrier function and activates inflammation. Aberrant mitochondrial dynamics have been implicated in various forms of cell death, but it is currently unclear if they play a role in IEC death and colitis pathogenesis. This study aims to investigate the contribution of aberrant mitochondrial dynamics to colitis progression using cellular models, animal models, and clinical samples. The results revealed that IEC in mice with Dextran sulfate sodium salt (DSS)-induced colitis exhibited dynamin-related protein 1 (Drp1)-mediated mitochondrial fission and Z-DNA binding protein 1 (ZBP1)-dependent PANoptosis, which is a combination of apoptosis, necroptosis, and pyroptosis. However, these processes and the pathogenesis of DSS-induced colitis were significantly attenuated in IEC-specific Drp1 heterozygous knockout mice. Importantly, ZBP1-PANoptosis and Drp1-mediated mitochondrial fission were observed in IEC of UC patients, exhibiting a positive correlation with disease severity. Mechanistically, hyperactivated mitochondrial fission induced mitochondrial reactive oxygen species production leading to PANoptosis through ZBP1 sulfenylation at Cys327 independently of its Zα domain. Saquinavir, an FDA-approved drug identified through in-silico screening alongside in vivo and in vitro experiments, inhibits mitochondrial fission thereby enhancing therapeutic efficacy in mice with colitis.
Collapse
Affiliation(s)
- Zhiming Ye
- Center for Drug Research and Development, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Mingxia Deng
- The Guangzhou Laboratory, Guangzhou 510000, China
| | - Yang Yang
- Department of Pharmacy, Zhuhai People's Hospital (Zhuhai hospital affiliated with Jinan University), Zhuhai 519000, China; School of Pharmacy, Faculty of Medicine, Macau University of Science and Technology, Macao
| | - Yuanming Song
- Center for Drug Research and Development, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Liangkun Weng
- Center for Drug Research and Development, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Wanchen Qi
- Department of Pharmacy, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou 519000, China
| | - Ping Ding
- Center for Drug Research and Development, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Yihang Huang
- College of Pharmacy, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Can Yu
- Center for Drug Research and Development, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Yan Wang
- College of Nursing, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Yixing Wu
- College of Nursing, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Yan Zhang
- College of Pharmacy, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Shaoying Yuan
- College of Nursing, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Wenkai Nie
- College of Pharmacy, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Luyong Zhang
- Center for Drug Research and Development, Guangdong Pharmaceutical University, Guangzhou 510006, China; Jiangsu Key Laboratory of Drug Screening, China Pharmaceutical University, Nanjing 210009, China
| | - Cheng Zeng
- Center for Drug Research and Development, Guangdong Pharmaceutical University, Guangzhou 510006, China; Key specialty of Clinical Pharmacy, The first Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou 510699, China.
| |
Collapse
|
43
|
Mocarski ES. Cytomegalovirus Biology Viewed Through a Cell Death Suppression Lens. Viruses 2024; 16:1820. [PMID: 39772130 PMCID: PMC11680106 DOI: 10.3390/v16121820] [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: 10/28/2024] [Revised: 11/22/2024] [Accepted: 11/22/2024] [Indexed: 01/11/2025] Open
Abstract
Cytomegaloviruses, species-specific members of the betaherpesviruses, encode an impressive array of immune evasion strategies committed to the manipulation of the host immune system enabling these viruses to remain for life in a stand-off with host innate and adaptive immune mechanisms. Even though they are species-restricted, cytomegaloviruses are distributed across a wide range of different mammalian species in which they cause systemic infection involving many different cell types. Regulated, or programmed cell death has a recognized potential to eliminate infected cells prior to completion of viral replication and release of progeny. Cell death also naturally terminates replication during the final stages of replication. Over the past two decades, the host defense potential of known programmed cell death pathways (apoptosis, necroptosis, and pyroptosis), as well as a novel mitochondrial serine protease pathway have been defined through studies of cytomegalovirus-encoded cell death suppressors. Such virus-encoded inhibitors prevent virus-induced, cytokine-induced, and stress-induced death of infected cells while also moderating inflammation. By evading cell death and consequent inflammation as well as innate and adaptive immune clearance, cytomegaloviruses represent successful pathogens that become a critical disease threat when the host immune system is compromised. This review will discuss cell death programs acquired for mammalian host defense against cytomegaloviruses and enumerate the range of modulatory strategies this type of virus employs to balance host defense in favor of lifelong persistence.
