1
|
Huang R, Hu Y, Wang YF, Zhang S, Wang ZG, Pang DW, Liu SL. Targeted Degradation of ZBP1 with Covalent PROTACs for Anti-Inflammatory Treatment of Infections. Angew Chem Int Ed Engl 2025; 64:e202423524. [PMID: 40013409 DOI: 10.1002/anie.202423524] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2024] [Revised: 02/24/2025] [Accepted: 02/26/2025] [Indexed: 02/28/2025]
Abstract
Z-DNA binding protein 1 (ZBP1) has emerged as a critical pathogen-sensing protein that upon activation, triggers necroptotic signaling cascades, leading to a potent inflammatory response and potentially causing significant tissue damage. However, available drugs specifically developed for the effective inhibition or degradation of ZBP1 is still lacking so far. In this study, we developed a potent covalent recognition-based PROTAC (C-PROTAC) molecule for the degradation of ZBP1. It consists of a DNA aptamer as the recognition moiety and an E3 enzyme-recruiting unit, connected by a linker containing N-acyl-N-alkyl sulfonamides (NASA) groups. The DNA aptamer specifically binds to ZBP1, while the NASA-containing linker facilitates the formation of a covalent bond between the PROTAC and the target protein. The E3 ligase-recruiting unit then directs the ubiquitin-proteasome system to degrade the ZBP1-PROTAC complex. This approach combines the high specificity of DNA aptamers with the efficiency of covalent binding and the degradation-inducing capabilities of PROTACs, providing a powerful tool for targeted protein degradation. The successful application of this technology to ZBP1 highlights its potential for the selective elimination of disease-associated proteins and the development of novel therapeutic strategies.
Collapse
Affiliation(s)
- Rentang Huang
- State Key Laboratory of Medicinal Chemical Biology, Frontiers Science Centre for New Organic Matter, Tianjin Key Laboratory of Biosensing and Molecular Recognition, Research Centre for Analytical Sciences, College of Chemistry, School of Medicine, and Frontiers Science Centre for Cell Responses, Nankai University, Tianjin, 300071, P.R. China
| | - Yusi Hu
- State Key Laboratory of Medicinal Chemical Biology, Frontiers Science Centre for New Organic Matter, Tianjin Key Laboratory of Biosensing and Molecular Recognition, Research Centre for Analytical Sciences, College of Chemistry, School of Medicine, and Frontiers Science Centre for Cell Responses, Nankai University, Tianjin, 300071, P.R. China
| | - Yi-Fan Wang
- State Key Laboratory of Medicinal Chemical Biology, Frontiers Science Centre for New Organic Matter, Tianjin Key Laboratory of Biosensing and Molecular Recognition, Research Centre for Analytical Sciences, College of Chemistry, School of Medicine, and Frontiers Science Centre for Cell Responses, Nankai University, Tianjin, 300071, P.R. China
| | - Shiyu Zhang
- State Key Laboratory of Medicinal Chemical Biology, Frontiers Science Centre for New Organic Matter, Tianjin Key Laboratory of Biosensing and Molecular Recognition, Research Centre for Analytical Sciences, College of Chemistry, School of Medicine, and Frontiers Science Centre for Cell Responses, Nankai University, Tianjin, 300071, P.R. China
| | - Zhi-Gang Wang
- State Key Laboratory of Medicinal Chemical Biology, Frontiers Science Centre for New Organic Matter, Tianjin Key Laboratory of Biosensing and Molecular Recognition, Research Centre for Analytical Sciences, College of Chemistry, School of Medicine, and Frontiers Science Centre for Cell Responses, Nankai University, Tianjin, 300071, P.R. China
| | - Dai-Wen Pang
- State Key Laboratory of Medicinal Chemical Biology, Frontiers Science Centre for New Organic Matter, Tianjin Key Laboratory of Biosensing and Molecular Recognition, Research Centre for Analytical Sciences, College of Chemistry, School of Medicine, and Frontiers Science Centre for Cell Responses, Nankai University, Tianjin, 300071, P.R. China
| | - Shu-Lin Liu
- State Key Laboratory of Medicinal Chemical Biology, Frontiers Science Centre for New Organic Matter, Tianjin Key Laboratory of Biosensing and Molecular Recognition, Research Centre for Analytical Sciences, College of Chemistry, School of Medicine, and Frontiers Science Centre for Cell Responses, Nankai University, Tianjin, 300071, P.R. China
| |
Collapse
|
2
|
Ma W, Wang Q, Guo L, Ju X. The molecular mechanisms, roles, and potential applications of PANoptosis in cancer treatment. Front Immunol 2025; 16:1550800. [PMID: 40364845 PMCID: PMC12069359 DOI: 10.3389/fimmu.2025.1550800] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2024] [Accepted: 04/07/2025] [Indexed: 05/15/2025] Open
Abstract
PANoptosis, a newly identified form of programmed cell death regulated by the panoptosome complex, exhibits key characteristics of apoptosis, pyroptosis and necroptosis. It exerts a substantial influence on the initiation and progression of a spectrum of diseases, particularly in cancer, where its impact is increasingly being recognized. PANoptosis is closely related to tumorigenesis, carcinogenesis, metastasis, chemotherapy resistance, as well as the prediction of therapeutic responses and prognosis in cancer patients. In this review, we first review the discovery of PANoptosis and systematically analyze the composition of the panoptosome. Subsequently, we examine the role of PANoptosis in various types of cancer, encompassing its function within the tumor microenvironment, its role in tumor drug resistance, and its predictive role in cancer prognosis. Ultimately, we delve into strategies for targeting PANoptosis in cancer therapy, including targeting various molecules in the PANoptosis pathway, such as ZBP1, RIPK1, RIPK3, Caspases and other novel strategies like nanoinducers and viral vectors. This review aims to provide references and assistance for the research and application of PANoptosis in cancer treatment.
Collapse
Affiliation(s)
- Wenyuan Ma
- Department of Pathology, The People’s Hospital of Danyang, Affiliated Danyang Hospital of Nantong University, Danyang Clinical Medical College of Jiangsu University, Danyang, Jiangsu, China
| | - Qiang Wang
- School of Life Sciences, Jiangsu University, Zhenjiang, Jiangsu, China
| | - Lanfang Guo
- Department of Clinical Laboratory Medicine, The Fourth People’s Hospital of Jiangsu University, Zhenjiang, Zhenjiang, Jiangsu, China
| | - Xiaoli Ju
- Department of Pathology, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu, China
| |
Collapse
|
3
|
Hou Y, Feng Q, Wei C, Cao F, Liu D, Pan S, Shi Y, Liu Z, Liu F. Emerging role of PANoptosis in kidney diseases: molecular mechanisms and therapeutic opportunities. Apoptosis 2025; 30:579-596. [PMID: 39833634 DOI: 10.1007/s10495-024-02072-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/26/2024] [Indexed: 01/22/2025]
Abstract
Kidney diseases represent a significant global public health challenge, characterized by complex pathogenesis, high incidence, low awareness, insufficient early screening, and substantial treatment disparities. Effective therapeutic options remain lacking. Programmed cell death (PCD), including apoptosis, pyroptosis, and necroptosis, play pivotal roles in the pathogenesis of various kidney diseases. In 2019, PANoptosis, a novel form of inflammatory cell death, was introduced, providing new insights into innate immunity and PCD research. Although research on PANoptosis in kidney diseases is still limited, identifying key molecules within PANoptosomes and understanding their regulatory roles is critical for disease prevention and management. This review summarizes the various forms of PCD implicated in kidney diseases, along with PANoptosomes activated by Z-DNA binding protein 1 (ZBP1), absent in melanoma 2 (AIM2), receptor-interacting protein kinase 1 (RIPK1), NOD-like receptor family CARD domain containing 12 (NLRP12), and NOD-like receptor family member C5 (NLRC5). It also reviews the advancements in PANoptosis research in the field of kidney diseases, particularly in renal tumors and acute kidney injuries (AKI). The goal is to establish a foundation for future research into the role of PANoptosis in kidney diseases.
Collapse
Affiliation(s)
- Yi Hou
- Department of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, P. R. China
- Henan Province Research Center for Kidney Disease, Zhengzhou, 450052, P. R. China
| | - Qi Feng
- Department of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, P. R. China
- Research Institute of Nephrology, Zhengzhou University, Zhengzhou, 450052, P. R. China
- Henan Province Research Center for Kidney Disease, Zhengzhou, 450052, P. R. China
- Key Laboratory of Precision Diagnosis and Treatment for Chronic Kidney Disease in Henan Province, Zhengzhou, 450052, P. R. China
| | - Cien Wei
- Department of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, P. R. China
- Henan Province Research Center for Kidney Disease, Zhengzhou, 450052, P. R. China
| | - Fengyu Cao
- Department of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, P. R. China
- Henan Province Research Center for Kidney Disease, Zhengzhou, 450052, P. R. China
| | - Dongwei Liu
- Department of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, P. R. China
- Traditional Chinese Medicine Integrated Department of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, P. R. China
- Research Institute of Nephrology, Zhengzhou University, Zhengzhou, 450052, P. R. China
- Henan Province Research Center for Kidney Disease, Zhengzhou, 450052, P. R. China
- Key Laboratory of Precision Diagnosis and Treatment for Chronic Kidney Disease in Henan Province, Zhengzhou, 450052, P. R. China
| | - Shaokang Pan
- Department of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, P. R. China
- Traditional Chinese Medicine Integrated Department of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, P. R. China
- Research Institute of Nephrology, Zhengzhou University, Zhengzhou, 450052, P. R. China
- Henan Province Research Center for Kidney Disease, Zhengzhou, 450052, P. R. China
- Key Laboratory of Precision Diagnosis and Treatment for Chronic Kidney Disease in Henan Province, Zhengzhou, 450052, P. R. China
| | - Yan Shi
- Department of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, P. R. China
- Henan Province Research Center for Kidney Disease, Zhengzhou, 450052, P. R. China
| | - Zhangsuo Liu
- Department of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, P. R. China.
- Traditional Chinese Medicine Integrated Department of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, P. R. China.
- Research Institute of Nephrology, Zhengzhou University, Zhengzhou, 450052, P. R. China.
- Henan Province Research Center for Kidney Disease, Zhengzhou, 450052, P. R. China.
- Key Laboratory of Precision Diagnosis and Treatment for Chronic Kidney Disease in Henan Province, Zhengzhou, 450052, P. R. China.
| | - Fengxun Liu
- Department of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, P. R. China.
- Traditional Chinese Medicine Integrated Department of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, P. R. China.
- Research Institute of Nephrology, Zhengzhou University, Zhengzhou, 450052, P. R. China.
- Henan Province Research Center for Kidney Disease, Zhengzhou, 450052, P. R. China.
- Key Laboratory of Precision Diagnosis and Treatment for Chronic Kidney Disease in Henan Province, Zhengzhou, 450052, P. R. China.
| |
Collapse
|
4
|
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
|
5
|
Zhou H, Zhou M, Liao X, Zhang L, Wei H, Lu Y, Zhang Y, Huang H, Hu Y, Chen T, Lv Z. The Innate Immune Sensor Zbp1 Mediates Central Nervous System Inflammation Induced by Angiostrongylus Cantonensis by Promoting Macrophage Inflammatory Phenotypes. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2413675. [PMID: 39853924 PMCID: PMC11923990 DOI: 10.1002/advs.202413675] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/25/2024] [Revised: 01/14/2025] [Indexed: 01/26/2025]
Abstract
Angiostrongylus cantonensis (AC) is the leading cause of eosinophilic meningoencephalitis worldwide. The neuroimmune interactions between peripheral and central immune systems in angiostrongyliasis remain unclear. In this study, significant infiltration of eosinophils, myeloid cells, macrophages, neutrophils, and Ly6C monocytes is observed in the brains of AC-infected mice, with macrophages being the most abundant. RNA-seq and SMART-seq analysis of pattern recognition receptor (PRR) and DNA sensor gene sets revealed a marked increase in Z-DNA binding protein 1 (Zbp1) expression in infected mice. Confocal microscopy, RT-qPCR, western blotting, and immunohistochemistry further confirmed that Zbp1 is specifically upregulated in macrophages and microglia. Using Zbp1-knockout mice and flow cytometry, it is found that knockout of Zbp1 enhanced lymphocyte infiltration and natural killer cell cytotoxicity, modulating the immune microenvironment in the central nervous system (CNS) during AC infection. Mechanistically, it is revealed that in macrophage Zbp1 directly binds to receptor-interacting protein 3 (RIP3) to promote its phosphorylation, subsequently facilitating the phosphorylation of mixed lineage kinase domain-like protein (Mlkl). The activated Zbp1-pRIP3-pMlkl axis leads to necroptosis and upregulates pro-inflammatory cytokines including TNF-α, IL-1α, CXCL9, CXCL10 in macrophages, which recruits and activates immune cells. These findings offer new insights into the pathogenic mechanisms of angiostrongyliasis and suggest potential therapeutic strategies.
