1
|
Xie H, Li W, Han X, Li M, Zhao Q, Xu Y, Su H, Meng W. Identification of RIPK3 as a target of flavonoids for anti-necroptosis in vitro. Bioorg Chem 2025; 161:108503. [PMID: 40328155 DOI: 10.1016/j.bioorg.2025.108503] [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: 02/15/2025] [Revised: 04/16/2025] [Accepted: 04/20/2025] [Indexed: 05/08/2025]
Abstract
Receptor-interacting protein kinase 3 (RIPK3), a key regulator of necroptosis, has emerged as an important target for therapeutic intervention. Flavonoids are natural compounds known for their anti-inflammatory and antioxidant properties, with recent studies highlighting their potential to modulate necroptosis. In this study, we explored the potential of RIPK3 as a target for flavonoids to achieve anti-necroptosis and anti-inflammatory effects. A library of 63 flavonoids was tested for RIPK3 binding and kinase inhibition using fluorescence polarization (FP) competition assay and ADP-Glo kinase activity assay. Six flavonoids, including scutellarein, robinetin, baicalin, myricetin, baicalein, and tricetin, showed significant inhibition of RIPK3, with IC50 values ranging from 2.5 to 13.7 μM. Structural studies of tricetin and robinetin through co-crystallization and molecular docking revealed distinct binding modes of these flavonoids within the ATP-binding pocket of RIPK3. The anti-necroptosis effects of these flavonoids were further evaluated in human HT-29 cells and mouse embryonic fibroblasts (MEFs) using a TSZ-induced cell death assay, resulting in EC50 values in the tens of micromolar range. Western blot analysis demonstrated that these flavonoids inhibit the phosphorylation of RIPK3 and its downstream effector, mixed lineage kinase domain-like protein (MLKL), and disrupt the formation of RIPK1 and RIPK3 aggregates in the necroptosis pathway. These findings identify RIPK3 as a target of natural flavonoids for the first time and elucidate the molecular mechanism underlying the anti-necroptotic activity of these flavonoids.
Collapse
Affiliation(s)
- Hang Xie
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Wanchen Li
- Nottingham Ningbo China Beacons of Excellence Research and Innovation Institute, University of Nottingham Ningbo China, Ningbo 315100, China
| | - Xiaoyu Han
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Minjun Li
- Shanghai Synchrotron Radiation Facility, Shanghai Advanced Research Institute, Chinese Academy of Sciences, Shanghai 201210, China
| | - Qiang Zhao
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China; University of Chinese Academy of Sciences, Beijing 100049, China; School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Yechun Xu
- School of Pharmaceutical Science and Technology, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou 310024, China; State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China; University of Chinese Academy of Sciences, Beijing 100049, China; School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing 210023, China.
| | - Haixia Su
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China; University of Chinese Academy of Sciences, Beijing 100049, China.
| | - Weihua Meng
- Nottingham Ningbo China Beacons of Excellence Research and Innovation Institute, University of Nottingham Ningbo China, Ningbo 315100, China; Division of Population Health and Genomics, Ninewells Hospital and Medical School, University of Dundee, Dundee DD2 4BF, UK; Center for Public Health, Faculty of Medicine, Health and Life Sciences, School of Medicine, Dentistry and Biomedical Sciences, Queen's University Belfast, Belfast BT12 6BA, UK.