Collapse
Affiliation(s)
- Edward S. Mocarski
- Department of Microbiology & Immunology, Stanford Medical School, Stanford University, Stanford, CA 94305, USA;
- Department of Microbiology & Immunology, Emory Medical School, Emory Vaccine Center, Emory University, Atlanta, GA 30322, USA
| |
Collapse
|
44
|
Jia Y, Liu Y, Zuo Y, Zhang J, Li Y, Liu X, Lv S. The Potential Therapeutic Prospect of PANoptosis in Heart Failure. J Inflamm Res 2024; 17:9147-9168. [PMID: 39583864 PMCID: PMC11585275 DOI: 10.2147/jir.s485901] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2024] [Accepted: 11/15/2024] [Indexed: 11/26/2024] Open
Abstract
Heart failure (HF) represents a serious manifestation or advanced stage of various cardiac diseases. HF continues to impose a significant global disease burden, characterized by high rates of hospitalization and fatality. Furthermore, the pathogenesis and pathophysiological processes underlying HF remain incompletely understood, complicating its prevention and treatment strategies. One significant pathophysiological mechanism associated with HF is the systemic inflammatory response. PANoptosis, a novel mode of inflammatory cell death, has been extensively studied in the context of infectious diseases, neurodegenerative disorders, cancers, and other inflammatory conditions. Recent investigations have revealed that PANoptosis-related genes are markedly dysregulated in HF specimens. Consequently, the PANoptosis-mediated inflammatory response may represent a potential mechanism and therapeutic target for HF. This paper conducts a comprehensive analysis of the molecular pathways that drive PANoptosis. We discuss its role and potential therapeutic targets in HF, thereby providing valuable insights for clinical treatment and the development of novel therapies.
Collapse
Affiliation(s)
- Yunfeng Jia
- Department of Geriatrics, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine (National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion), Tianjin, 300381, People’s Republic of China
| | - Yayi Liu
- Department of Geriatrics, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine (National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion), Tianjin, 300381, People’s Republic of China
| | - Yiming Zuo
- Department of Geriatrics, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine (National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion), Tianjin, 300381, People’s Republic of China
| | - Junping Zhang
- Department of Geriatrics, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine (National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion), Tianjin, 300381, People’s Republic of China
| | - Yanyang Li
- Department of Integrated Traditional and Western Medicine, Tianjin Medical University Cancer Institute and Hospital, Tianjin, 300060, People’s Republic of China
- Graduate School, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, People’s Republic of China
| | - Xuezheng Liu
- Department of Geriatrics, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine (National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion), Tianjin, 300381, People’s Republic of China
| | - Shichao Lv
- Department of Geriatrics, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine (National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion), Tianjin, 300381, People’s Republic of China
| |
Collapse
|
45
|
Wang Q, Wei J, He J, Ming S, Li X, Huang X, Hong Z, Wu Y. HSP70 contributes to pathogenesis of fulminant hepatitis induced by coronavirus. Int Immunopharmacol 2024; 141:112963. [PMID: 39159560 DOI: 10.1016/j.intimp.2024.112963] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Revised: 08/07/2024] [Accepted: 08/15/2024] [Indexed: 08/21/2024]
Abstract
Fulminant viral hepatitis (FH) represents a significant clinical challenge, with its pathogenesis not yet fully elucidated. Heat shock protein (HSP)70, a molecular chaperone protein with a broad range of cytoprotective functions, is upregulated in response to stress. However, the role of HSP70 in FH remains to be investigated. Notably, HSP70 expression is upregulated in the livers of coronavirus-infected mice and patients. Therefore, we investigated the mechanistic role of HSP70 in coronavirus-associated FH pathogenesis. FH was induced in HSP70-deficient (HSP70 KO) mice or in WT mice treated with the HSP70 inhibitor VER155008 when infected with the mouse hepatitis virus strain A59 (MHV-A59). MHV-A59-infected HSP70 KO mice exhibited significantly reduced liver damage and mortality. This effect was attributed to decreased infiltration of monocyte-macrophages and neutrophils in the liver of HSP70 KO mice, resulting in lower levels of inflammatory cytokines such as IL-1β, TNFα, and IL-6, and a reduced viral load. Moreover, treatment with the HSP70 inhibitor VER155008 protected mice from MHV-A59-induced liver damage and FH mortality. In summary, HSP70 promotes coronavirus-induced FH pathogenesis by enhancing the infiltration of monocyte-macrophages and neutrophils and promoting the secretion of inflammatory cytokines. Therefore, HSP70 is a potential therapeutic target in viral FH intervention.