Collapse
Affiliation(s)
- Hongli Zhou
- Key Laboratory of Tropical Disease Control (Sun Yat‐Sen University)Ministry of EducationGuangzhouGuangdong510080China
- Clinical Medical Research CenterThe Second Affiliated HospitalArmy Medical UniversityChongqing400037China
| | - Minyu Zhou
- Key Laboratory of Tropical Disease Control (Sun Yat‐Sen University)Ministry of EducationGuangzhouGuangdong510080China
- Provincial Engineering Technology Research Center for Biological Vector ControlSun Yat‐sen UniversityGuangzhou510080China
| | - XiPing Liao
- Clinical Medical Research CenterThe Second Affiliated HospitalArmy Medical UniversityChongqing400037China
| | - Liangyu Zhang
- Key Laboratory of Tropical Disease Control (Sun Yat‐Sen University)Ministry of EducationGuangzhouGuangdong510080China
- Provincial Engineering Technology Research Center for Biological Vector ControlSun Yat‐sen UniversityGuangzhou510080China
| | - Hang Wei
- Key Laboratory of Tropical Disease Control (Sun Yat‐Sen University)Ministry of EducationGuangzhouGuangdong510080China
- Provincial Engineering Technology Research Center for Biological Vector ControlSun Yat‐sen UniversityGuangzhou510080China
| | - Yuting Lu
- Key Laboratory of Tropical Disease Control (Sun Yat‐Sen University)Ministry of EducationGuangzhouGuangdong510080China
- Provincial Engineering Technology Research Center for Biological Vector ControlSun Yat‐sen UniversityGuangzhou510080China
| | - Yiqing Zhang
- Key Laboratory of Tropical Disease Control (Sun Yat‐Sen University)Ministry of EducationGuangzhouGuangdong510080China
- Provincial Engineering Technology Research Center for Biological Vector ControlSun Yat‐sen UniversityGuangzhou510080China
| | - Hui Huang
- Key Laboratory of Tropical Disease Control (Sun Yat‐Sen University)Ministry of EducationGuangzhouGuangdong510080China
- Provincial Engineering Technology Research Center for Biological Vector ControlSun Yat‐sen UniversityGuangzhou510080China
| | - Yue Hu
- Key Laboratory of Tropical Disease Control (Sun Yat‐Sen University)Ministry of EducationGuangzhouGuangdong510080China
- Provincial Engineering Technology Research Center for Biological Vector ControlSun Yat‐sen UniversityGuangzhou510080China
| | - Tao Chen
- Department of NeurologyHainan General Hospital,Hainan Affiliated Hospital of Hainan Medical UniversityHaikouHainan570311China
| | - Zhiyue Lv
- Key Laboratory of Tropical Disease Control (Sun Yat‐Sen University)Ministry of EducationGuangzhouGuangdong510080China
- Provincial Engineering Technology Research Center for Biological Vector ControlSun Yat‐sen UniversityGuangzhou510080China
- Department of NeurologyHainan General Hospital,Hainan Affiliated Hospital of Hainan Medical UniversityHaikouHainan570311China
- Key Laboratory of Tropical Translational Medicine of Ministry of EducationHainan Medical UniversityHaikouHainan570216China
| |
Collapse
|
6
|
Yang W, Xu Y, Liu S, Gao L, Li S, Xie X, Zhang Q, Habib O, Chen R, Sun X, Li Z. Mebendazole induces ZBP-1 mediated PANoptosis of acute myeloid leukemia cells by targeting TUBA1A and exerts antileukemia effect. J Adv Res 2025:S2090-1232(25)00111-0. [PMID: 39952321 DOI: 10.1016/j.jare.2025.02.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Revised: 12/23/2024] [Accepted: 02/10/2025] [Indexed: 02/17/2025] Open
Abstract
BACKGROUND Despite notable advancements in AML therapy in recent years, a substantial proportion of patients remain refractory or at high risk of recurrence with limited efficacy. Therefore, it's urgent to develop novel drugs for treating AML. METHODS The small molecule drug library was utilized to screen for drugs that elicit the inflammatory death of AML cells. Cell viability, cell morphological analysis, western blotting, and RNA-seq were used to determine the pathway of Mebendazole (MBD)-induced AML cell death. Cell cycle analysis, protein expression profiling, molecular docking, western blotting and lentivirus overexpression were used to analyze the target protein of MBD in AML cells. The anti-AML activity of MBD in vivo was evaluated using tumor xenograft models constructed by AML cell lines and patient-derived primary AML cells. RESULTS In this study, we have identified Mebendazole (MBD), a conventional anthelmintic drug known for its low toxicity and cost, as a potent agent that exerts significant anti-AML effects in vitro. Furthermore, we have observed its inhibitory effects on the invasion of AML cell lines and primary AML cells in xenograft mouse models, while noting its negligible toxic side effects in normal mice in vivo. Mechanically, MBD inhibits the cell cycle in G2/M phase by inhibiting tubulin α1A (TUBA1A) and promotes ZBP-1 mediated PANoptosis in AML cells. Our results confirm that MBD exerts anti-AML activity in preclinical models. CONCLUSION These results highlight the remarkable clinical translational potential of MBD, providing new potential medicine for AML patients. In addition, TUBA1A can be used potential novel therapeutic target in tumors with abnormal TUBA1A expression.
Collapse
Affiliation(s)
- Wei Yang
- Guangdong Provincial Key Laboratory of Systems Biology and Synthetic Biology for Urogenital Tumors, Shenzhen Key Laboratory of Genitourinary Tumor, Department of Urology, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital (Shenzhen Institute of Translational Medicine), Shenzhen, China; Department of Critical Care Medicine, Longgang Central Hospital, Shenzhen 6082 longgang Road, Shenzhen 518116 Guangdong, China
| | - Ying Xu
- Department of hematopathology, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University, The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen 518020 Guangdong, PR China
| | - Shuai Liu
- Department of Laboratory, Shenzhen Samii International Medical Center (Shenzhen Fourth People's Hospital), Shenzhen 518118, P.R. China
| | - Lin Gao
- Guangdong Provincial Key Laboratory of Systems Biology and Synthetic Biology for Urogenital Tumors, Shenzhen Key Laboratory of Genitourinary Tumor, Department of Urology, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital (Shenzhen Institute of Translational Medicine), Shenzhen, China
| | - Shi Li
- Guangdong Provincial Key Laboratory of Systems Biology and Synthetic Biology for Urogenital Tumors, Shenzhen Key Laboratory of Genitourinary Tumor, Department of Urology, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital (Shenzhen Institute of Translational Medicine), Shenzhen, China
| | - Xina Xie
- Guangdong Provincial Key Laboratory of Systems Biology and Synthetic Biology for Urogenital Tumors, Shenzhen Key Laboratory of Genitourinary Tumor, Department of Urology, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital (Shenzhen Institute of Translational Medicine), Shenzhen, China
| | - Qiaoxia Zhang
- Shenzhen Bone Marrow Transplantation Public Service Platform, Shenzhen Institute of Hematology, Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University, Shenzhen University Health Sciences Center, Shenzhen 518035, PR China
| | - Obaid Habib
- Guangdong Provincial Key Laboratory of Systems Biology and Synthetic Biology for Urogenital Tumors, Shenzhen Key Laboratory of Genitourinary Tumor, Department of Urology, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital (Shenzhen Institute of Translational Medicine), Shenzhen, China
| | - Ronglin Chen
- Department of Critical Care Medicine, Longgang Central Hospital, Shenzhen 6082 longgang Road, Shenzhen 518116 Guangdong, China.
| | - Xiongfei Sun
- Department of hematopathology, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University, The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen 518020 Guangdong, PR China; Shenzhen Clinical Research Centre for Geriatrics, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University, The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen 518020 Guangdong, PR China.
| | - Zesong Li
- Guangdong Provincial Key Laboratory of Systems Biology and Synthetic Biology for Urogenital Tumors, Shenzhen Key Laboratory of Genitourinary Tumor, Department of Urology, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital (Shenzhen Institute of Translational Medicine), Shenzhen, China; Institute of Basic Medicine and Forensic Medicine, North Sichuan Medical College, Nanchong, Sichuan 637199, China.
| |
Collapse
|
7
|
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
|
8
|
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
|
9
|
Luo J, Lu Y, Dai E, Yin N, Wang T, Qian H, Jiang Q, Cao X, Wang C, Zeng Y. The PB1 protein of H9N2 influenza A virus inhibits antiviral innate immunity by targeting MAVS for TRIM25-mediated autophagic degradation. Poult Sci 2025; 104:104639. [PMID: 39647358 PMCID: PMC11666973 DOI: 10.1016/j.psj.2024.104639] [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: 08/27/2024] [Revised: 11/21/2024] [Accepted: 12/03/2024] [Indexed: 12/10/2024] Open
Abstract
The proteins encoded by Influenza A virus (IAV) evade the innate immune system through diverse strategies to facilitate their replication. However, the regulatory mechanisms remain not fully understood. In this study, we identified that the H9N2 PB1 protein suppressed the activities of the IFN-β, ISRE, and NF-κB promoters. Furthermore, H9N2 PB1 inhibited the phosphorylation of IRF3, IκBα, and TBK1 and the secretion of IFN-β. The results demonstrated H9N2 PB1 as a negative regulator of the RIG-I signaling pathway. Subsequent investigations revealed a specific interaction between H9N2 PB1 and MAVS, which disturbed the stability of MAVS. Notably, we discovered that H9N2 PB1 exploited the function of TRIM25, leading to the autophagic degradation of MAVS through K48-linked polyubiquitination. In conclusion, we uncovered a negative regulatory axis consisting of H9N2 PB1-TRIM25-MAVS-IFN-I. These findings provide valuable insights into the molecular interactions involved in the regulation of the host's innate immune antiviral response by IAV.
Collapse
Affiliation(s)
- Jiawei Luo
- College of Veterinary Medicine, Jilin Agricultural University, Changchun, 130118, PR China
| | - Yiyuan Lu
- College of Veterinary Medicine, Jilin Agricultural University, Changchun, 130118, PR China; Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Agricultural University, Changchun, 130118, PR China; Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Jilin Agricultural University, Changchun, 130118, PR China; Engineering Research Center of Microecological Vaccines (Drugs) for Major Animal Diseases, Ministry of Education, Jilin Agricultural University, Changchun, 130118, PR China
| | - Enqi Dai
- College of Veterinary Medicine, Jilin Agricultural University, Changchun, 130118, PR China
| | - Nianchun Yin
- Suining Municipal Agricultural and Rural Bureau of Sichuan Province, Suining, 629000, PR China
| | - Ting Wang
- College of Veterinary Medicine, Jilin Agricultural University, Changchun, 130118, PR China
| | - Hongxi Qian
- TECHLEX Food Co. Ltd, Mianyang, 621000, PR China
| | - Qingrong Jiang
- Sichuan Sundaily Farm Ecological Food Co. Ltd, Mianyang, 621000, PR China
| | - Xin Cao
- College of Veterinary Medicine, Jilin Agricultural University, Changchun, 130118, PR China; Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Agricultural University, Changchun, 130118, PR China; Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Jilin Agricultural University, Changchun, 130118, PR China; Engineering Research Center of Microecological Vaccines (Drugs) for Major Animal Diseases, Ministry of Education, Jilin Agricultural University, Changchun, 130118, PR China.
| | - Chunfeng Wang
- College of Veterinary Medicine, Jilin Agricultural University, Changchun, 130118, PR China; Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Agricultural University, Changchun, 130118, PR China; Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Jilin Agricultural University, Changchun, 130118, PR China; Engineering Research Center of Microecological Vaccines (Drugs) for Major Animal Diseases, Ministry of Education, Jilin Agricultural University, Changchun, 130118, PR China.
| | - Yan Zeng
- College of Veterinary Medicine, Jilin Agricultural University, Changchun, 130118, PR China; Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Agricultural University, Changchun, 130118, PR China; Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Jilin Agricultural University, Changchun, 130118, PR China; Engineering Research Center of Microecological Vaccines (Drugs) for Major Animal Diseases, Ministry of Education, Jilin Agricultural University, Changchun, 130118, PR China.
| |
Collapse
|
10
|
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
|
11
|
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
|
12
|
Hoover AR, Liu K, Furrer C, Lam SSK, Anderson DW, Zhou Z, Yang J, Wong CF, Medcalf AD, Sun XH, Hode T, Alleruzzo L, Delawder A, Raker J, Abousleiman G, Valerio TI, Sun Y, Papin JF, Li M, Chen WR. N-Dihydrogalactochitosan Drives Conventional and Alternative Activations of STING to Synergize Type I IFN and IL-1β Productions for Antitumor Immunity. ADVANCED FUNCTIONAL MATERIALS 2024; 34:2410079. [PMID: 39896882 PMCID: PMC11784597 DOI: 10.1002/adfm.202410079] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/10/2024] [Indexed: 02/04/2025]
Abstract
N-dihydrogalactochitosan (GC) is an immune stimulant/adjuvant. Synthesized from chitosan and galactose, GC is a new chemical entity that significantly enhances the immune-stimulating properties of its parental material, chitosan, making it a promising therapeutic agent. When used in combination with antigenic material, GC stimulates innate and adaptive antitumor and antiviral immunities. However, its mechanism has not been fully investigated. Herein we demonstrate that GC drives type I IFN activation in antigen-presenting cells (APCs). More importantly, GC drives alternative STING pathways, leading to inflammatory cell death that enhances dendritic cell (DC) activation. GC-activated DCs trigger a variety of nucleic acid sensing pattern recognition receptors (PRRs) pathways and IL-1β production via the activation of the inflammasome. In vivo, GC induces a potent response of type I IFNs and upregulates genes associated with STING signaling within the tumor microenvironment (TME). Moreover, intratumoral delivery of GC reduces the numbers of M2-like macrophages and increases M1-like macrophages residing within the TME, while subsequently increasing the number of activated DCs. Our findings demonstrate that GC acts as a multimodal immune stimulant via STING to generate a broad type I IFN response. This uniquely broad response holds therapeutic promise in generating enhanced antitumor and antiviral immunities.
Collapse
Affiliation(s)
- Ashley R. Hoover
- Stephenson School of Biomedical Engineering, University of Oklahoma, Norman, OK, USA
- Arthritis and Clinical Immunology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
| | - Kaili Liu
- Stephenson School of Biomedical Engineering, University of Oklahoma, Norman, OK, USA
| | - Coline Furrer
- Stephenson School of Biomedical Engineering, University of Oklahoma, Norman, OK, USA
| | | | | | - Zhijun Zhou
- Department of Medicine, Department of Surgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - Jingxuan Yang
- Department of Medicine, Department of Surgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | | | - Alexandra D. Medcalf
- Arthritis and Clinical Immunology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
| | - Xiao-Hong Sun
- Arthritis and Clinical Immunology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
| | - Tomas Hode
- Immunophotonics, Inc., Saint Louis, MO, USA
| | | | | | | | - Ghainaa Abousleiman
- Stephenson School of Biomedical Engineering, University of Oklahoma, Norman, OK, USA
| | - Trisha I. Valerio
- Stephenson School of Biomedical Engineering, University of Oklahoma, Norman, OK, USA
| | - Yuanhong Sun
- Stephenson School of Biomedical Engineering, University of Oklahoma, Norman, OK, USA
| | - James F. Papin
- Department Pathology and Division of Comparative Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, USA
| | - Min Li
- Department of Medicine, Department of Surgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
- Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - Wei R. Chen
- Stephenson School of Biomedical Engineering, University of Oklahoma, Norman, OK, USA
- Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| |
Collapse
|
13
|
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
|
14
|
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
|
15
|
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
|
16
|
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
|
17
|
Yang F, Zhang G, An N, Dai Q, Cho W, Shang H, Xing Y. Interplay of ferroptosis, cuproptosis, and PANoptosis in cancer treatment-induced cardiotoxicity: Mechanisms and therapeutic implications. Semin Cancer Biol 2024; 106-107:106-122. [PMID: 39299410 DOI: 10.1016/j.semcancer.2024.09.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Revised: 09/14/2024] [Accepted: 09/16/2024] [Indexed: 09/22/2024]
Abstract
With the prolonged survival of individuals with cancer, the emergence of cardiovascular diseases (CVD) induced by cancer treatment has become a significant concern, ranking as the second leading cause of death among cancer survivors. This review explores three distinct types of programmed cell death (PCD): ferroptosis, cuproptosis, and PANoptosis, focusing on their roles in chemotherapy-induced cardiotoxicity. While ferroptosis and cuproptosis are triggered by excess iron and copper (Cu), PANoptosis is an inflammatory PCD with features of pyroptosis, apoptosis, and necroptosis. Recent studies reveal intricate connections among these PCD types, emphasizing the interplay between cuproptosis and ferroptosis. Notably, the role of intracellular Cu in promoting ferroptosis through GPX4 is highlighted. Additionally, ROS-induced PANoptosis is influenced by ferroptosis and cuproptosis, suggesting a complex interrelationship. This review provides insights into the molecular mechanisms of these PCD modalities and their distinct contributions to chemotherapy-induced cardiotoxicity. Furthermore, we discuss the potential application of cardioprotective drugs in managing these PCD types. This comprehensive analysis aims to advance the understanding, diagnosis, and therapeutic strategies for cardiotoxicity associated with cancer treatment.