| |
Collapse
|
2
|
Jarabicová I, Horváth C, Marciníková A, Adameová A. Receptor-interacting protein kinase 3: A macromolecule with multiple cellular actions and its perspective in the diagnosis and treatment of heart disease. Int J Biol Macromol 2025; 314:144280. [PMID: 40389003 DOI: 10.1016/j.ijbiomac.2025.144280] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2025] [Revised: 05/02/2025] [Accepted: 05/14/2025] [Indexed: 05/21/2025]
Abstract
Receptor-interacting protein kinase 3 (RIP3), a serine/threonine kinase of the RIP family, has emerged as a critical regulator of necroptosis, a necrosis-like form of cell demise. However, recent research has revealed that overactivated RIP3 might also be involved in the regulation of other cell death forms, such as pyroptosis, autophagy, mitochondrial permeability transition pore (mPTP)-necrosis and ferroptosis, and operates in diverse cellular compartments. RIP3 can therefore affect inflammation, oxidative stress and energy metabolism, further underscoring its pivotal role in cellular homeostasis. Furthermore, elevated circulating levels of RIP3 have been observed in cardiac disorders such as heart failure, myocardial infarction, and coronary artery disease and might correlate with disease severity and worse prognostic outcomes. On the contrary, the pharmacological inhibition of RIP3 has shown protective effects due to complex mechanisms involving necroptosis retardation, prevention of immune cell infiltration, and mitigation of cardiac cells mitochondrial damage. A detailed understanding of the complexity of RIP3's function in the heart may favour its diagnostic potential and lead to the development of future therapeutic interventions.
Collapse
Affiliation(s)
- Izabela Jarabicová
- Faculty of Pharmacy, Department of Pharmacology and Toxicology, Comenius University, Bratislava, Slovak Republic.
| | - Csaba Horváth
- Faculty of Pharmacy, Department of Pharmacology and Toxicology, Comenius University, Bratislava, Slovak Republic.
| | - Andrea Marciníková
- Faculty of Pharmacy, Department of Pharmacology and Toxicology, Comenius University, Bratislava, Slovak Republic.
| | - Adriana Adameová
- Faculty of Pharmacy, Department of Pharmacology and Toxicology, Comenius University, Bratislava, Slovak Republic; Centre of Experimental Medicine, Institute for Heart Research, Slovak Academy of Sciences, Bratislava, Slovak Republic.
| |
Collapse
|
3
|
Long D, Xu Y, Li X, Zeng Y, Tang Z, Liu L, Liu Y, Zong X, Yang S, Wang D. TET2 promotes UVB-induced cell death by activating RIPK3-MLKL-necroptosis signaling. Chem Biol Interact 2025; 416:111550. [PMID: 40350054 DOI: 10.1016/j.cbi.2025.111550] [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: 03/03/2025] [Revised: 04/10/2025] [Accepted: 05/03/2025] [Indexed: 05/14/2025]
Abstract
Ultraviolet B(UVB) radiation is a leading environmental factor that induces severe photodamage. However, its pathogenic mechanisms remain incompletely understood. Our previous research has found that Ten-eleven translocation 2 (TET2) is significantly upregulated in UVB-irradiated keratinocytes. Here, this study revealed that TET2 was upregulated in photodamaged skin, including specimens from actinic keratosis (AK) patients, UVB-exposed human skin sites, and a photodamaged mouse model. TET2 deficiency in keratinocytes mitigated UVB-induced cell death and photodamage, while TET2 overexpression exacerbated these effects. Furthermore, TET2 prompted keratinocyte death and photodamage mainly by activating the RIPK3-MLKL signaling pathway, with caspase-8 activation contributing secondarily. As for the mechanism, firstly, TET2 increases the expression of RIPK3 and MLKL by promoting their DNA demethylation, and secondly, TET2 directs the binding of PLK3 to RIPK3 and MLKL, thus enhancing the RIPK3-MLKL signaling pathway activation. This work showed that TET2 increases UVB-induced keratinocyte death and photodamage by activating the RIPK3-MLKL signaling pathway. TET2 appears to have a second function that orchestrates host responses to UVB exposure.