Collapse
Affiliation(s)
- Qiaohua Wang
- Center for Infection and Immunity, Guangdong Provincial Engineering Research Center of Molecular Imaging, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong Province 519000, China
| | - Jiayou Wei
- Center for Infection and Immunity, Guangdong Provincial Engineering Research Center of Molecular Imaging, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong Province 519000, China
| | - Jianzhong He
- Department of Pathology, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai 519000, China
| | - Siqi Ming
- Department of Laboratory Medicine, Guangdong Provincial Hospital of Chinese Medicine, Zhuhai, Guangdong Province 519015, China
| | - Xingyu Li
- Center for Infection and Immunity, Guangdong Provincial Engineering Research Center of Molecular Imaging, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong Province 519000, China
| | - Xi Huang
- Center for Infection and Immunity, Guangdong Provincial Engineering Research Center of Molecular Imaging, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong Province 519000, China
| | - Zhongsi Hong
- Center of Infectious Disease, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong Province 519000, China; Key Laboratory of Traditional Chinese Medicine for the Prevention and Treatment of Infectious Diseases, Traditional Chinese Medicine Bureau of Guangdong Province, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai 519000, China.
| | - Yongjian Wu
- Center for Infection and Immunity, Guangdong Provincial Engineering Research Center of Molecular Imaging, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong Province 519000, China; Key Laboratory of Traditional Chinese Medicine for the Prevention and Treatment of Infectious Diseases, Traditional Chinese Medicine Bureau of Guangdong Province, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai 519000, China.
| |
Collapse
|
46
|
Zhang X, Tang B, Luo J, Yang Y, Weng Q, Fang S, Zhao Z, Tu J, Chen M, Ji J. Cuproptosis, ferroptosis and PANoptosis in tumor immune microenvironment remodeling and immunotherapy: culprits or new hope. Mol Cancer 2024; 23:255. [PMID: 39543600 PMCID: PMC11566504 DOI: 10.1186/s12943-024-02130-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2024] [Accepted: 09/19/2024] [Indexed: 11/17/2024] Open
Abstract
Normal life requires cell division to produce new cells, but cell death is necessary to maintain balance. Dysregulation of cell death can lead to the survival and proliferation of abnormal cells, promoting tumor development. Unlike apoptosis, necrosis, and autophagy, the newly recognized forms of regulated cell death (RCD) cuproptosis, ferroptosis, and PANoptosis provide novel therapeutic strategies for tumor treatment. Increasing research indicates that the death of tumor and immune cells mediated by these newly discovered forms of cell death can regulate the tumor microenvironment (TME) and influence the effectiveness of tumor immunotherapy. This review primarily elucidates the molecular mechanisms of cuproptosis, ferroptosis, and PANoptosis and their complex effects on tumor cells and the TME. This review also summarizes the exploration of nanoparticle applications in tumor therapy based on in vivo and in vitro evidence derived from the induction or inhibition of these new RCD pathways.
Collapse
Affiliation(s)
- Xiaojie Zhang
- Zhejiang Key Laboratory of Imaging and Interventional Medicine, Zhejiang Engineering Research Csaenter of Interventional Medicine Engineering and Biotechnology, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui, Zhejiang, 323000, China
| | - Bufu Tang
- Zhejiang Key Laboratory of Imaging and Interventional Medicine, Zhejiang Engineering Research Csaenter of Interventional Medicine Engineering and Biotechnology, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui, Zhejiang, 323000, China
- Department of Radiation Oncology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Jinhua Luo
- Zhejiang Key Laboratory of Imaging and Interventional Medicine, Zhejiang Engineering Research Csaenter of Interventional Medicine Engineering and Biotechnology, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui, Zhejiang, 323000, China
| | - Yang Yang
- Zhejiang Key Laboratory of Imaging and Interventional Medicine, Zhejiang Engineering Research Csaenter of Interventional Medicine Engineering and Biotechnology, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui, Zhejiang, 323000, China
- Key Laboratory of Precision Medicine of Lishui City, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui, 323000, China
- School of Medcine, Clinical College of The Affiliated Central Hospital, Lishui University, Lishui, 323000, China
| | - Qiaoyou Weng
- Zhejiang Key Laboratory of Imaging and Interventional Medicine, Zhejiang Engineering Research Csaenter of Interventional Medicine Engineering and Biotechnology, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui, Zhejiang, 323000, China
- Key Laboratory of Precision Medicine of Lishui City, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui, 323000, China
- School of Medcine, Clinical College of The Affiliated Central Hospital, Lishui University, Lishui, 323000, China
| | - Shiji Fang
- Zhejiang Key Laboratory of Imaging and Interventional Medicine, Zhejiang Engineering Research Csaenter of Interventional Medicine Engineering and Biotechnology, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui, Zhejiang, 323000, China
- Key Laboratory of Precision Medicine of Lishui City, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui, 323000, China
- School of Medcine, Clinical College of The Affiliated Central Hospital, Lishui University, Lishui, 323000, China
| | - Zhongwei Zhao
- Zhejiang Key Laboratory of Imaging and Interventional Medicine, Zhejiang Engineering Research Csaenter of Interventional Medicine Engineering and Biotechnology, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui, Zhejiang, 323000, China
- Key Laboratory of Precision Medicine of Lishui City, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui, 323000, China
- School of Medcine, Clinical College of The Affiliated Central Hospital, Lishui University, Lishui, 323000, China
| | - Jianfei Tu
- Zhejiang Key Laboratory of Imaging and Interventional Medicine, Zhejiang Engineering Research Csaenter of Interventional Medicine Engineering and Biotechnology, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui, Zhejiang, 323000, China.