Collapse
Affiliation(s)
- Fan Yang
- Guang'anmen Hospital, Chinese Academy of Chinese Medical Sciences, Beijing 100053, China
| | - Guoxia Zhang
- Guang'anmen Hospital, Chinese Academy of Chinese Medical Sciences, Beijing 100053, China; Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Beijing 100700, China
| | - Na An
- Key Laboratory of Chinese Internal Medicine of the Ministry of Education, Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Beijing 100700, China
| | - Qianqian Dai
- Key Laboratory of Chinese Internal Medicine of the Ministry of Education, Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Beijing 100700, China
| | - William Cho
- Department of Clinical Oncology, Queen Elizabeth Hospital, Kowloon, Hong Kong, China.
| | - Hongcai Shang
- Key Laboratory of Chinese Internal Medicine of the Ministry of Education, Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Beijing 100700, China.
| | - Yanwei Xing
- Guang'anmen Hospital, Chinese Academy of Chinese Medical Sciences, Beijing 100053, China.
| |
Collapse
|
18
|
Gong T, Wang QD, Loughran PA, Li YH, Scott MJ, Billiar TR, Liu YT, Fan J. Mechanism of lactic acidemia-promoted pulmonary endothelial cells death in sepsis: role for CIRP-ZBP1-PANoptosis pathway. Mil Med Res 2024; 11:71. [PMID: 39465383 PMCID: PMC11514876 DOI: 10.1186/s40779-024-00574-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/19/2024] [Accepted: 09/30/2024] [Indexed: 10/29/2024] Open
Abstract
BACKGROUND Sepsis is often accompanied by lactic acidemia and acute lung injury (ALI). Clinical studies have established that high serum lactate levels are associated with increased mortality rates in septic patients. We further observed a significant correlation between the levels of cold-inducible RNA-binding protein (CIRP) in plasma and bronchoalveolar lavage fluid (BALF), as well as lactate levels, and the severity of post-sepsis ALI. The underlying mechanism, however, remains elusive. METHODS C57BL/6 wild type (WT), Casp8-/-, Ripk3-/-, and Zbp1-/- mice were subjected to the cecal ligation and puncture (CLP) sepsis model. In this model, we measured intra-macrophage CIRP lactylation and the subsequent release of CIRP. We also tracked the internalization of extracellular CIRP (eCIRP) in pulmonary vascular endothelial cells (PVECs) and its interaction with Z-DNA binding protein 1 (ZBP1). Furthermore, we monitored changes in ZBP1 levels in PVECs and the consequent activation of cell death pathways. RESULTS In the current study, we demonstrate that lactate, accumulating during sepsis, promotes the lactylation of CIRP in macrophages, leading to the release of CIRP. Once eCIRP is internalized by PVEC through a Toll-like receptor 4 (TLR4)-mediated endocytosis pathway, it competitively binds to ZBP1 and effectively blocks the interaction between ZBP1 and tripartite motif containing 32 (TRIM32), an E3 ubiquitin ligase targeting ZBP1 for proteasomal degradation. This interference mechanism stabilizes ZBP1, thereby enhancing ZBP1-receptor-interacting protein kinase 3 (RIPK3)-dependent PVEC PANoptosis, a form of cell death involving the simultaneous activation of multiple cell death pathways, thereby exacerbating ALI. CONCLUSIONS These findings unveil a novel pathway by which lactic acidemia promotes macrophage-derived eCIRP release, which, in turn, mediates ZBP1-dependent PVEC PANoptosis in sepsis-induced ALI. This finding offers new insights into the molecular mechanisms driving sepsis-related pulmonary complications and provides potential new therapeutic strategies.
Collapse
Affiliation(s)
- Ting Gong
- Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15213, USA.
- Department of Anesthesiology, Shenzhen Hospital of Southern Medical University, Shenzhen, 518110, Guangdong, China.
| | - Qing-De Wang
- Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15213, USA
| | - Patricia A Loughran
- Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15213, USA
| | - Yue-Hua Li
- Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15213, USA
| | - Melanie J Scott
- Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15213, USA
| | - Timothy R Billiar
- Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15213, USA
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA, 15219, USA
| | - You-Tan Liu
- Department of Anesthesiology, Shenzhen Hospital of Southern Medical University, Shenzhen, 518110, Guangdong, China.
| | - Jie Fan
- Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15213, USA.
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA, 15219, USA.
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15213, USA.
- Research and Development, Veterans Affairs Pittsburgh Healthcare System, Pittsburgh, PA, 15240, USA.
| |
Collapse
|
19
|
Xiang Q, Geng ZX, Yi X, Wei X, Zhu XH, Jiang DS. PANoptosis: a novel target for cardiovascular diseases. Trends Pharmacol Sci 2024; 45:739-756. [PMID: 39003157 DOI: 10.1016/j.tips.2024.06.002] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Revised: 06/18/2024] [Accepted: 06/19/2024] [Indexed: 07/15/2024]
Abstract
PANoptosis is a unique innate immune inflammatory lytic cell death pathway initiated by an innate immune sensor and driven by caspases and RIPKs. As a distinct pathway, the execution of PANoptosis cannot be hindered by targeting other cell death pathways, such as pyroptosis, apoptosis, or necroptosis. Instead, targeting key PANoptosome components can serve as a strategy to prevent this form of cell death. Given the physiological relevance in several diseases, PANoptosis is a pivotal therapeutic target. Notably, previous research has primarily focused on the role of PANoptosis in cancer and infectious and inflammatory diseases. By contrast, its role in cardiovascular diseases has not been comprehensively discussed. Here, we review the available evidence on PANoptosis in cardiovascular diseases, including cardiomyopathy, atherosclerosis, myocardial infarction, myocarditis, and aortic aneurysm and dissection, and explore a variety of agents that target PANoptosis, with the overarching goal of providing a novel complementary approach to combatting cardiovascular diseases.
Collapse
Affiliation(s)
- Qi Xiang
- Division of Cardiovascular Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Zhen-Xi Geng
- Division of Cardiovascular Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Xin Yi
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Xiang Wei
- Division of Cardiovascular Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China; Key Laboratory of Organ Transplantation, Ministry of Education; NHC Key Laboratory of Organ Transplantation; Key Laboratory of Organ Transplantation, Chinese Academy of Medical Sciences, Wuhan, Hubei, China
| | - Xue-Hai Zhu
- Division of Cardiovascular Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China; Key Laboratory of Organ Transplantation, Ministry of Education; NHC Key Laboratory of Organ Transplantation; Key Laboratory of Organ Transplantation, Chinese Academy of Medical Sciences, Wuhan, Hubei, China.
| | - Ding-Sheng Jiang
- Division of Cardiovascular Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China; Key Laboratory of Organ Transplantation, Ministry of Education; NHC Key Laboratory of Organ Transplantation; Key Laboratory of Organ Transplantation, Chinese Academy of Medical Sciences, Wuhan, Hubei, China.
| |
Collapse
|
20
|
Liang Y, Yan Y, Liu N, Wang J, Fang C. Shengxian decoction improves lung function in rats with bleomycin-induced idiopathic pulmonary fibrosis through the inhibition of PANoptosis. JOURNAL OF ETHNOPHARMACOLOGY 2024; 329:118153. [PMID: 38604513 DOI: 10.1016/j.jep.2024.118153] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Revised: 04/02/2024] [Accepted: 04/03/2024] [Indexed: 04/13/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Shengxian decoction (SXD) is a classic Chinese medicinal formula that can effectively improve clinical symptoms and quality of life and delay disease progression in idiopathic pulmonary fibrosis (IPF) patients; however, the underlying mechanisms remain unclear. AIM OF THE STUDY This study aimed to observe PANoptosis in bleomycin-induced IPF and to assess the efficacy and mechanism of action of SXD in the treatment of IPF. MATERIALS AND METHODS Fifty SD rats were randomly divided into the sham, IPF, IPF + pirfenidone (PFD), IPF + SXD-medium dose (SXD-M), and IPF + SXD-low dose (SXD-L) groups. Lung function analysis and microcomputed tomography imaging of the rats with IPF treated with oral pirfenidone or oral SXD for 28 days were performed. Hematoxylin and eosin (HE) staining and Masson's trichrome staining were used to observe pathological lung damage. Enzyme-linked immunosorbent assays (ELISAs) were used to determine the serum levels of IL-1β, IL-18, TNF-α, and IFN-γ. Pyroptosis, apoptosis, and necroptosis were assessed using TUNEL, TUNEL/caspase-1, and PI fluorescence staining, respectively. GSDMD, caspase-3, and MLKL were examined by immunohistochemistry. The expression of fibrin-, ZBP1-, pyroptosis-, apoptosis-, and necroptosis-related proteins in the lung tissue was determined by western blotting. RESULTS SXD normalized lung function in rats with bleomycin-induced IPF and reduced serum inflammatory factor levels and lung tissue fibrosis. The underlying mechanism of action involves the inhibition of pyroptosis pathway proteins, such as NLRP3, caspase-1, cleaved caspase-1, and GSDMD; apoptotic pathway proteins, such as Bax, Bcl-2, cleaved caspase-3, and caspase-3; and necroptosis pathway proteins, such as RIPK1, RIPK3, p-MLKL and MLKL. These pathways are modulated by the PANoptosis initiator ZBP1. Notably, the efficacy of SXD is concentration dependent, with a medium dose exhibiting superior effectiveness compared to a low dose. CONCLUSION Bleomycin induced PANoptosis in the lung tissue of rats with IPF. Additionally, SXD effectively delayed or reversed the early pathological changes in bleomycin-induced pulmonary fibrosis by inhibiting PANoptosis.
Collapse
Affiliation(s)
- Yulei Liang
- Graduate School, Hebei University of Chinese Medicine, Shijiazhuang 050200, China
| | - Yanan Yan
- Graduate School, Hebei University of Chinese Medicine, Shijiazhuang 050200, China
| | - Na Liu
- Graduate School, Hebei University of Chinese Medicine, Shijiazhuang 050200, China
| | - Jiepeng Wang
- Graduate School, Hebei University of Chinese Medicine, Shijiazhuang 050200, China; Hebei Key Laboratory of Lung Disease Research of Integrated Traditional Chinese and Western Medicine, Shijiazhuang 050091, China
| | - Chaoyi Fang
- Graduate School, Hebei University of Chinese Medicine, Shijiazhuang 050200, China; Hebei Key Laboratory of Lung Disease Research of Integrated Traditional Chinese and Western Medicine, Shijiazhuang 050091, China.
| |
Collapse
|
21
|
Liang K, Barnett KC, Hsu M, Chou WC, Bais SS, Riebe K, Xie Y, Nguyen TT, Oguin TH, Vannella KM, Hewitt SM, Chertow DS, Blasi M, Sempowski GD, Karlsson A, Koller BH, Lenschow DJ, Randell SH, Ting JPY. Initiator cell death event induced by SARS-CoV-2 in the human airway epithelium. Sci Immunol 2024; 9:eadn0178. [PMID: 38996010 PMCID: PMC11970318 DOI: 10.1126/sciimmunol.adn0178] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Accepted: 06/14/2024] [Indexed: 07/14/2024]
Abstract
Virus-induced cell death is a key contributor to COVID-19 pathology. Cell death induced by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is well studied in myeloid cells but less in its primary host cell type, angiotensin-converting enzyme 2 (ACE2)-expressing human airway epithelia (HAE). SARS-CoV-2 induces apoptosis, necroptosis, and pyroptosis in HAE organotypic cultures. Single-cell and limiting-dilution analysis revealed that necroptosis is the primary cell death event in infected cells, whereas uninfected bystanders undergo apoptosis, and pyroptosis occurs later during infection. Mechanistically, necroptosis is induced by viral Z-RNA binding to Z-DNA-binding protein 1 (ZBP1) in HAE and lung tissues from patients with COVID-19. The Delta (B.1.617.2) variant, which causes more severe disease than Omicron (B1.1.529) in humans, is associated with orders of magnitude-greater Z-RNA/ZBP1 interactions, necroptosis, and disease severity in animal models. Thus, Delta induces robust ZBP1-mediated necroptosis and more disease severity.
Collapse
Affiliation(s)
- Kaixin Liang
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Center for Translational Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Oral and Craniofacial Biomedicine Program, Adams School of Dentistry, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Katherine C. Barnett
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Center for Translational Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Martin Hsu
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Center for Translational Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Wei-chun Chou
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Center for Translational Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Sachendra S. Bais
- Department of Medicine, Washington University School of Medicine, Saint Louis, Missouri, USA
| | | | - Yuying Xie
- Department of Computational Mathematics, Science and Engineering, Michigan State University, East Lansing, MI 48824, USA
- Department of Statistics and Probability, Michigan State University, East Lansing, MI 48824, USA
| | - Tuong Thien Nguyen
- Marsico Lung Institute, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | | | - Kevin M. Vannella
- Emerging Pathogens Section, Critical Care Medicine Department, Clinical Center, National Institutes of Health, Bethesda, MD, USA
- Laboratory of Virology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, USA
| | - Stephen M. Hewitt
- Laboratory of Pathology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Daniel S. Chertow
- Emerging Pathogens Section, Critical Care Medicine Department, Clinical Center, National Institutes of Health, Bethesda, MD, USA
- Laboratory of Virology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, USA
| | - Maria Blasi
- Duke Human Vaccine Institute, Durham, NC 27701, USA
- Department of Medicine, Duke University School of Medicine, Durham, North Carolina, USA
| | - Gregory D. Sempowski
- Duke Human Vaccine Institute, Durham, NC 27701, USA
- Present address: RTI International, Research Triangle Park, Durham, NC 27709, USA
| | - Amelia Karlsson
- Duke Human Vaccine Institute, Durham, NC 27701, USA
- Present address: RTI International, Research Triangle Park, Durham, NC 27709, USA
| | - Beverly H. Koller
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Deborah J. Lenschow
- Department of Medicine, Washington University School of Medicine, Saint Louis, Missouri, USA
- Department of Pathology and Immunology, Washington University School of Medicine, Saint Louis, Missouri, USA
- Department of Molecular Microbiology, Washington University School of Medicine, Saint Louis, Missouri, USA
| | - Scott H. Randell
- Marsico Lung Institute, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Jenny P.-Y. Ting
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Center for Translational Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| |
Collapse
|
22
|
An W, Lakhina S, Leong J, Rawat K, Husain M. Host Innate Antiviral Response to Influenza A Virus Infection: From Viral Sensing to Antagonism and Escape. Pathogens 2024; 13:561. [PMID: 39057788 PMCID: PMC11280125 DOI: 10.3390/pathogens13070561] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Revised: 06/26/2024] [Accepted: 07/01/2024] [Indexed: 07/28/2024] Open
Abstract
Influenza virus possesses an RNA genome of single-stranded, negative-sensed, and segmented configuration. Influenza virus causes an acute respiratory disease, commonly known as the "flu" in humans. In some individuals, flu can lead to pneumonia and acute respiratory distress syndrome. Influenza A virus (IAV) is the most significant because it causes recurring seasonal epidemics, occasional pandemics, and zoonotic outbreaks in human populations, globally. The host innate immune response to IAV infection plays a critical role in sensing, preventing, and clearing the infection as well as in flu disease pathology. Host cells sense IAV infection through multiple receptors and mechanisms, which culminate in the induction of a concerted innate antiviral response and the creation of an antiviral state, which inhibits and clears the infection from host cells. However, IAV antagonizes and escapes many steps of the innate antiviral response by different mechanisms. Herein, we review those host and viral mechanisms. This review covers most aspects of the host innate immune response, i.e., (1) the sensing of incoming virus particles, (2) the activation of downstream innate antiviral signaling pathways, (3) the expression of interferon-stimulated genes, (4) and viral antagonism and escape.