Collapse
Affiliation(s)
- Daijing Long
- Department of Dermatology, Central South University Third Xiangya Hospital, 138 Tongzipo Road, Yuelu District, Changsha, Hunan Province, 410013, PR China
| | - Yangfan Xu
- Department of Dermatology, Central South University Third Xiangya Hospital, 138 Tongzipo Road, Yuelu District, Changsha, Hunan Province, 410013, PR China
| | - Xuemei Li
- Department of Dermatology, Central South University Third Xiangya Hospital, 138 Tongzipo Road, Yuelu District, Changsha, Hunan Province, 410013, PR China
| | - Yilan Zeng
- Department of Dermatology, Central South University Third Xiangya Hospital, 138 Tongzipo Road, Yuelu District, Changsha, Hunan Province, 410013, PR China
| | - Ziting Tang
- Department of Dermatology, Central South University Third Xiangya Hospital, 138 Tongzipo Road, Yuelu District, Changsha, Hunan Province, 410013, PR China
| | - Lulu Liu
- Department of Dermatology, Central South University Third Xiangya Hospital, 138 Tongzipo Road, Yuelu District, Changsha, Hunan Province, 410013, PR China
| | - Yuanhong Liu
- Department of Dermatology, Central South University Third Xiangya Hospital, 138 Tongzipo Road, Yuelu District, Changsha, Hunan Province, 410013, PR China
| | - Xiule Zong
- Department of Dermatology, Central South University Third Xiangya Hospital, 138 Tongzipo Road, Yuelu District, Changsha, Hunan Province, 410013, PR China
| | - Shengbo Yang
- Department of Dermatology, Central South University Third Xiangya Hospital, 138 Tongzipo Road, Yuelu District, Changsha, Hunan Province, 410013, PR China
| | - Dan Wang
- Department of Dermatology, Central South University Third Xiangya Hospital, 138 Tongzipo Road, Yuelu District, Changsha, Hunan Province, 410013, PR China.
| |
Collapse
|
4
|
Su H, Chen G, Xie H, Li W, Xiong M, He J, Hu H, Zhao W, Shao Q, Li M, Zhao Q, Xu Y. Structure-based design of potent and selective inhibitors targeting RIPK3 for eliminating on-target toxicity in vitro. Nat Commun 2025; 16:4288. [PMID: 40341069 PMCID: PMC12062462 DOI: 10.1038/s41467-025-59432-8] [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: 08/19/2024] [Accepted: 04/18/2025] [Indexed: 05/10/2025] Open
Abstract
The essential role of RIPK3 in necroptosis makes its inhibition a promising therapeutic strategy. However, the development of RIPK3 inhibitors has been hampered by on-target apoptosis and limited kinase selectivity. Inspired by the R69H mutation, which prevents on-target apoptosis by disrupting RIPK3 dimerization, we design LK-series inhibitors that effectively inhibit RIPK3 in biochemical assays and block TNF-α-induced necroptosis in both mouse L929 and human HT29 cells without inducing apoptosis. The representative compound, LK01003, shows high selectivity across a panel of 379 kinases. Our structural studies reveal that LK compounds act as Type I1/2 inhibitors, engaging a unique hydrophobic site and stabilizing an inactive conformation of RIPK3. Moreover, several type II inhibitors are also revealed to maintain RIPK3 in the inactive conformation and do not induce on-target apoptosis. These findings suggest a promising strategy for rational design of safe and selective inhibitors by locking the inactive conformation of RIPK3.
Collapse
Affiliation(s)
- Haixia Su
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, University of Chinese Academy of Sciences, Shanghai, 201203, China.
- University of Chinese Academy of Sciences, Beijing, 100049, China.
| | - Guofeng Chen
- School of Pharmaceutical Science and Technology, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, 310024, China
| | - Hang Xie
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Wanchen Li
- Nottingham Ningbo China Beacons of Excellence Research and Innovation Institute, University of Nottingham Ningbo China, Ningbo, 315100, China
| | - Muya Xiong
- School of Pharmaceutical Science and Technology, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, 310024, China
| | - Jian He
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, University of Chinese Academy of Sciences, Shanghai, 201203, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Hangchen Hu
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, University of Chinese Academy of Sciences, Shanghai, 201203, China
| | - Wenfeng Zhao
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, University of Chinese Academy of Sciences, Shanghai, 201203, China
| | - Qiang Shao
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, University of Chinese Academy of Sciences, Shanghai, 201203, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Minjun Li
- Shanghai Synchrotron Radiation Facility, Shanghai Advanced Research Institute, Chinese Academy of Sciences, Shanghai, 201204, China
| | - Qiang Zhao
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, University of Chinese Academy of Sciences, Shanghai, 201203, China.