- Key Laboratory of Precision Medicine of Lishui City, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui, 323000, China.
- School of Medcine, Clinical College of The Affiliated Central Hospital, Lishui University, Lishui, 323000, China.
| | - Minjiang Chen
- Zhejiang Key Laboratory of Imaging and Interventional Medicine, Zhejiang Engineering Research Csaenter of Interventional Medicine Engineering and Biotechnology, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui, Zhejiang, 323000, China.
- Key Laboratory of Precision Medicine of Lishui City, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui, 323000, China.
- School of Medcine, Clinical College of The Affiliated Central Hospital, Lishui University, Lishui, 323000, China.
| | - Jiansong Ji
- Zhejiang Key Laboratory of Imaging and Interventional Medicine, Zhejiang Engineering Research Csaenter of Interventional Medicine Engineering and Biotechnology, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui, Zhejiang, 323000, China.
- Key Laboratory of Precision Medicine of Lishui City, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui, 323000, China.
- School of Medcine, Clinical College of The Affiliated Central Hospital, Lishui University, Lishui, 323000, China.
| |
Collapse
|
47
|
Sun Y, Liu K. Mechanistic Insights into Influenza A Virus-Induced Cell Death and Emerging Treatment Strategies. Vet Sci 2024; 11:555. [PMID: 39591329 PMCID: PMC11598850 DOI: 10.3390/vetsci11110555] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Revised: 10/30/2024] [Accepted: 11/08/2024] [Indexed: 11/28/2024] Open
Abstract
Influenza A virus (IAV) infection initiates a complex interplay of cell death modalities, including apoptosis, necroptosis, pyroptosis, and their integration, known as PANoptosis, which significantly impacts host immune responses and tissue integrity. These pathways are intricately regulated by viral proteins and host factors, contributing to both viral clearance and pathogenesis-related tissue damage. This review comprehensively explores the molecular mechanisms underlying these cell death processes in influenza infection. We highlight the roles of key regulatory proteins, such as ZBP1 (Z-DNA binding protein 1) and RIPK3 (receptor-interacting protein kinase 3), in orchestrating these responses, emphasizing the dual roles of cell death in both antiviral defense and tissue injury. Furthermore, we discuss emerging therapeutic strategies targeting these pathways, aiming to enhance antiviral efficacy while minimizing collateral tissue damage. Future research should focus on targeted approaches to modulate cell death mechanisms, aiming to reduce tissue damage and improve clinical outcomes for patients with severe influenza.