Collapse
Affiliation(s)
| | | | | | | | - Matloob Husain
- Department of Microbiology and Immunology, University of Otago, P.O. Box 56, Dunedin 9054, New Zealand; (W.A.); (S.L.); (J.L.); (K.R.)
| |
Collapse
|
23
|
Bae H, Jang Y, Karki R, Han JH. Implications of inflammatory cell death-PANoptosis in health and disease. Arch Pharm Res 2024; 47:617-631. [PMID: 38987410 DOI: 10.1007/s12272-024-01506-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Accepted: 07/06/2024] [Indexed: 07/12/2024]
Abstract
Regulated cell death (RCD) pathways, such as pyroptosis, apoptosis, and necroptosis, are essential for maintaining the body's balance, defending against pathogens, and eliminating abnormal cells that could lead to diseases like cancer. Although these pathways operate through distinct mechanisms, recent genetic and pharmacological studies have shown that they can interact and influence each other. The concept of "PANoptosis" has emerged, highlighting the interplay between pyroptosis, apoptosis, and necroptosis, especially during cellular responses to infections. This article provides a concise overview of PANoptosis and its molecular mechanisms, exploring its implications in various diseases. The review focuses on the extensive interactions among different RCD pathways, emphasizing the role of PANoptosis in infections, cytokine storms, inflammatory diseases, and cancer. Understanding PANoptosis is crucial for developing novel treatments for conditions involving infections, sterile inflammations, and cancer.
Collapse
Affiliation(s)
- Hyun Bae
- Department of Biological Sciences, College of Natural Science, Seoul National University, Seoul, 08826, South Korea
| | - Yeonseo Jang
- Department of Biological Sciences, College of Natural Science, Seoul National University, Seoul, 08826, South Korea
| | - Rajendra Karki
- Department of Biological Sciences, College of Natural Science, Seoul National University, Seoul, 08826, South Korea.
- Nexus Institute of Research and Innovation (NIRI), Kathmandu, Nepal.
| | - Joo-Hui Han
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Woosuk University, Wanju, 55338, Republic of Korea.
| |
Collapse
|
24
|
Song Q, Fan Y, Zhang H, Wang N. Z-DNA binding protein 1 orchestrates innate immunity and inflammatory cell death. Cytokine Growth Factor Rev 2024; 77:15-29. [PMID: 38548490 DOI: 10.1016/j.cytogfr.2024.03.005] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Revised: 03/17/2024] [Accepted: 03/20/2024] [Indexed: 06/22/2024]
Abstract
Innate immunity is not only the first line of host defense against microbial infections but is also crucial for the host responses against a variety of noxious stimuli. Z-DNA binding protein 1 (ZBP1) is a cytosolic nucleic acid sensor that can induce inflammatory cell death in both immune and nonimmune cells upon sensing of incursive virus-derived Z-form nucleic acids and self-nucleic acids via its Zα domain. Mechanistically, aberrantly expressed or activated ZBP1 induced by pathogens or noxious stimuli enables recruitment of TANK binding kinase 1 (TBK1), interferon regulatory factor 3 (IRF3), receptor-interacting serine/threonine-protein kinase 1 (RIPK1) and RIPK3 to drive type I interferon (IFN-I) responses and activation of nuclear factor kappa B (NF-κB) signaling. Meanwhile, ZBP1 promotes the assembly of ZBP1- and absent in melanoma 2 (AIM2)-PANoptosome, which ultimately triggers PANoptosis through caspase 3-mediated apoptosis, mixed lineage kinase domain like pseudokinase (MLKL)-mediated necroptosis, and gasdermin D (GSDMD)-mediated pyroptosis. In response to damaged mitochondrial DNA, ZBP1 can interact with cyclic GMP-AMP synthase to augment IFN-I responses but inhibits toll like receptor 9-mediated inflammatory responses. This review summarizes the structure and expression pattern of ZBP1, discusses its roles in human diseases through immune-dependent (e.g., the production of IFN-I and pro-inflammatory cytokines) and -independent (e.g., the activation of cell death) functions, and highlights the attractive prospect of manipulating ZBP1 as a promising therapeutic target in diseases.
Collapse
Affiliation(s)
- Qixiang Song
- Department of Pathophysiology, School of Basic Medical Science, Central South University, 110 Xiangya Road, Changsha 410083, China; Key Laboratory of Sepsis Translational Medicine of Hunan, Central South University, 110 Xiangya Road, Changsha 410083, China
| | - Yuhang Fan
- Department of Pathophysiology, School of Basic Medical Science, Central South University, 110 Xiangya Road, Changsha 410083, China; Key Laboratory of Sepsis Translational Medicine of Hunan, Central South University, 110 Xiangya Road, Changsha 410083, China
| | - Huali Zhang
- Department of Pathophysiology, School of Basic Medical Science, Central South University, 110 Xiangya Road, Changsha 410083, China; Key Laboratory of Sepsis Translational Medicine of Hunan, Central South University, 110 Xiangya Road, Changsha 410083, China.
| | - Nian Wang
- Department of Pathophysiology, School of Basic Medical Science, Central South University, 110 Xiangya Road, Changsha 410083, China; Key Laboratory of Sepsis Translational Medicine of Hunan, Central South University, 110 Xiangya Road, Changsha 410083, China.
| |
Collapse
|
25
|
Zheng Z, Li K, Yang Z, Wang X, Shen C, Zhang Y, Lu H, Yin Z, Sha M, Ye J, Zhu L. Transcriptomic analysis reveals molecular characterization and immune landscape of PANoptosis-related genes in atherosclerosis. Inflamm Res 2024; 73:961-978. [PMID: 38587531 DOI: 10.1007/s00011-024-01877-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Revised: 03/01/2024] [Accepted: 03/27/2024] [Indexed: 04/09/2024] Open
Abstract
BACKGROUND Atherosclerosis is a chronic inflammatory disease characterized by abnormal lipid deposition in the arteries. Programmed cell death is involved in the inflammatory response of atherosclerosis, but PANoptosis, as a new form of programmed cell death, is still unclear in atherosclerosis. This study explored the key PANoptosis-related genes involved in atherosclerosis and their potential mechanisms through bioinformatics analysis. METHODS We evaluated differentially expressed genes (DEGs) and immune infiltration landscape in atherosclerosis using microarray datasets and bioinformatics analysis. By intersecting PANoptosis-related genes from the GeneCards database with DEGs, we obtained a set of PANoptosis-related genes in atherosclerosis (PANoDEGs). Functional enrichment analysis of PANoDEGs was performed and protein-protein interaction (PPI) network of PANoDEGs was established. The machine learning algorithms were used to identify the key PANoDEGs closely linked to atherosclerosis. Receiver operating characteristic (ROC) analysis was used to assess the diagnostic potency of key PANoDEGs. CIBERSORT was used to analyze the immune infiltration patterns in atherosclerosis, and the Spearman method was used to study the relationship between key PANoDEGs and immune infiltration abundance. The single gene enrichment analysis of key PANoDEGs was investigated by GSEA. The transcription factors and target miRNAs of key PANoDEGs were predicted by Cytoscape and online database, respectively. The expression of key PANoDEGs was validated through animal and cell experiments. RESULTS PANoDEGs in atherosclerosis were significantly enriched in apoptotic process, pyroptosis, necroptosis, cytosolic DNA-sensing pathway, NOD-like receptor signaling pathway, lipid and atherosclerosis. Four key PANoDEGs (ZBP1, SNHG6, DNM1L, and AIM2) were found to be closely related to atherosclerosis. The ROC curve analysis demonstrated that the key PANoDEGs had a strong diagnostic potential in distinguishing atherosclerotic samples from control samples. Immune cell infiltration analysis revealed that the proportion of initial B cells, plasma cells, CD4 memory resting T cells, and M1 macrophages was significantly higher in atherosclerotic tissues compared to normal tissues. Spearman analysis showed that key PANoDEGs showed strong correlations with immune cells such as T cells, macrophages, plasma cells, and mast cells. The regulatory networks of the four key PANoDEGs were established. The expression of key PANoDEGs was verified in further cell and animal experiments. CONCLUSIONS This study evaluated the expression changes of PANoptosis-related genes in atherosclerosis, providing a reference direction for the study of PANoptosis in atherosclerosis and offering potential new avenues for further understanding the pathogenesis and treatment strategies of atherosclerosis.
Collapse
Affiliation(s)
| | - Kaiyuan Li
- Dalian Medical University, Dalian, 116000, China
| | - Zhiyuan Yang
- Dalian Medical University, Dalian, 116000, China
| | - Xiaowen Wang
- Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Cheng Shen
- Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Yubin Zhang
- Dalian Medical University, Dalian, 116000, China
| | - Huimin Lu
- Taizhou People's Hospital Affiliated to Nanjing Medical University, Taizhou, 225399, China
| | - Zhifeng Yin
- Jiangsu Hanjiang Biotechnology Co., LTD, Taizhou, 225399, China
| | - Min Sha
- Taizhou People's Hospital Affiliated to Nanjing Medical University, Taizhou, 225399, China.
| | - Jun Ye
- Taizhou People's Hospital Affiliated to Nanjing Medical University, Taizhou, 225399, China.
| | - Li Zhu
- Dalian Medical University, Dalian, 116000, China.
- Nanjing University of Chinese Medicine, Nanjing, 210023, China.
- Taizhou People's Hospital Affiliated to Nanjing Medical University, Taizhou, 225399, China.
| |
Collapse
|
26
|
Liu S, Joshi K, Zhang L, Li W, Mack R, Runde A, Hagen PA, Barton K, Breslin P, Ji HL, Kini AR, Wang Z, Zhang J. Caspase 8 deletion causes infection/inflammation-induced bone marrow failure and MDS-like disease in mice. Cell Death Dis 2024; 15:278. [PMID: 38637559 PMCID: PMC11026525 DOI: 10.1038/s41419-024-06660-3] [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: 10/20/2023] [Revised: 04/01/2024] [Accepted: 04/08/2024] [Indexed: 04/20/2024]
Abstract
Myelodysplastic syndromes (MDS) are a heterogeneous group of pre-leukemic hematopoietic disorders characterized by cytopenia in peripheral blood due to ineffective hematopoiesis and normo- or hypercellularity and morphologic dysplasia in bone marrow (BM). An inflammatory BM microenvironment and programmed cell death of hematopoietic stem/progenitor cells (HSPCs) are thought to be the major causes of ineffective hematopoiesis in MDS. Pyroptosis, apoptosis and necroptosis (collectively, PANoptosis) are observed in BM tissues of MDS patients, suggesting an important role of PANoptosis in MDS pathogenesis. Caspase 8 (Casp8) is a master regulator of PANoptosis, which is downregulated in HSPCs from most MDS patients and abnormally spliced in HSPCs from MDS patients with SRSF2 mutation. To study the role of PANoptosis in hematopoiesis, we generated inducible Casp8 knockout mice (Casp8-/-). Mx1-Cre-Casp8-/- mice died of BM failure within 10 days of polyI:C injections due to depletion of HSPCs. Rosa-ERT2Cre-Casp8-/- mice are healthy without significant changes in BM hematopoiesis within the first 1.5 months after Casp8 deletion. Such mice developed BM failure upon infection or low dose polyI:C/LPS injections due to the hypersensitivity of Casp8-/- HSPCs to infection or inflammation-induced necroptosis which can be prevented by Ripk3 deletion. However, impaired self-renewal capacity of Casp8-/- HSPCs cannot be rescued by Ripk3 deletion due to activation of Ripk1-Tbk1 signaling. Most importantly, mice transplanted with Casp8-/- BM cells developed MDS-like disease within 4 months of transplantation as demonstrated by anemia, thrombocytopenia and myelodysplasia. Our study suggests an essential role for a balance in Casp8, Ripk3-Mlkl and Ripk1-Tbk1 activities in the regulation of survival and self-renewal of HSPCs, the disruption of which induces inflammation and BM failure, resulting in MDS-like disease.
Collapse
Affiliation(s)
- Shanhui Liu
- Oncology Institute, Cardinal Bernardin Cancer Canter, Loyola University Chicago Medical Center, Maywood, IL, 60153, USA
- Department of Cancer Biology, Loyola University Chicago Medical Center, Maywood, IL, 60153, USA
- Lanzhou University Second Hospital, Key Laboratory of Urological Diseases in Gansu Province, Lanzhou, Gansu, 730030, China
| | - Kanak Joshi
- Oncology Institute, Cardinal Bernardin Cancer Canter, Loyola University Chicago Medical Center, Maywood, IL, 60153, USA
- Department of Cancer Biology, Loyola University Chicago Medical Center, Maywood, IL, 60153, USA
| | - Lei Zhang
- Oncology Institute, Cardinal Bernardin Cancer Canter, Loyola University Chicago Medical Center, Maywood, IL, 60153, USA
- Department of Cancer Biology, Loyola University Chicago Medical Center, Maywood, IL, 60153, USA
- Cyrus Tang Hematology Center, Collaborative Innovation Center of Hematology, National Clinical Research Center for Hematologic Diseases, Soochow University, Suzhou, 215123, China
| | - Wenyan Li
- Lanzhou University Second Hospital, Key Laboratory of Urological Diseases in Gansu Province, Lanzhou, Gansu, 730030, China
| | - Ryan Mack
- Oncology Institute, Cardinal Bernardin Cancer Canter, Loyola University Chicago Medical Center, Maywood, IL, 60153, USA
- Department of Cancer Biology, Loyola University Chicago Medical Center, Maywood, IL, 60153, USA
| | - Austin Runde
- Oncology Institute, Cardinal Bernardin Cancer Canter, Loyola University Chicago Medical Center, Maywood, IL, 60153, USA
- Department of Cancer Biology, Loyola University Chicago Medical Center, Maywood, IL, 60153, USA
| | - Patrick A Hagen
- Oncology Institute, Cardinal Bernardin Cancer Canter, Loyola University Chicago Medical Center, Maywood, IL, 60153, USA
- Department of Medicine, Loyola University Chicago Medical Center, Maywood, IL, 60153, USA
| | - Kevin Barton
- Oncology Institute, Cardinal Bernardin Cancer Canter, Loyola University Chicago Medical Center, Maywood, IL, 60153, USA
- Department of Medicine, Loyola University Chicago Medical Center, Maywood, IL, 60153, USA
| | - Peter Breslin
- Oncology Institute, Cardinal Bernardin Cancer Canter, Loyola University Chicago Medical Center, Maywood, IL, 60153, USA
- Department of Cancer Biology, Loyola University Chicago Medical Center, Maywood, IL, 60153, USA
- Departments of Biology and Molecular/Cellular Physiology, Loyola University Chicago, Maywood, IL, 60153, USA
| | - Hong-Long Ji
- Department of Surgery, Loyola University Chicago Medical Center, Maywood, IL, 60153, USA
| | - Ameet R Kini
- Departments of Pathology and Radiation Oncology, Loyola University Chicago Medical Center, Maywood, IL, 60153, USA
| | - Zhiping Wang
- Lanzhou University Second Hospital, Key Laboratory of Urological Diseases in Gansu Province, Lanzhou, Gansu, 730030, China.
| | - Jiwang Zhang
- Oncology Institute, Cardinal Bernardin Cancer Canter, Loyola University Chicago Medical Center, Maywood, IL, 60153, USA.