- University of Chinese Academy of Sciences, Beijing, 100049, China.
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, 210023, China.
| | - Yechun Xu
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, University of Chinese Academy of Sciences, Shanghai, 201203, China.
- University of Chinese Academy of Sciences, Beijing, 100049, China.
- School of Pharmaceutical Science and Technology, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, 310024, China.
| |
Collapse
|
5
|
Hong YL, Wu ZX, Jiang JX, Sun YM, Ma JH, Ai JX, Wang Y, Ma D, Zhang J, Yang CQ, Li YX, Li C, Chen QL, Liu XH, Liu XS, Ma JT, Liu MM, Shi JB. Discovery of quinazoline derivatives as RIPK3 inhibitors that switch cell death from necroptosis to apoptosis for psoriasis treatment. Eur J Med Chem 2025; 294:117716. [PMID: 40398153 DOI: 10.1016/j.ejmech.2025.117716] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2025] [Revised: 04/26/2025] [Accepted: 04/30/2025] [Indexed: 05/23/2025]
Abstract
In this study, we employed a structure-based approach and step-by-step structural optimization to identify a series of quinazoline derivatives as potent receptor interacting protein kinase 3 (RIPK3) inhibitors. Among these, compound 32 emerged as the most effective inhibitor, with strong inhibition of RIPK3 (IC50 = 27 nM) and necroptosis (EC50 = 0.45 μM). Biological evaluation showed that compound 32 binds directly to RIPK3, inhibiting the phosphorylation of both RIPK3 and its downstream substrate, MLKL, thus suppressing necroptosis. Additionally, compound 32 induces Caspase-8/3-mediated apoptosis, resulting in moderate anti-proliferative effects. By converting inflammatory necroptosis to non-inflammatory apoptosis, compound 32 not only exerts anti-inflammatory effects but also reduces inflammatory hyperplasia. More importantly, compared to the known RIPK3 inhibitor HS-1371, compound 32 significantly lower toxicity in vivo in mice. In an IMQ-induced mouse model of psoriasis, compound 32 significantly alleviates skin inflammation, scaling, and hyperkeratosis, without inducing notable toxicity. This study highlights a promising therapeutic strategy for inflammatory proliferative diseases, such as psoriasis, by inhibiting RIPK3 and shifting the mode of cell death from necroptosis to apoptosis.
Collapse
Affiliation(s)
- Ya-Ling Hong
- Anhui Province Key Laboratory of Inflammation and Immune Diseases, The Key Laboratory of Antiinflammatory and Immune Medicine, Ministry of Education, School of Pharmacy, Anhui Medical University, Hefei, 230032, Anhui Province, PR China
| | - Zheng-Xing Wu
- Anhui Province Key Laboratory of Inflammation and Immune Diseases, The Key Laboratory of Antiinflammatory and Immune Medicine, Ministry of Education, School of Pharmacy, Anhui Medical University, Hefei, 230032, Anhui Province, PR China
| | - Jia-Xing Jiang
- Anhui Province Key Laboratory of Inflammation and Immune Diseases, The Key Laboratory of Antiinflammatory and Immune Medicine, Ministry of Education, School of Pharmacy, Anhui Medical University, Hefei, 230032, Anhui Province, PR China
| | - Yi-Meng Sun
- Anhui Province Key Laboratory of Inflammation and Immune Diseases, The Key Laboratory of Antiinflammatory and Immune Medicine, Ministry of Education, School of Pharmacy, Anhui Medical University, Hefei, 230032, Anhui Province, PR China
| | - Jia-Hai Ma