Collapse
Affiliation(s)
- Yuling Sun
- Joint International Research Laboratory of Agriculture and Agri-Product Safety, The Ministry of Education of China, Yangzhou University, Yangzhou 225009, China
- College of Veterinary Medicine, Institute of Comparative Medicine, Yangzhou University, Yangzhou 225009, China
| | - Kaituo Liu
- Joint International Research Laboratory of Agriculture and Agri-Product Safety, The Ministry of Education of China, Yangzhou University, Yangzhou 225009, China
- College of Veterinary Medicine, Institute of Comparative Medicine, Yangzhou University, Yangzhou 225009, China
| |
Collapse
|
48
|
Hou G, Chen Y, Lei H, Lu Y, Liu L, Han Z, Sun S, Li J, Cheng L. Bimetallic peroxide nanoparticles induce PANoptosis by disrupting ion homeostasis for enhanced immunotherapy. SCIENCE ADVANCES 2024; 10:eadp7160. [PMID: 39514658 PMCID: PMC11546811 DOI: 10.1126/sciadv.adp7160] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Accepted: 10/04/2024] [Indexed: 11/16/2024]
Abstract
PANoptosis has recently emerged as a potential approach to improve the immune microenvironment. However, current methods for inducing PANoptosis are limited. Herein, through biological screening, the rational use of the nutrient metal ions Cu2+ and Zn2+ had great potential to induce PANoptosis. Inspired by these findings, we successfully developed hydrazided hyaluronic acid-modified zinc copper oxide (HZCO) nanoparticles as a PANoptosis inducer to potentiate immunotherapy. Bioactive HZCO actively delivered Cu2+ and Zn2+ while disrupting the cellular intrinsic ion metabolism pathway, resulting in double-stranded DNA release and organelle damage in cancer cells. Simultaneously, this process triggered the formation of PANoptosome and the activation of PANoptosis. HZCO-induced PANoptosis inhibited tumor growth and activated potent antitumor immune response, thereby enhancing the effectiveness of anti-programmed cell death 1 therapy. Overall, our work provides an insight into the development of PANoptosis inducers and the design of synergistic immunotherapy strategies.
Collapse
Affiliation(s)
- Guanghui Hou
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Soochow University, Suzhou 215123, China
| | - Youdong Chen
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Soochow University, Suzhou 215123, China
| | - Huali Lei
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Soochow University, Suzhou 215123, China
| | - Yujie Lu
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Soochow University, Suzhou 215123, China
- Institute of State Key Laboratory of Targeting Oncology, National Center for International Research of Bio-targeting Theranostics, Guangxi Key Laboratory of Bio-targeting Theranostics, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Medical University, Nanning, Guangxi 530021, China
| | - Lin Liu
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Soochow University, Suzhou 215123, China
| | - Zhihui Han
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Soochow University, Suzhou 215123, China
| | - Shumin Sun
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Soochow University, Suzhou 215123, China
| | - Jingrui Li
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Soochow University, Suzhou 215123, China
| | - Liang Cheng
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Soochow University, Suzhou 215123, China
| |
Collapse
|
49
|
Zhao Y, Liang L, Jeon JE, Keshavjee S, Liu M. Ischemia/Reperfusion Upregulates Genes Related to PANoptosis in Human Lung Transplants. Transplantation 2024:00007890-990000000-00937. [PMID: 39528335 DOI: 10.1097/tp.0000000000005268] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2024]
Abstract
BACKGROUND Activation of multiple programmed cell death (PCD) pathways has been reported in cellular and animal studies of ischemia/reperfusion injury in lung transplantation. However, the status of these pathways in human lung transplants remains unknown. This study investigates the involvement of PCD pathways and their relationship with inflammation and signaling pathways in human lung transplants. METHODS Transcriptomic analysis was conducted on 54 paired human lung tissue samples at the end of cold preservation time and 2 h after reperfusion, collected between 2008 and 2011. Gene Set Enrichment Analysis (GSEA) and single-sample GSEA were used to examine the activation of genes in 6 PCD pathways. The relationships between PCD pathways and inflammation, as well as signaling pathways, were assessed via single-gene GSEA. RESULTS GSEA results indicated that apoptosis and necroptosis were significantly upregulated after reperfusion in human lung transplants, whereas the gene sets related to pyroptosis, ferroptosis, autophagy, and cuproptosis were not significantly upregulated. Notably, single-sample GSEA demonstrated an intricate interplay among pyroptosis, apoptosis, and necroptosis, collectively referred to as PANoptosis, which is further supported by enrichment of genes related to PANoptosome, inflammatory response, and nuclear factor-κB and interferon signaling pathways, via single-gene GSEA assays. CONCLUSIONS This study demonstrated the genes of PANoptosis are upregulated in human lung grafts during reperfusion. The discovery of PANoptosis as an underlying mechanism of cell death in human lung grafts implies that effective therapeutics to prevent or reduce PANoptosis may alleviate ischemia/reperfusion injury and improve clinical lung transplant outcomes.