- Department of Cancer Biology, Loyola University Chicago Medical Center, Maywood, IL, 60153, USA.
- Departments of Pathology and Radiation Oncology, Loyola University Chicago Medical Center, Maywood, IL, 60153, USA.
| |
Collapse
|
27
|
Zheng Y, Li J, Liu B, Xie Z, He Y, Xue D, Zhao D, Hao C. Global trends in PANoptosis research: bibliometrics and knowledge graph analysis. Apoptosis 2024; 29:229-242. [PMID: 37751105 DOI: 10.1007/s10495-023-01889-3] [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] [Accepted: 08/30/2023] [Indexed: 09/27/2023]
Abstract
PANoptosis has recently been discovered as a new type of cell death. PANoptosis mainly refers to the significant interaction among the three programmed cell death pathways of apoptosis, necroptosis, and pyroptosis. Despite this, only a few studies have examined the systematic literature in this area. By analyzing the bibliometric data for PANoptosis, we can visualize the current hotspots and predicted trends in research. This study analyzed bibliometric indicators using the Histcite Pro 2.0 tool, which searches the Web of Science for PANoptosis literature published between 2016 and 2022. A bibliometric analysis was performed using Histcite Pro 2.0, while research trends and hotspots were visualized using VOSviewer, CiteSpace and BioBERT. The output of related literature was low in the four years from the first presentation of PANoptosis in 2016 to 2020. The volume of relevant literature grew exponentially between 2020 and 2022. The United States and China play a leading role in this field. Although China started late, its research in this field is developing rapidly. As research progressed, more focus was placed on the relationship between PANoptosis and pyroptosis, as well as apoptosis and necrosis. Now is a rapid development stage of PANoptosis research. Most of the research focuses on the cellular level, and the focus is more on the treatment of tumor-related diseases. The current focus of this area is PANoptosis mechanisms in cancer and inflammation. It can be seen from the burst analysis of keywords that caspase1 and host defense have consistently been research hotspots in the field of PANoptosis, while the frequency of NLRC4, causes of autoinflammation, recognition, NLRP3, and Gasdermin D has gradually increased, all of which have become research hotspots in recent years. Finally, we used the BioBERT biomedical language model to mine the most documented genes and diseases in the PANoptosis field articles, pointing out the direction for subsequent research steps. According to a bibliometric analysis, researchers have shown an increased interest in PANoptosis over the past few years. Researchers initially focused on the molecular mechanism of PANoptosis and pyroptosis, apoptosis, and necroptosis. The role of PANoptosis in diseases and conditions such as inflammation and tumors is one of the current research hotspots in this area. The focus is more on treating inflammation-related diseases, which will become the key development direction of future research.
Collapse
Affiliation(s)
- Yi Zheng
- Department of General Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China
- Key Laboratory of Hepatosplenic Surgery, Ministry of Education, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Jiachen Li
- Department of General Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China
- Key Laboratory of Hepatosplenic Surgery, Ministry of Education, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Bo Liu
- Department of General Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China
- Key Laboratory of Hepatosplenic Surgery, Ministry of Education, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Zhihong Xie
- Department of General Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China
- Key Laboratory of Hepatosplenic Surgery, Ministry of Education, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Yuanhang He
- Department of General Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China
- Key Laboratory of Hepatosplenic Surgery, Ministry of Education, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Dongbo Xue
- Department of General Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China.
- Key Laboratory of Hepatosplenic Surgery, Ministry of Education, The First Affiliated Hospital of Harbin Medical University, Harbin, China.
| | - Dali Zhao
- Department of General Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China.
- Key Laboratory of Hepatosplenic Surgery, Ministry of Education, The First Affiliated Hospital of Harbin Medical University, Harbin, China.
| | - Chenjun Hao
- Department of General Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China.
- Key Laboratory of Hepatosplenic Surgery, Ministry of Education, The First Affiliated Hospital of Harbin Medical University, Harbin, China.
| |
Collapse
|
28
|
Wu K, Li B, Zhang X, Fang Y, Zeng S, Hu W, Liu X, Liu X, Lu Z, Li X, Chen W, Qin Y, Zhou B, Zou L, Zhao F, Yi L, Zhao M, Fan S, Chen J. CSFV restricts necroptosis to sustain infection by inducing autophagy/mitophagy-targeted degradation of RIPK3. Microbiol Spectr 2024; 12:e0275823. [PMID: 38100396 PMCID: PMC10782971 DOI: 10.1128/spectrum.02758-23] [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: 07/04/2023] [Accepted: 11/10/2023] [Indexed: 12/17/2023] Open
Abstract
IMPORTANCE CSFV infection in pigs causes persistent high fever, hemorrhagic necrotizing multi-organ inflammation, and high mortality, which seriously threatens the global swine industry. Cell death is an essential immune response of the host against pathogen invasion, and lymphopenia is the most typical clinical feature in the acute phase of CSFV infection, which affects the initial host antiviral immunity. As an "old" virus, CSFV has evolved mechanisms to evade host immune response after a long genetic evolution. Here, we show that necroptosis is a limiting host factor for CSFV infection and that CSFV-induced autophagy can subvert this host defense mechanism to promote its sustained replication. Our findings reveal a complex link between necroptosis and autophagy in the process of cell death, provide evidence supporting the important role for CSFV in counteracting host cell necrosis, and enrich our knowledge of pathogens that may subvert and evade this host defense.
Collapse
Affiliation(s)
- Keke Wu
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
- Agro-Biological Gene Research Center, Guangdong Academy of Agricultural Sciences, State Key Laboratory of Livestock and Poultry Breeding industry, Guangzhou, China
| | - Bingke Li
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
- Agro-Biological Gene Research Center, Guangdong Academy of Agricultural Sciences, State Key Laboratory of Livestock and Poultry Breeding industry, Guangzhou, China
| | - Xiaoai Zhang
- Agro-Biological Gene Research Center, Guangdong Academy of Agricultural Sciences, State Key Laboratory of Livestock and Poultry Breeding industry, Guangzhou, China
| | - Yiqi Fang
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
- Key Laboratory of Zoonosis Prevention and Control of Guangdong Province, South China Agricultural University, Guangzhou, China
| | - Sen Zeng
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
- Key Laboratory of Zoonosis Prevention and Control of Guangdong Province, South China Agricultural University, Guangzhou, China
| | - Wenshuo Hu
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
- Key Laboratory of Zoonosis Prevention and Control of Guangdong Province, South China Agricultural University, Guangzhou, China
| | - Xiaodi Liu
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
- Key Laboratory of Zoonosis Prevention and Control of Guangdong Province, South China Agricultural University, Guangzhou, China
| | - Xueyi Liu
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
- Key Laboratory of Zoonosis Prevention and Control of Guangdong Province, South China Agricultural University, Guangzhou, China
| | - Zhimin Lu
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
- Key Laboratory of Zoonosis Prevention and Control of Guangdong Province, South China Agricultural University, Guangzhou, China
| | - Xiaowen Li
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
- Key Laboratory of Zoonosis Prevention and Control of Guangdong Province, South China Agricultural University, Guangzhou, China
| | - Wenxian Chen
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
- Key Laboratory of Zoonosis Prevention and Control of Guangdong Province, South China Agricultural University, Guangzhou, China
| | - Yuwei Qin
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
- Key Laboratory of Zoonosis Prevention and Control of Guangdong Province, South China Agricultural University, Guangzhou, China
| | - Bolun Zhou
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
- Key Laboratory of Zoonosis Prevention and Control of Guangdong Province, South China Agricultural University, Guangzhou, China
| | - Linke Zou
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
- Key Laboratory of Zoonosis Prevention and Control of Guangdong Province, South China Agricultural University, Guangzhou, China
| | - Feifan Zhao
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
- Key Laboratory of Zoonosis Prevention and Control of Guangdong Province, South China Agricultural University, Guangzhou, China
| | - Lin Yi
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
- Agro-Biological Gene Research Center, Guangdong Academy of Agricultural Sciences, State Key Laboratory of Livestock and Poultry Breeding industry, Guangzhou, China
- Key Laboratory of Zoonosis Prevention and Control of Guangdong Province, South China Agricultural University, Guangzhou, China
| | - Mingqiu Zhao
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
- Agro-Biological Gene Research Center, Guangdong Academy of Agricultural Sciences, State Key Laboratory of Livestock and Poultry Breeding industry, Guangzhou, China
- Key Laboratory of Zoonosis Prevention and Control of Guangdong Province, South China Agricultural University, Guangzhou, China
| | - Shuangqi Fan
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
- Agro-Biological Gene Research Center, Guangdong Academy of Agricultural Sciences, State Key Laboratory of Livestock and Poultry Breeding industry, Guangzhou, China
- Key Laboratory of Zoonosis Prevention and Control of Guangdong Province, South China Agricultural University, Guangzhou, China
| | - Jinding Chen
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
- Agro-Biological Gene Research Center, Guangdong Academy of Agricultural Sciences, State Key Laboratory of Livestock and Poultry Breeding industry, Guangzhou, China
- Key Laboratory of Zoonosis Prevention and Control of Guangdong Province, South China Agricultural University, Guangzhou, China
| |
Collapse
|
29
|
Mei X, Zhang Y, Wang S, Wang H, Chen R, Ma K, Yang Y, Jiang P, Feng Z, Zhang C, Zhang Z. Necroptosis in Pneumonia: Therapeutic Strategies and Future Perspectives. Viruses 2024; 16:94. [PMID: 38257794 PMCID: PMC10818625 DOI: 10.3390/v16010094] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Revised: 01/02/2024] [Accepted: 01/04/2024] [Indexed: 01/24/2024] Open
Abstract
Pneumonia remains a major global health challenge, necessitating the development of effective therapeutic approaches. Recently, necroptosis, a regulated form of cell death, has garnered attention in the fields of pharmacology and immunology for its role in the pathogenesis of pneumonia. Characterized by cell death and inflammatory responses, necroptosis is a key mechanism contributing to tissue damage and immune dysregulation in various diseases, including pneumonia. This review comprehensively analyzes the role of necroptosis in pneumonia and explores potential pharmacological interventions targeting this cell death pathway. Moreover, we highlight the intricate interplay between necroptosis and immune responses in pneumonia, revealing a bidirectional relationship between necrotic cell death and inflammatory signaling. Importantly, we assess current therapeutic strategies modulating necroptosis, encompassing synthetic inhibitors, natural products, and other drugs targeting key components of the programmed necrosis pathway. The article also discusses challenges and future directions in targeting programmed necrosis for pneumonia treatment, proposing novel therapeutic strategies that combine antibiotics with necroptosis inhibitors. This review underscores the importance of understanding necroptosis in pneumonia and highlights the potential of pharmacological interventions to mitigate tissue damage and restore immune homeostasis in this devastating respiratory infection.
Collapse
Affiliation(s)
- Xiuzhen Mei
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
- Institute of Veterinary Medicine, Jiangsu Academy of Agricultural Sciences, Nanjing 210014, China
- GuoTai (Taizhou) Center of Technology Innovation for Veterinary Biologicals, Taizhou 225300, China
| | - Yuchen Zhang
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
- Institute of Veterinary Medicine, Jiangsu Academy of Agricultural Sciences, Nanjing 210014, China
- GuoTai (Taizhou) Center of Technology Innovation for Veterinary Biologicals, Taizhou 225300, China
| | - Shu Wang
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
- Institute of Veterinary Medicine, Jiangsu Academy of Agricultural Sciences, Nanjing 210014, China
- GuoTai (Taizhou) Center of Technology Innovation for Veterinary Biologicals, Taizhou 225300, China
| | - Hui Wang
- Institute of Veterinary Medicine, Jiangsu Academy of Agricultural Sciences, Nanjing 210014, China
- GuoTai (Taizhou) Center of Technology Innovation for Veterinary Biologicals, Taizhou 225300, China
- Jiangsu Key Laboratory for Aquatic Crustacean Diseases, College of Marine Science and Engineering, Nanjing Normal University, Nanjing 210023, China
| | - Rong Chen
- Institute of Veterinary Medicine, Jiangsu Academy of Agricultural Sciences, Nanjing 210014, China
- GuoTai (Taizhou) Center of Technology Innovation for Veterinary Biologicals, Taizhou 225300, China
| | - Ke Ma
- Institute of Veterinary Medicine, Jiangsu Academy of Agricultural Sciences, Nanjing 210014, China
- GuoTai (Taizhou) Center of Technology Innovation for Veterinary Biologicals, Taizhou 225300, China
- College of Life Sciences, Nanjing Agricultural University, Nanjing 210095, China
| | - Yue Yang
- Institute of Veterinary Medicine, Jiangsu Academy of Agricultural Sciences, Nanjing 210014, China
- GuoTai (Taizhou) Center of Technology Innovation for Veterinary Biologicals, Taizhou 225300, China
- Jiangsu Key Laboratory for Aquatic Crustacean Diseases, College of Marine Science and Engineering, Nanjing Normal University, Nanjing 210023, China
| | - Ping Jiang
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
| | - Zhixin Feng
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
- Institute of Veterinary Medicine, Jiangsu Academy of Agricultural Sciences, Nanjing 210014, China
- GuoTai (Taizhou) Center of Technology Innovation for Veterinary Biologicals, Taizhou 225300, China
- Jiangsu Key Laboratory for Aquatic Crustacean Diseases, College of Marine Science and Engineering, Nanjing Normal University, Nanjing 210023, China
| | - Chao Zhang
- School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Zhenzhen Zhang
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
- Institute of Veterinary Medicine, Jiangsu Academy of Agricultural Sciences, Nanjing 210014, China
- GuoTai (Taizhou) Center of Technology Innovation for Veterinary Biologicals, Taizhou 225300, China
| |
Collapse
|
30
|
Cai H, Lv M, Wang T. PANoptosis in cancer, the triangle of cell death. Cancer Med 2023; 12:22206-22223. [PMID: 38069556 PMCID: PMC10757109 DOI: 10.1002/cam4.6803] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Revised: 11/08/2023] [Accepted: 11/27/2023] [Indexed: 12/31/2023] Open
Abstract
BACKGROUND PANoptosis is a novel form of programmed cell death (PCD) found in 2019 that is regulated by the PANoptosome. PANoptosis combines essential features of pyroptosis, apoptosis, and necroptosis, forming a "death triangle" of cells. While apoptosis, pyroptosis, and necroptosis have been extensively studied for their roles in human inflammatory diseases and many other clinical conditions, historically they were considered as independent processes. However, emerging evidence indicates that these PCDs exhibit cross talk and interactions, resulting in the development of the concept of PANoptosis. METHODS In this review, we offer a concise summary of the fundamental mechanisms of apoptosis, pyroptosis, and necroptosis. We subsequently introduce the notion of PANoptosis and detail the assembly mechanism of the PANoptosome complex which is responsible for inducing cell death. We also describe some regulatory networks of PANoptosis. RESULTS PANoptosis now has been associated with various human diseases including cancer. Although the exact function of PANoptosis in each tumor is not fully understood, it represents a prospective avenue for cancer therapy, offering promise for advancements in cancer therapy. CONCLUSIONS In the future, in-depth study of PANoptosis will continue to help us in understanding the fundamental processes underlying cell death and provide scientific support for cancer research.