- Anhui Province Key Laboratory of Inflammation and Immune Diseases, The Key Laboratory of Antiinflammatory and Immune Medicine, Ministry of Education, School of Pharmacy, Anhui Medical University, Hefei, 230032, Anhui Province, PR China
| | - Jia-Xin Ai
- Anhui Province Key Laboratory of Inflammation and Immune Diseases, The Key Laboratory of Antiinflammatory and Immune Medicine, Ministry of Education, School of Pharmacy, Anhui Medical University, Hefei, 230032, Anhui Province, PR China
| | - Yu Wang
- Anhui Province Key Laboratory of Inflammation and Immune Diseases, The Key Laboratory of Antiinflammatory and Immune Medicine, Ministry of Education, School of Pharmacy, Anhui Medical University, Hefei, 230032, Anhui Province, PR China
| | - Duo Ma
- Anhui Province Key Laboratory of Inflammation and Immune Diseases, The Key Laboratory of Antiinflammatory and Immune Medicine, Ministry of Education, School of Pharmacy, Anhui Medical University, Hefei, 230032, Anhui Province, PR China
| | - Jing Zhang
- Anhui Province Key Laboratory of Inflammation and Immune Diseases, The Key Laboratory of Antiinflammatory and Immune Medicine, Ministry of Education, School of Pharmacy, Anhui Medical University, Hefei, 230032, Anhui Province, PR China
| | - Chang-Qi Yang
- Anhui Province Key Laboratory of Inflammation and Immune Diseases, The Key Laboratory of Antiinflammatory and Immune Medicine, Ministry of Education, School of Pharmacy, Anhui Medical University, Hefei, 230032, Anhui Province, PR China
| | - Yi-Xiang Li
- Anhui Province Key Laboratory of Inflammation and Immune Diseases, The Key Laboratory of Antiinflammatory and Immune Medicine, Ministry of Education, School of Pharmacy, Anhui Medical University, Hefei, 230032, Anhui Province, PR China
| | - Chong Li
- Anhui Province Key Laboratory of Inflammation and Immune Diseases, The Key Laboratory of Antiinflammatory and Immune Medicine, Ministry of Education, School of Pharmacy, Anhui Medical University, Hefei, 230032, Anhui Province, PR China
| | - Qing-Ling Chen
- Anhui Province Key Laboratory of Inflammation and Immune Diseases, The Key Laboratory of Antiinflammatory and Immune Medicine, Ministry of Education, School of Pharmacy, Anhui Medical University, Hefei, 230032, Anhui Province, PR China
| | - Xin-Hua Liu
- Anhui Province Key Laboratory of Inflammation and Immune Diseases, The Key Laboratory of Antiinflammatory and Immune Medicine, Ministry of Education, School of Pharmacy, Anhui Medical University, Hefei, 230032, Anhui Province, PR China
| | - Xue-Song Liu
- Anhui Province Key Laboratory of Inflammation and Immune Diseases, The Key Laboratory of Antiinflammatory and Immune Medicine, Ministry of Education, School of Pharmacy, Anhui Medical University, Hefei, 230032, Anhui Province, PR China
| | - Jun-Ting Ma
- Anhui Province Key Laboratory of Inflammation and Immune Diseases, The Key Laboratory of Antiinflammatory and Immune Medicine, Ministry of Education, School of Pharmacy, Anhui Medical University, Hefei, 230032, Anhui Province, PR China.
| | - Ming-Ming Liu
- Anhui Province Key Laboratory of Inflammation and Immune Diseases, The Key Laboratory of Antiinflammatory and Immune Medicine, Ministry of Education, School of Pharmacy, Anhui Medical University, Hefei, 230032, Anhui Province, PR China.
| | - Jing-Bo Shi
- Anhui Province Key Laboratory of Inflammation and Immune Diseases, The Key Laboratory of Antiinflammatory and Immune Medicine, Ministry of Education, School of Pharmacy, Anhui Medical University, Hefei, 230032, Anhui Province, PR China.