Collapse
Affiliation(s)
- Yajin Zhao
- Latner Thoracic Surgery Research Laboratories, Toronto General Hospital Research Institute, University Health Network, Toronto, ON, Canada
| | - Lubiao Liang
- Latner Thoracic Surgery Research Laboratories, Toronto General Hospital Research Institute, University Health Network, Toronto, ON, Canada
- Department of Thoracic Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, China
| | - Jamie E Jeon
- Latner Thoracic Surgery Research Laboratories, Toronto General Hospital Research Institute, University Health Network, Toronto, ON, Canada
- Department of Physiology, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - Shaf Keshavjee
- Latner Thoracic Surgery Research Laboratories, Toronto General Hospital Research Institute, University Health Network, Toronto, ON, Canada
- Institute of Medical Science, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada
- Department of Surgery, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - Mingyao Liu
- Latner Thoracic Surgery Research Laboratories, Toronto General Hospital Research Institute, University Health Network, Toronto, ON, Canada
- Department of Physiology, 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 Surgery, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
50
|
Lou J, Mao Y, Jiang W, Shen H, Fan Y, Yu Q, Zhou C, Wei Z, Zhou K, Jin M, Wu J. TRIM56 Modulates YBX1 Degradation to Ameliorate ZBP1-Mediated Neuronal PANoptosis in Spinal Cord Injury. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2407132. [PMID: 39291396 PMCID: PMC11558135 DOI: 10.1002/advs.202407132] [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: 06/26/2024] [Revised: 09/10/2024] [Indexed: 09/19/2024]
Abstract
Spinal cord injury (SCI) is a severe injury to the central nervous system, and its treatment is always a major medical challenge. Proinflammatory cell death is considered an important factor affecting neuroinflammation and the prognosis after injury. PANoptosis, a newly discovered type of proinflammatory cell death, regulates the activation of executioner molecules of apoptosis, pyroptosis and necroptosis through the PANoptosome, providing a new target for therapeutic intervention after SCI. However, its role and regulatory mechanism in SCI are not yet elucidated. Here, based on proteomic data, YBX1 expression is significantly increased in neurons after SCI. Guided by RIP-seq, subsequent experiments reveal that YBX1 promotes ZBP1 expression by stabilizing the Zbp1 mRNA, thereby aggravating ZBP1-mediated PANoptosis. Furthermore, the E3 ubiquitin ligase TRIM56 is identified as an endogenous inhibitor of YBX1 via molecular docking and IP/MS analysis. Mechanistically, TRIM56 bound to YBX1 and promoted its ubiquitination, thereby accelerating its degradation. Taken together, these findings reveal a novel function of YBX1 in regulating ZBP1-mediated PANoptosis in the pathogenesis of SCI and verified that TRIM56 functions as an endogenous inhibitor to promote the ubiquitin-proteasomal degradation of YBX1, providing new insights into SCI treatment strategies.
Collapse
Affiliation(s)
- Junsheng Lou
- Department of Orthopedic SurgeryThe First Affiliated HospitalZhejiang University School of MedicineNo. 79 Qingchun RoadHangzhou310003China
| | - Yiting Mao
- Obstetrics and Gynecology HospitalInstitute of Reproduction and DevelopmentFudan UniversityShanghai200090China
| | - Wu Jiang
- Department of OrthopeadicsAffiliated Hangzhou First People's HospitalSchool of MedicineWestlake UniversityNo. 261 Huansha RoadHangzhou310006China
| | - Honghao Shen
- Department of Orthopedic SurgeryThe First Affiliated HospitalZhejiang University School of MedicineNo. 79 Qingchun RoadHangzhou310003China
| | - Yunpeng Fan
- Department of Orthopedic SurgeryThe First Affiliated HospitalZhejiang University School of MedicineNo. 79 Qingchun RoadHangzhou310003China
| | - Qing Yu
- Department of Critical Care Medicinethe Second Affiliated Hospital of Zhejiang University School of MedicineHangzhou310009China
| | - Conghui Zhou
- Department of Orthopedic SurgeryThe First Affiliated HospitalZhejiang University School of MedicineNo. 79 Qingchun RoadHangzhou310003China
| | - Ziyao Wei
- Department of Orthopedic SurgeryThe First Affiliated HospitalZhejiang University School of MedicineNo. 79 Qingchun RoadHangzhou310003China
| | - Kailiang Zhou
- Department of OrthopeadicsThe Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical UniversityWenzhou325027China
| | - Mengran Jin
- Department of Orthopedic SurgeryThe First Affiliated HospitalZhejiang University School of MedicineNo. 79 Qingchun RoadHangzhou310003China
| | - Junsong Wu
- Department of Orthopedic SurgeryThe First Affiliated HospitalZhejiang University School of MedicineNo. 79 Qingchun RoadHangzhou310003China
| |
Collapse
|