Collapse
Affiliation(s)
- Hantao Cai
- The State Key Laboratory of Pharmaceutical Biotechnology, Division of Immunology, Medical School, Nanjing University, Nanjing, China
- Jiangsu Key Laboratory of Molecular Medicine, Division of Immunology, Medical School, Nanjing University, Nanjing, China
| | - Mingming Lv
- Department of Breast, Women's Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, Nanjing, China
| | - Tingting Wang
- The State Key Laboratory of Pharmaceutical Biotechnology, Division of Immunology, Medical School, Nanjing University, Nanjing, China
- Jiangsu Key Laboratory of Molecular Medicine, Division of Immunology, Medical School, Nanjing University, Nanjing, China
| |
Collapse
|
31
|
Xiong Y. The emerging role of PANoptosis in cancer treatment. Biomed Pharmacother 2023; 168:115696. [PMID: 37837884 DOI: 10.1016/j.biopha.2023.115696] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Revised: 10/05/2023] [Accepted: 10/09/2023] [Indexed: 10/16/2023] Open
Abstract
Programmed cell death (PCD) is a key mechanism for the study of anticancer drugs and has a significant impact on the development and management of cancer. A growing amount of data indicates that different kinds of PCD, particularly pyroptosis, apoptosis, and necroptosis, interact closely. Recent research has revealed the existence of the distinct inflammatory PCD modality known as PANoptosis, which is controlled by complex PANoptosome complexes built by combining elements from different PCD pathways. No single PCD route is sufficient to explain all of the physiologic effects seen in PANoptosis. Numerous studies have demonstrated that PANoptosis can successfully stop cancer cells from growing, proliferating, and developing drug resistance. As a result, it has changed the focus of targeted anticancer therapy. In this review, we outlined the molecular processes of PANoptosis activation and modulation as well as the mechanisms of innate immune cell death. In order to provide a theoretical foundation for the development of drugs targeting PANoptosis as an anti-cancer target, we also highlight the PANoptosomes discovered to date and give an overview of the implications of PANoptosis in cancer treatment.
Collapse
Affiliation(s)
- Yongai Xiong
- Department of Pharmaceutics, Key Laboratory of Basic Pharmacology of Guizhou Province, and School of Pharmacy, Zunyi Medical University, Zunyi, China; Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International, Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi, China.
| |
Collapse
|
32
|
Ke D, Zhang Z, Liu J, Chen P, Dai Y, Sun X, Chu Y, Li L. RIPK1 and RIPK3 inhibitors: potential weapons against inflammation to treat diabetic complications. Front Immunol 2023; 14:1274654. [PMID: 37954576 PMCID: PMC10639174 DOI: 10.3389/fimmu.2023.1274654] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Accepted: 10/05/2023] [Indexed: 11/14/2023] Open
Abstract
Diabetes mellitus is a metabolic disease that is characterized by chronic hyperglycemia due to a variety of etiological factors. Long-term metabolic stress induces harmful inflammation leading to chronic complications, mainly diabetic ophthalmopathy, diabetic cardiovascular complications and diabetic nephropathy. With diabetes complications being one of the leading causes of disability and death, the use of anti-inflammatories in combination therapy for diabetes is increasing. There has been increasing interest in targeting significant regulators of the inflammatory pathway, notably receptor-interacting serine/threonine-kinase-1 (RIPK1) and receptor-interacting serine/threonine-kinase-3 (RIPK3), as drug targets for managing inflammation in treating diabetes complications. In this review, we aim to provide an up-to-date summary of current research on the mechanism of action and drug development of RIPK1 and RIPK3, which are pivotal in chronic inflammation and immunity, in relation to diabetic complications which may be benefit for explicating the potential of selective RIPK1 and RIPK3 inhibitors as anti-inflammatory therapeutic agents for diabetic complications.
Collapse
Affiliation(s)
- Dan Ke
- College of Life Sciences, Mudanjiang Medical University, Mudanjiang, China
- Heilongjiang Key Laboratory of Tissue Damage and Repair, Mudanjiang Medical University, Mudanjiang, China
| | - Zhen Zhang
- Heilongjiang Key Laboratory of Tissue Damage and Repair, Mudanjiang Medical University, Mudanjiang, China
- School of First Clinical Medical College, Mudanjiang Medical University, Mudanjiang, China
| | - Jieting Liu
- College of Life Sciences, Mudanjiang Medical University, Mudanjiang, China
- Heilongjiang Key Laboratory of Tissue Damage and Repair, Mudanjiang Medical University, Mudanjiang, China
| | - Peijian Chen
- College of Life Sciences, Mudanjiang Medical University, Mudanjiang, China
- Heilongjiang Key Laboratory of Tissue Damage and Repair, Mudanjiang Medical University, Mudanjiang, China
| | - Yucen Dai
- College of Life Sciences, Mudanjiang Medical University, Mudanjiang, China
- Heilongjiang Key Laboratory of Tissue Damage and Repair, Mudanjiang Medical University, Mudanjiang, China
| | - Xinhai Sun
- Department of Thoracic Surgery, Union Hospital, Fujian Medical University, Fuzhou, China
| | - Yanhui Chu
- Heilongjiang Key Laboratory of Tissue Damage and Repair, Mudanjiang Medical University, Mudanjiang, China
| | - Luxin Li
- College of Life Sciences, Mudanjiang Medical University, Mudanjiang, China
- Heilongjiang Key Laboratory of Tissue Damage and Repair, Mudanjiang Medical University, Mudanjiang, China
| |
Collapse
|
33
|
Malireddi RKS, Sharma BR, Bynigeri RR, Wang Y, Lu J, Kanneganti TD. ZBP1 Drives IAV-Induced NLRP3 Inflammasome Activation and Lytic Cell Death, PANoptosis, Independent of the Necroptosis Executioner MLKL. Viruses 2023; 15:2141. [PMID: 38005819 PMCID: PMC10674287 DOI: 10.3390/v15112141] [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: 10/04/2023] [Revised: 10/20/2023] [Accepted: 10/21/2023] [Indexed: 11/26/2023] Open
Abstract
Influenza A virus (IAV) continues to pose a significant global health threat, causing severe respiratory infections that result in substantial annual morbidity and mortality. Recent research highlights the pivotal role of innate immunity, cell death, and inflammation in exacerbating the severity of respiratory viral diseases. One key molecule in this process is ZBP1, a well-recognized innate immune sensor for IAV infection. Upon activation, ZBP1 triggers the formation of a PANoptosome complex containing ASC, caspase-8, and RIPK3, among other molecules, leading to inflammatory cell death, PANoptosis, and NLRP3 inflammasome activation for the maturation of IL-1β and IL-18. However, the role for other molecules in this process requires further evaluation. In this study, we investigated the role of MLKL in regulating IAV-induced cell death and NLRP3 inflammasome activation. Our data indicate IAV induced inflammatory cell death through the ZBP1-PANoptosome, where caspases and RIPKs serve as core components. However, IAV-induced lytic cell death was only partially dependent on RIPK3 at later timepoints and was fully independent of MLKL throughout all timepoints tested. Additionally, NLRP3 inflammasome activation was unaffected in MLKL-deficient cells, establishing that MLKL and MLKL-dependent necroptosis do not act upstream of NLRP3 inflammasome activation, IL-1β maturation, and lytic cell death during IAV infection.
Collapse
Affiliation(s)
- R K Subbarao Malireddi
- Department of Immunology, St. Jude Children's Research Hospital, 262 Danny Thomas Pl, Memphis, TN 38105, USA
| | - Bhesh Raj Sharma
- Department of Immunology, St. Jude Children's Research Hospital, 262 Danny Thomas Pl, Memphis, TN 38105, USA
| | - Ratnakar R Bynigeri
- Department of Immunology, St. Jude Children's Research Hospital, 262 Danny Thomas Pl, Memphis, TN 38105, USA
| | - Yaqiu Wang
- Department of Immunology, St. Jude Children's Research Hospital, 262 Danny Thomas Pl, Memphis, TN 38105, USA
| | - Jianlin Lu
- Department of Immunology, St. Jude Children's Research Hospital, 262 Danny Thomas Pl, Memphis, TN 38105, USA
| | - Thirumala-Devi Kanneganti
- Department of Immunology, St. Jude Children's Research Hospital, 262 Danny Thomas Pl, Memphis, TN 38105, USA
| |
Collapse
|
34
|
Yang X, Wang J, Liu W. Molecular markers of type II alveolar epithelial cells in acute lung injury by bioinformatics analysis. Sci Rep 2023; 13:17797. [PMID: 37853056 PMCID: PMC10584938 DOI: 10.1038/s41598-023-45129-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2023] [Accepted: 10/16/2023] [Indexed: 10/20/2023] Open
Abstract
In this study, we aimed to identify molecular markers associated with type II alveolar epithelial cell injury in acute lung injury (ALI) models using bioinformatics methods. The objective was to provide new insights for the diagnosis and treatment of ALI/ARDS. We downloaded RNA SEQ datasets (GSE109913, GSE179418, and GSE119123) from the Gene Expression Omnibus (GEO) and used R language package to screen differentially expressed genes (DEGs). DEGs were annotated using Gene Ontology (GO), and their pathways were analyzed using Kyoto Encyclopedia of Genes and Genomes (KEGG). DEGs were imported into the STRING database and analyzed using Cytoscape software to determine the protein network of DEGs and calculate the top 10 nodes for the hub genes. Finally, potential therapeutic drugs for the hub genes were predicted using the DGIdb database. We identified 78 DEGs, including 70 up-regulated genes and 8 down-regulated genes. GO analysis revealed that the DEGs were mainly involved in biological processes such as granulocyte migration, response to bacterial-derived molecules, and cytokine-mediated signaling pathways. Additionally, they had cytokine activity, chemokine activity, and receptor ligand activity, and functioned in related receptor binding, CXCR chemokine receptor binding, G protein-coupled receptor binding, and other molecular functions. KEGG analysis indicated that the DEGs were mainly involved in TNF signaling pathway, IL-17 signaling pathway, NF-κB signal pathway, chemokine signal pathway, cytokine-cytokine receptor interaction signal pathway, and others. We identified eight hub genes, including IRF7, IFIT1, IFIT3, PSMB8, PSMB9, BST2, OASL2, and ZBP1, which were all up-regulated genes. We identified several hub genes of type II alveolar epithelial cells in ALI mouse models using bioinformatics analysis. These results provide new targets for understanding and treating of ALI.
Collapse
Affiliation(s)
- Xiaoting Yang
- Emergency Department, The First Hospital of China Medical University, No.155 of North Street Nanjing, Heping District, Shenyang City, 110001, Liaoning Province, China
| | - Jing Wang
- Emergency Department, The First Hospital of China Medical University, No.155 of North Street Nanjing, Heping District, Shenyang City, 110001, Liaoning Province, China
| | - Wei Liu
- Emergency Department, The First Hospital of China Medical University, No.155 of North Street Nanjing, Heping District, Shenyang City, 110001, Liaoning Province, China.
| |
Collapse
|
35
|
Wei L, Wang X, Zhou H. Interaction among inflammasome, PANoptosise, and innate immune cells in infection of influenza virus: Updated review. Immun Inflamm Dis 2023; 11:e997. [PMID: 37773712 PMCID: PMC10521376 DOI: 10.1002/iid3.997] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Revised: 08/15/2023] [Accepted: 08/18/2023] [Indexed: 10/01/2023] Open
Abstract
BACKGROUND Influenza virus (IV) is a leading cause of respiratory tract infections, eliciting responses from key innate immune cells such as Macrophages (MQs), Neutrophils, and Dendritic Cells (DCs). These cells employ diverse mechanisms to combat IV, with Inflammasomes playing a pivotal role in viral infection control. Cellular death mechanisms, including Pyroptosis, Apoptosis, and Necroptosis (collectively called PANoptosis), significantly contribute to the innate immune response. METHODS In this updated review, we delve into the intricate relationship between PANoptosis and Inflammasomes within innate immune cells (MQs, Neutrophils, and DCs) during IV infections. We explore the strategies employed by IV to evade these immune defenses and the consequences of unchecked PANoptosis and inflammasome activation, including the potential development of severe complications such as cytokine storms and tissue damage. RESULTS Our analysis underscores the interplay between PANoptosis and Inflammasomes as a critical aspect of the innate immune response against IV. We provide insights into IV's various mechanisms to subvert these immune pathways and highlight the importance of understanding these interactions to develop effective antiviral medications. CONCLUSION A comprehensive understanding of the dynamic interactions between PANoptosis, Inflammasomes, and IV is essential for advancing our knowledge of innate immune responses to viral infections. This knowledge will be invaluable in developing targeted antiviral therapies to combat IV and mitigate potential complications, including cytokine storms and tissue damage.
Collapse
Affiliation(s)
- Li Wei
- Intensive Care Unit, Huzhou Third Municipal hospitalThe Affiliated hospital of Huzhou UniversityHuzhouChina
| | - Xufang Wang
- Intensive Care Unit, Huzhou Third Municipal hospitalThe Affiliated hospital of Huzhou UniversityHuzhouChina
| | - Huifei Zhou
- Intensive Care Unit, Huzhou Third Municipal hospitalThe Affiliated hospital of Huzhou UniversityHuzhouChina
| |
Collapse
|
36
|
Chai Q, Lei Z, Liu CH. Pyroptosis modulation by bacterial effector proteins. Semin Immunol 2023; 69:101804. [PMID: 37406548 DOI: 10.1016/j.smim.2023.101804] [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: 03/21/2023] [Revised: 06/28/2023] [Accepted: 06/29/2023] [Indexed: 07/07/2023]
Abstract
Pyroptosis is a proinflammatory form of programmed cell death featured with membrane pore formation that causes cellular swelling and allows the release of intracellular inflammatory mediators. This cell death process is elicited by the activation of the pore-forming proteins named gasdermins, and is intricately orchestrated by diverse regulatory factors in mammalian hosts to exert a prompt immune response against infections. However, growing evidence suggests that bacterial pathogens have evolved to regulate host pyroptosis for evading immune clearance and establishing progressive infection. In this review, we highlight current understandings of the functional role and regulatory network of pyroptosis in host antibacterial immunity. Thereafter, we further discuss the latest advances elucidating the mechanisms by which bacterial pathogens modulate pyroptosis through adopting their effector proteins to drive infections. A better understanding of regulatory mechanisms underlying pyroptosis at the interface of host-bacterial interactions will shed new light on the pathogenesis of infectious diseases and contribute to the development of promising therapeutic strategies against bacterial pathogens.
Collapse
Affiliation(s)
- Qiyao Chai
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, China.
| | - Zehui Lei
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, China; Savaid Medical School, University of Chinese Academy of Sciences, Beijing 101408, China
| | - Cui Hua Liu
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, China; Savaid Medical School, University of Chinese Academy of Sciences, Beijing 101408, China.
| |
Collapse
|
37
|
Maelfait J, Rehwinkel J. The Z-nucleic acid sensor ZBP1 in health and disease. J Exp Med 2023; 220:e20221156. [PMID: 37450010 PMCID: PMC10347765 DOI: 10.1084/jem.20221156] [Citation(s) in RCA: 32] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Revised: 06/28/2023] [Accepted: 07/05/2023] [Indexed: 07/18/2023] Open
Abstract
Nucleic acid sensing is a central process in the immune system, with far-reaching roles in antiviral defense, autoinflammation, and cancer. Z-DNA binding protein 1 (ZBP1) is a sensor for double-stranded DNA and RNA helices in the unusual left-handed Z conformation termed Z-DNA and Z-RNA. Recent research established ZBP1 as a key upstream regulator of cell death and proinflammatory signaling. Recognition of Z-DNA/RNA by ZBP1 promotes host resistance to viral infection but can also drive detrimental autoinflammation. Additionally, ZBP1 has interesting roles in cancer and other disease settings and is emerging as an attractive target for therapy.
Collapse
Affiliation(s)
- Jonathan Maelfait
- VIB-UGent Center for Inflammation Research, Ghent, Belgium
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Jan Rehwinkel
- Medical Research Council Human Immunology Unit, Medical Research Council Weatherall Institute of Molecular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| |
Collapse
|
38
|
Karki R, Kanneganti TD. PANoptosome signaling and therapeutic implications in infection: central role for ZBP1 to activate the inflammasome and PANoptosis. Curr Opin Immunol 2023; 83:102348. [PMID: 37267644 PMCID: PMC10524556 DOI: 10.1016/j.coi.2023.102348] [Citation(s) in RCA: 38] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Revised: 04/14/2023] [Accepted: 05/03/2023] [Indexed: 06/04/2023]
Abstract
The innate immune response provides the first line of defense against infection and disease. Regulated cell death (RCD) is a key component of innate immune activation, and RCD must be tightly controlled to clear pathogens while preventing excess inflammation. Recent studies have highlighted a central role for the innate immune sensor Z-DNA-binding protein 1 (ZBP1) as an activator of a form of inflammatory RCD called PANoptosis, which is regulated by a multifaceted cell death complex called the PANoptosome. In response to influenza A virus infection, ZBP1 activates the nucleotide-binding oligomerization domain-like receptor family pyrin domain-containing protein 3 (NLRP3) inflammasome, which then acts as an integral component of the ZBP1-PANoptosome to drive inflammatory cell death, PANoptosis. In this context, the NLRP3 inflammasome is critical for caspase-1 activation and proinflammatory cytokine interleukin (IL)-1β and IL-18 maturation, but dispensable for cell death due to functional redundancies between PANoptosome molecules. Similarly, ZBP1 is also central to the absent in melanoma 2 (AIM2)-PANoptosome; this PANoptosome forms in response to Francisella novicida and herpes simplex virus 1 infection and incorporates the AIM2 inflammasome as an integral component. In this review, we will discuss the critical roles of ZBP1 in mediating innate immune responses through inflammasomes, PANoptosomes, and PANoptosis during infection. An improved understanding of the molecular mechanisms of innate immunity and cell death will be essential for the development of targeted modalities that can improve patient outcomes by mitigating severe disease.
Collapse
Affiliation(s)
- Rajendra Karki
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA; Department of Biological Sciences, Seoul National University, Seoul 08826, Republic of Korea.
| | | |
Collapse
|
39
|
Park W, Wei S, Kim BS, Kim B, Bae SJ, Chae YC, Ryu D, Ha KT. Diversity and complexity of cell death: a historical review. Exp Mol Med 2023; 55:1573-1594. [PMID: 37612413 PMCID: PMC10474147 DOI: 10.1038/s12276-023-01078-x] [Citation(s) in RCA: 120] [Impact Index Per Article: 60.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Revised: 06/22/2023] [Accepted: 07/11/2023] [Indexed: 08/25/2023] Open
Abstract
Death is the inevitable fate of all living organisms, whether at the individual or cellular level. For a long time, cell death was believed to be an undesirable but unavoidable final outcome of nonfunctioning cells, as inflammation was inevitably triggered in response to damage. However, experimental evidence accumulated over the past few decades has revealed different types of cell death that are genetically programmed to eliminate unnecessary or severely damaged cells that may damage surrounding tissues. Several types of cell death, including apoptosis, necrosis, autophagic cell death, and lysosomal cell death, which are classified as programmed cell death, and pyroptosis, necroptosis, and NETosis, which are classified as inflammatory cell death, have been described over the years. Recently, several novel forms of cell death, namely, mitoptosis, paraptosis, immunogenic cell death, entosis, methuosis, parthanatos, ferroptosis, autosis, alkaliptosis, oxeiptosis, cuproptosis, and erebosis, have been discovered and advanced our understanding of cell death and its complexity. In this review, we provide a historical overview of the discovery and characterization of different forms of cell death and highlight their diversity and complexity. We also briefly discuss the regulatory mechanisms underlying each type of cell death and the implications of cell death in various physiological and pathological contexts. This review provides a comprehensive understanding of different mechanisms of cell death that can be leveraged to develop novel therapeutic strategies for various diseases.
Collapse
Affiliation(s)
- Wonyoung Park
- Department of Korean Medical Science, School of Korean Medicine, Pusan National University, Yangsan, Gyeongsangnam-do, 50612, Republic of Korea
- Korean Medical Research Center for Healthy Aging, Pusan National University, Yangsan, Gyeongsangnam-do, 50612, Republic of Korea
| | - Shibo Wei
- Department of Precision Medicine, School of Medicine, Sungkyunkwan University School of Medicine, Suwon, Gyeonggi-do, 16419, Republic of Korea
| | - Bo-Sung Kim
- Department of Korean Medical Science, School of Korean Medicine, Pusan National University, Yangsan, Gyeongsangnam-do, 50612, Republic of Korea
- Korean Medical Research Center for Healthy Aging, Pusan National University, Yangsan, Gyeongsangnam-do, 50612, Republic of Korea
| | - Bosung Kim
- Department of Korean Medical Science, School of Korean Medicine, Pusan National University, Yangsan, Gyeongsangnam-do, 50612, Republic of Korea
- Korean Medical Research Center for Healthy Aging, Pusan National University, Yangsan, Gyeongsangnam-do, 50612, Republic of Korea
| | - Sung-Jin Bae
- Department of Molecular Biology and Immunology, Kosin University College of Medicine, Busan, 49267, Republic of Korea
| | - Young Chan Chae
- Department of Biological Sciences, UNIST, Ulsan, 44919, Republic of Korea
| | - Dongryeol Ryu
- Department of Biomedical Science and Engineering, Gwangju Institute of Science and Technology, Gwangju, 61005, Republic of Korea
| | - Ki-Tae Ha
- Department of Korean Medical Science, School of Korean Medicine, Pusan National University, Yangsan, Gyeongsangnam-do, 50612, Republic of Korea.
- Korean Medical Research Center for Healthy Aging, Pusan National University, Yangsan, Gyeongsangnam-do, 50612, Republic of Korea.
| |
Collapse
|
40
|
Deng CH, Li TQ, Zhang W, Zhao Q, Wang Y. Targeting Inflammasome Activation in Viral Infection: A Therapeutic Solution? Viruses 2023; 15:1451. [PMID: 37515138 PMCID: PMC10384481 DOI: 10.3390/v15071451] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Revised: 06/24/2023] [Accepted: 06/26/2023] [Indexed: 07/30/2023] Open
Abstract
Inflammasome activation is exclusively involved in sensing activation of innate immunity and inflammatory response during viral infection. Accumulating evidence suggests that the manipulation of inflammasome assembly or its interaction with viral proteins are critical factors in viral pathogenesis. Results from pilot clinical trials show encouraging results of NLRP3 inflammasome suppression in reducing mortality and morbidity in SARS-CoV-2-infected patients. In this article, we summarize the up-to-date understanding of inflammasomes, including NLRP3, AIM2, NLRP1, NLRP6, and NLRC4 in various viral infections, with particular focus on RNA viruses such as SARS-CoV-2, HIV, IAV, and Zika virus and DNA viruses such as herpes simplex virus 1. We also discuss the current achievement of the mechanisms involved in viral infection-induced inflammatory response, host defense, and possible therapeutic solutions.
Collapse
Affiliation(s)
- Chuan-Han Deng
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Avenida da Universidade, Taipa, Macao 999078, China
| | - Tian-Qi Li
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Avenida da Universidade, Taipa, Macao 999078, China
| | - Wei Zhang
- State Key Laboratory of Quality Research in Chinese Medicines, Macau University of Science and Technology, Av. Wai Long, Taipa, Macao 999078, China
| | - Qi Zhao
- Cancer Centre, Institute of Translational Medicine, Faculty of Health Sciences, University of Macau, Taipa, Macau 999078, China
- MoE Frontiers Science Center for Precision Oncology, University of Macau, Avenida da Universidade, Taipa, Macao 999078, China
| | - Ying Wang
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Avenida da Universidade, Taipa, Macao 999078, China
- MoE Frontiers Science Center for Precision Oncology, University of Macau, Avenida da Universidade, Taipa, Macao 999078, China
- Department of Pharmaceutical Sciences, Faculty of Health Science, University of Macau, Avenida da Universidade, Taipa, Macao 999078, China
- Minister of Education Key Laboratory of Tumor Molecular Biology, Jinan University, Guangzhou 510632, China
| |
Collapse
|
41
|
She R, Liu D, Liao J, Wang G, Ge J, Mei Z. Mitochondrial dysfunctions induce PANoptosis and ferroptosis in cerebral ischemia/reperfusion injury: from pathology to therapeutic potential. Front Cell Neurosci 2023; 17:1191629. [PMID: 37293623 PMCID: PMC10244524 DOI: 10.3389/fncel.2023.1191629] [Citation(s) in RCA: 51] [Impact Index Per Article: 25.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Accepted: 05/05/2023] [Indexed: 06/10/2023] Open
Abstract
Ischemic stroke (IS) accounts for more than 80% of the total stroke, which represents the leading cause of mortality and disability worldwide. Cerebral ischemia/reperfusion injury (CI/RI) is a cascade of pathophysiological events following the restoration of blood flow and reoxygenation, which not only directly damages brain tissue, but also enhances a series of pathological signaling cascades, contributing to inflammation, further aggravate the damage of brain tissue. Paradoxically, there are still no effective methods to prevent CI/RI, since the detailed underlying mechanisms remain vague. Mitochondrial dysfunctions, which are characterized by mitochondrial oxidative stress, Ca2+ overload, iron dyshomeostasis, mitochondrial DNA (mtDNA) defects and mitochondrial quality control (MQC) disruption, are closely relevant to the pathological process of CI/RI. There is increasing evidence that mitochondrial dysfunctions play vital roles in the regulation of programmed cell deaths (PCDs) such as ferroptosis and PANoptosis, a newly proposed conception of cell deaths characterized by a unique form of innate immune inflammatory cell death that regulated by multifaceted PANoptosome complexes. In the present review, we highlight the mechanisms underlying mitochondrial dysfunctions and how this key event contributes to inflammatory response as well as cell death modes during CI/RI. Neuroprotective agents targeting mitochondrial dysfunctions may serve as a promising treatment strategy to alleviate serious secondary brain injuries. A comprehensive insight into mitochondrial dysfunctions-mediated PCDs can help provide more effective strategies to guide therapies of CI/RI in IS.
Collapse
Affiliation(s)
- Ruining She
- Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-Cerebral Diseases, College of Integrated Traditional Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Danhong Liu
- Medical School, Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Jun Liao
- Medical School, Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Guozuo Wang
- Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-Cerebral Diseases, College of Integrated Traditional Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Jinwen Ge
- Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-Cerebral Diseases, College of Integrated Traditional Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, China
- Hunan Academy of Traditional Chinese Medicine, Changsha, Hunan, China
| | - Zhigang Mei
- Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-Cerebral Diseases, College of Integrated Traditional Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, China
- Third-Grade Pharmacological Laboratory on Chinese Medicine Approved by State Administration of Traditional Chinese Medicine, China Three Gorges University, Yichang, Hubei, China
| |
Collapse
|
42
|
Oh S, Lee S. Recent advances in ZBP1-derived PANoptosis against viral infections. Front Immunol 2023; 14:1148727. [PMID: 37261341 PMCID: PMC10228733 DOI: 10.3389/fimmu.2023.1148727] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Accepted: 05/03/2023] [Indexed: 06/02/2023] Open
Abstract
Innate immunity is an important first line of defense against pathogens, including viruses. These pathogen- and damage-associated molecular patterns (PAMPs and DAMPs, respectively), resulting in the induction of inflammatory cell death, are detected by specific innate immune sensors. Recently, Z-DNA binding protein 1 (ZBP1), also called the DNA-dependent activator of IFN regulatory factor (DAI) or DLM1, is reported to regulate inflammatory cell death as a central mediator during viral infection. ZBP1 is an interferon (IFN)-inducible gene that contains two Z-form nucleic acid-binding domains (Zα1 and Zα2) in the N-terminus and two receptor-interacting protein homotypic interaction motifs (RHIM1 and RHIM2) in the middle, which interact with other proteins with the RHIM domain. By sensing the entry of viral RNA, ZBP1 induces PANoptosis, which protects host cells against viral infections, such as influenza A virus (IAV) and herpes simplex virus (HSV1). However, some viruses, particularly coronaviruses (CoVs), induce PANoptosis to hyperactivate the immune system, leading to cytokine storm, organ failure, tissue damage, and even death. In this review, we discuss the molecular mechanism of ZBP1-derived PANoptosis and pro-inflammatory cytokines that influence the double-edged sword of results in the host cell. Understanding the ZBP1-derived PANoptosis mechanism may be critical for improving therapeutic strategies.
Collapse
|
43
|
Necroptosis of macrophage is a key pathological feature in biliary atresia via GDCA/S1PR2/ZBP1/p-MLKL axis. Cell Death Dis 2023; 14:175. [PMID: 36859525 PMCID: PMC9977961 DOI: 10.1038/s41419-023-05615-4] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2022] [Revised: 01/19/2023] [Accepted: 01/23/2023] [Indexed: 03/03/2023]
Abstract
Biliary atresia (BA) is a severe inflammatory and fibrosing neonatal cholangiopathy disease characterized by progressive obstruction of extrahepatic bile ducts, resulting in cholestasis and progressive hepatic failure. Cholestasis may play an important role in the inflammatory and fibrotic pathological processes, but its specific mechanism is still unclear. Necroptosis mediated by Z-DNA-binding protein 1 (ZBP1)/phosphorylated-mixed lineage kinase domain-like pseudokinase (p-MLKL) is a prominent pathogenic factor in inflammatory and fibrotic diseases, but its function in BA remains unclear. Here, we aim to determine the effect of macrophage necroptosis in the BA pathology, and to explore the specific molecular mechanism. We found that necroptosis existed in BA livers, which was occurred in liver macrophages. Furthermore, this process was mediated by ZBP1/p-MLKL, and the upregulated expression of ZBP1 in BA livers was correlated with liver fibrosis and prognosis. Similarly, in the bile duct ligation (BDL) induced mouse cholestatic liver injury model, macrophage necroptosis mediated by ZBP1/p-MLKL was also observed. In vitro, conjugated bile acid-glycodeoxycholate (GDCA) upregulated ZBP1 expression in mouse bone marrow-derived monocyte/macrophages (BMDMs) through sphingosine 1-phosphate receptor 2 (S1PR2), and the induction of ZBP1 was a prerequisite for the enhanced necroptosis. Finally, after selectively knocking down of macrophage S1pr2 in vivo, ZBP1/p-MLKL-mediated necroptosis was decreased, and further collagen deposition was markedly attenuated in BDL mice. Furthermore, macrophage Zbp1 or Mlkl specific knockdown also alleviated BDL-induced liver injury/fibrosis. In conclusion, GDCA/S1PR2/ZBP1/p-MLKL mediated macrophage necroptosis plays vital role in the pathogenesis of BA liver fibrosis, and targeting this process may represent a potential therapeutic strategy for BA.
Collapse
|
44
|
Abstract
Heatstroke, which is associated with circulatory failure and multiple organ dysfunction, is a heat stress-induced life-threatening condition characterized by a raised core body temperature and central nervous system dysfunction. As global warming continues to worsen, heatstroke is expected to become the leading cause of death globally. Despite the severity of this condition, the detailed mechanisms that underlie the pathogenesis of heatstroke still remain largely unknown. Z-DNA-binding protein 1 (ZBP1), also referred to as DNA-dependent activator of IFN-regulatory factors (DAI) and DLM-1, was initially identified as a tumor-associated and interferon (IFN)-inducible protein, but has recently been reported to be a Z-nucleic acid sensor that regulates cell death and inflammation; however, its biological function is not yet fully understood. In the present study, a brief review of the main regulators is presented, in which the Z-nucleic acid sensor ZBP1 was identified to be a significant factor in regulating the pathological characteristics of heatstroke through ZBP1-dependent signaling. Thus, the lethal mechanism of heatstroke is revealed, in addition to a second function of ZBP1 other than as a nucleic acid sensor.
Collapse
Affiliation(s)
- Fanglin Li
- Critical Care Medicine, The Second Xiangya Hospital, Central South University, Changsha, China
- Department of Critical Care Medicine and Hematology, The 3rd Xiangya Hospital, Central South University, Changsha, China
| | - Jiayi Deng
- Critical Care Medicine, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Qiuli He
- Department of Nephrology, The First Affiliated Hospital of Gannan Medical University, Ganzhou, China
| | - Yanjun Zhong
- Critical Care Medicine, The Second Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
45
|
Zhu P, Ke ZR, Chen JX, Li SJ, Ma TL, Fan XL. Advances in mechanism and regulation of PANoptosis: Prospects in disease treatment. Front Immunol 2023; 14:1120034. [PMID: 36845112 PMCID: PMC9948402 DOI: 10.3389/fimmu.2023.1120034] [Citation(s) in RCA: 73] [Impact Index Per Article: 36.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Accepted: 01/31/2023] [Indexed: 02/11/2023] Open
Abstract
PANoptosis, a new research hotspot at the moment, is a cell death pattern in which pyroptosis, apoptosis, and necroptosis all occur in the same cell population. In essence, PANoptosis is a highly coordinated and dynamically balanced programmed inflammatory cell death pathway that combines the main features of pyroptosis, apoptosis, and necroptosis. Many variables, such as infection, injury, or self-defect, may be involved in the occurrence of PANoptosis, with the assembly and activation of the PANoptosome being the most critical. PANoptosis has been linked to the development of multiple systemic diseases in the human body, including infectious diseases, cancer, neurodegenerative diseases, and inflammatory diseases. Therefore, it is necessary to clarify the process of occurrence, the regulatory mechanism of PANoptosis, and its relation to diseases. In this paper, we summarized the differences and relations between PANoptosis and the three types of programmed cell death, and emphatically expounded molecular mechanism and regulatory patterns of PANoptosis, with the expectation of facilitating the application of PANoptosis regulation in disease treatment.
Collapse
Affiliation(s)
- Peng Zhu
- XiangYa School of Medicine, Central South University, Changsha, Hunan, China
| | - Zhuo-Ran Ke
- XiangYa School of Medicine, Central South University, Changsha, Hunan, China
| | - Jing-Xian Chen
- XiangYa School of Medicine, Central South University, Changsha, Hunan, China
| | - Shi-Jin Li
- School of Anesthesiology, Guizhou Medical University, Guiyang, Guizhou, China
| | - Tian-Liang Ma
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Xiao-Lei Fan
- Department of Orthopedics, Honghui Hospital, Xi’an Jiaotong University, Xi’an, China
| |
Collapse
|
46
|
Nassour J, Aguiar LG, Correia A, Schmidt TT, Mainz L, Przetocka S, Haggblom C, Tadepalle N, Williams A, Shokhirev MN, Akincilar SC, Tergaonkar V, Shadel GS, Karlseder J. Telomere-to-mitochondria signalling by ZBP1 mediates replicative crisis. Nature 2023; 614:767-773. [PMID: 36755096 PMCID: PMC9946831 DOI: 10.1038/s41586-023-05710-8] [Citation(s) in RCA: 75] [Impact Index Per Article: 37.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Accepted: 01/05/2023] [Indexed: 02/10/2023]
Abstract
Cancers arise through the accumulation of genetic and epigenetic alterations that enable cells to evade telomere-based proliferative barriers and achieve immortality. One such barrier is replicative crisis-an autophagy-dependent program that eliminates checkpoint-deficient cells with unstable telomeres and other cancer-relevant chromosomal aberrations1,2. However, little is known about the molecular events that regulate the onset of this important tumour-suppressive barrier. Here we identified the innate immune sensor Z-DNA binding protein 1 (ZBP1) as a regulator of the crisis program. A crisis-associated isoform of ZBP1 is induced by the cGAS-STING DNA-sensing pathway, but reaches full activation only when associated with telomeric-repeat-containing RNA (TERRA) transcripts that are synthesized from dysfunctional telomeres. TERRA-bound ZBP1 oligomerizes into filaments on the outer mitochondrial membrane of a subset of mitochondria, where it activates the innate immune adapter protein mitochondrial antiviral-signalling protein (MAVS). We propose that these oligomerization properties of ZBP1 serve as a signal amplification mechanism, where few TERRA-ZBP1 interactions are sufficient to launch a detrimental MAVS-dependent interferon response. Our study reveals a mechanism for telomere-mediated tumour suppression, whereby dysfunctional telomeres activate innate immune responses through mitochondrial TERRA-ZBP1 complexes to eliminate cells destined for neoplastic transformation.
Collapse
Affiliation(s)
- Joe Nassour
- The Salk Institute for Biological Studies, La Jolla, CA, USA
| | | | - Adriana Correia
- The Salk Institute for Biological Studies, La Jolla, CA, USA
- Departamento de Biologia Vegetal, Faculdade de Ciências da Universidade de Lisboa (FCUL), Lisbon, Portugal
| | | | - Laura Mainz
- The Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Sara Przetocka
- The Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Candy Haggblom
- The Salk Institute for Biological Studies, La Jolla, CA, USA
| | | | - April Williams
- The Salk Institute for Biological Studies, La Jolla, CA, USA
| | | | - Semih C Akincilar
- A*STAR Division of Cancer Genetics, Institute of Molecular and Cell Biology (IMCB), Singapore, Singapore
- Therapeutics Laboratory of NFκB Signaling, Institute of Molecular and Cell Biology (IMCB), Singapore, Singapore
| | - Vinay Tergaonkar
- A*STAR Division of Cancer Genetics, Institute of Molecular and Cell Biology (IMCB), Singapore, Singapore
- Therapeutics Laboratory of NFκB Signaling, Institute of Molecular and Cell Biology (IMCB), Singapore, Singapore
- Department of Pathology, Yong Loo Lin School of Medicine, National University of Singapore (NUS), Singapore, Singapore
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore (NUS), Singapore, Singapore
| | - Gerald S Shadel
- The Salk Institute for Biological Studies, La Jolla, CA, USA.
| | - Jan Karlseder
- The Salk Institute for Biological Studies, La Jolla, CA, USA.
| |
Collapse
|
47
|
Abstract
Herpes simplex virus (HSV)-1 and HSV-2 are ubiquitous human pathogens that infect keratinized epithelial surfaces and establish lifelong latent infection in sensory neurons of the peripheral nervous system. HSV-1 causes oral cold sores, and HSV-2 causes genital lesions characterized by recurrence at the site of the initial infection. In multicellular organisms, cell death plays a pivotal role in host defense by eliminating pathogen-infected cells. Apoptosis and necrosis are readily distinguished types of cell death. Apoptosis, the main form of programmed cell death, depends on the activity of certain caspases, a family of cysteine proteases. Necroptosis, a regulated form of necrosis that is unleashed when caspase activity is compromised, requires the activation of receptor-interacting protein (RIP) kinase 3 (RIPK3) through its interaction with other RIP homotypic interaction motif (RHIM)-containing proteins such as RIPK1. To ensure lifelong infection in the host, HSV carries out sophisticated molecular strategies to evade host cell death responses during viral infection. HSV-1 is a well-characterized pathogen that encodes potent viral inhibitors that modulate both caspase activation in the apoptosis pathway and RIPK3 activation in the necroptosis pathway in a dramatic, species-specific fashion. The viral UL39-encoded viral protein ICP6, the large subunit of the virus-encoded ribonucleotide reductase, functions as a suppressor of both caspase-8 and RHIM-dependent RIPK3 activities in the natural human host. In contrast, ICP6 RHIM-mediated recruitment of RIPK3 in the nonnatural mouse host drives the direct activation of necroptosis. This chapter provides an overview of the current state of the knowledge on molecular interactions between HSV-1 viral proteins and host cell death pathways and highlights how HSV-1 manipulates cell death signals for the benefit of viral propagation.
Collapse
Affiliation(s)
- Sudan He
- Center of Systems Medicine, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100005, China.
- Suzhou Institute of Systems Medicine, Suzhou, 215123, China.
| | - Jiahuai Han
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Xiamen University, Xiamen, China.
| |
Collapse
|
48
|
Abstract
Pyroptosis is a form of lytic, programmed cell death that functions as an innate immune effector mechanism to facilitate host defense against pathogenic microorganisms, including viruses. This type of proinflammatory cell death is orchestrated by proteolytic activation of human or mouse caspase-1, mouse caspase-11 and human caspase-4 and caspase-5 in response to infectious and inflammatory stimuli. Induction of pyroptosis requires either a canonical inflammasome responsible for caspase-1 activation or a noncanonical complex composed of caspase-11 in mice or caspase-4 or caspase-5 in humans. Recent studies have identified the pore-forming protein gasdermin D, a substrate of these inflammatory caspases, as an executioner of pyroptosis. The membrane pores formed by gasdermin D facilitate release of proinflammatory cytokines IL-1β and IL-18 and consequent biologic effects of these cytokines together with other released components. Pyroptosis, like other forms of programmed cell death, helps eliminate infected cells and thereby restricts the replicative niche, undermining survival and proliferation of intracellular pathogens. This includes viruses as well as bacteria, where ample evidence supports a critical role for inflammasome effector functions and cell death in host defense. Viruses have evolved their own mechanisms to modulate inflammasome signaling and pyroptosis. Here, we review the current literature regarding the role of pyroptosis in antiviral immune responses.
Collapse
Affiliation(s)
- Teneema Kuriakose
- Department of Immunology, St. Jude Children's Research Hospital, MS #351, 262 Danny Thomas Place, 38105-3678, Memphis, TN, USA
| | - Thirumala-Devi Kanneganti
- Department of Immunology, St. Jude Children's Research Hospital, MS #351, 262 Danny Thomas Place, 38105-3678, Memphis, TN, USA.
| |
Collapse
|
49
|
Thomas PG, Shubina M, Balachandran S. ZBP1/DAI-Dependent Cell Death Pathways in Influenza A Virus Immunity and Pathogenesis. Curr Top Microbiol Immunol 2023; 442:41-63. [PMID: 31970498 DOI: 10.1007/82_2019_190] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Influenza A viruses (IAV) are members of the Orthomyxoviridae family of negative-sense RNA viruses. The greatest diversity of IAV strains is found in aquatic birds, but a subset of strains infects other avian as well as mammalian species, including humans. In aquatic birds, infection is largely restricted to the gastrointestinal tract and spread is through feces, while in humans and other mammals, respiratory epithelial cells are the primary sites supporting productive replication and transmission. IAV triggers the death of most cell types in which it replicates, both in culture and in vivo. When well controlled, such cell death is considered an effective host defense mechanism that eliminates infected cells and limits virus spread. Unchecked or inopportune cell death also results in immunopathology. In this chapter, we discuss the impact of cell death in restricting virus spread, supporting the adaptive immune response and driving pathogenesis in the mammalian respiratory tract. Recent studies have begun to shed light on the signaling pathways underlying IAV-activated cell death. These pathways, initiated by the pathogen sensor protein ZBP1 (also called DAI and DLM1), cause infected cells to undergo apoptosis, necroptosis, and pyroptosis. We outline mechanisms of ZBP1-mediated cell death signaling following IAV infection.
Collapse
Affiliation(s)
- Paul G Thomas
- Department of Immunology, St. Jude Children's Research Hospital, MS 351, 262 Danny Thomas Place, 38105, Memphis, TN, USA.
| | - Maria Shubina
- Blood Cell Development and Function Program, Fox Chase Cancer Center, Room 224 Reimann Building, 333 Cottman Ave., 19111, Philadelphia, PA, USA
| | - Siddharth Balachandran
- Blood Cell Development and Function Program, Fox Chase Cancer Center, Room 224 Reimann Building, 333 Cottman Ave., 19111, Philadelphia, PA, USA.
| |
Collapse
|
50
|
Mocarski ES. Programmed Necrosis in Host Defense. Curr Top Microbiol Immunol 2023; 442:1-40. [PMID: 37563336 DOI: 10.1007/82_2023_264] [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: 08/12/2023]
Abstract
Host control over infectious disease relies on the ability of cells in multicellular organisms to detect and defend against pathogens to prevent disease. Evolution affords mammals with a wide variety of independent immune mechanisms to control or eliminate invading infectious agents. Many pathogens acquire functions to deflect these immune mechanisms and promote infection. Following successful invasion of a host, cell autonomous signaling pathways drive the production of inflammatory cytokines, deployment of restriction factors and induction of cell death. Combined, these innate immune mechanisms attract dendritic cells, neutrophils and macrophages as well as innate lymphoid cells such as natural killer cells that all help control infection. Eventually, the development of adaptive pathogen-specific immunity clears infection and provides immune memory of the encounter. For obligate intracellular pathogens such as viruses, diverse cell death pathways make a pivotal contribution to early control by eliminating host cells before progeny are produced. Pro-apoptotic caspase-8 activity (along with caspase-10 in humans) executes extrinsic apoptosis, a nonlytic form of cell death triggered by TNF family death receptors (DRs). Over the past two decades, alternate extrinsic apoptosis and necroptosis outcomes have been described. Programmed necrosis, or necroptosis, occurs when receptor interacting protein kinase 3 (RIPK3) activates mixed lineage kinase-like (MLKL), causing cell leakage. Thus, activation of DRs, toll-like receptors (TLRs) or pathogen sensor Z-nucleic acid binding protein 1 (ZBP1) initiates apoptosis as well as necroptosis if not blocked by virus-encoded inhibitors. Mammalian cell death pathways are blocked by herpesvirus- and poxvirus-encoded cell death suppressors. Growing evidence has revealed the importance of Z-nucleic acid sensor, ZBP1, in the cell autonomous recognition of both DNA and RNA virus infection. This volume will explore the detente between viruses and cells to manage death machinery and avoid elimination to support dissemination within the host animal.
Collapse
Affiliation(s)
- Edward S Mocarski
- Robert W. Woodruff Professor Emeritus, Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, GA, 30322, USA.
- Professor Emeritus, Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA, 94305, USA.
| |
Collapse
|