| |
Collapse
|
6
|
Boyd DF, Jordan SV, Balachandran S. ZBP1-driven cell death in severe influenza. Trends Microbiol 2025; 33:521-532. [PMID: 39809680 DOI: 10.1016/j.tim.2024.12.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2024] [Revised: 12/12/2024] [Accepted: 12/16/2024] [Indexed: 01/16/2025]
Abstract
Influenza A virus (IAV) infections can cause life-threatening illness in humans. The severity of disease is directly linked to virus replication in the alveoli of the lower respiratory tract. In particular, the lytic death of infected alveolar epithelial cells (AECs) is a major driver of influenza severity. Recent studies have begun to define the molecular mechanisms by which IAV triggers lytic cell death. Z-form nucleic-acid-binding protein 1 (ZBP1) senses replicating IAV and drives programmed cell death (PCD) in infected cells, including apoptosis and necroptosis in AECs and pyroptosis in myeloid cells. Necroptosis and pyroptosis, both lytic forms of death, contribute to pathogenesis during severe infections. Pharmacological blockade of necroptosis shows strong therapeutic potential in mouse models of lethal influenza. We suggest that targeting ZBP1-initiated necroinflammatory cell lysis, either alone or in combination antiviral drugs, will provide clinical benefit in severe influenza.
Collapse
Affiliation(s)
- David F Boyd
- Department of Molecular, Cell, and Developmental Biology, University of California, Santa Cruz, CA, USA.
| | - Summer Vaughn Jordan
- Department of Molecular, Cell, and Developmental Biology, University of California, Santa Cruz, CA, USA
| | | |
Collapse
|
7
|
Cao P, Jaeschke H, Ni HM, Ding WX. The Ways to Die: Cell Death in Liver Pathophysiology. Semin Liver Dis 2025. [PMID: 40199509 DOI: 10.1055/a-2576-4332] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 04/10/2025]
Abstract
Liver diseases are closely associated with various cell death mechanisms, including apoptosis, necroptosis, autophagy, pyroptosis, and ferroptosis. Each process contributes uniquely to the pathophysiology of liver injury and repair. Importantly, these mechanisms are not limited to hepatocytes; they also significantly involve nonparenchymal cells. This review examines the molecular pathways and regulatory mechanisms underlying these forms of cell death in hepatocytes, emphasizing their roles in several liver diseases, such as ischemia-reperfusion injury, metabolic dysfunction-associated steatotic liver disease, drug-induced liver injury, and alcohol-associated liver disease. Recent insights into ferroptosis and pyroptosis may reveal novel therapeutic targets for managing liver diseases. This review aims to provide a comprehensive overview of these cell death mechanisms in the context of liver diseases, detailing their molecular signaling pathways and implications for potential treatment strategies.
Collapse
Affiliation(s)
- Peng Cao
- Department of Pharmacology, Toxicology and Therapeutics, University of Kansas Medical Center, Kansas City, Kansas
| | - Hartmut Jaeschke
- Department of Pharmacology, Toxicology and Therapeutics, University of Kansas Medical Center, Kansas City, Kansas
| | - Hong-Min Ni
- Department of Pharmacology, Toxicology and Therapeutics, University of Kansas Medical Center, Kansas City, Kansas
| | - Wen-Xing Ding
- Department of Pharmacology, Toxicology and Therapeutics, University of Kansas Medical Center, Kansas City, Kansas
- Division of Gastroenterology, Hepatology and Mobility, Department of Internal Medicine, University of Kansas Medical Center, Kansas City, Kansas
| |
Collapse
|
8
|
Ji YW, Wen XY, Tang HP, Su WT, Xia ZY, Lei SQ. Necroptosis: a significant and promising target for intervention of cardiovascular disease. Biochem Pharmacol 2025; 237:116951. [PMID: 40268251 DOI: 10.1016/j.bcp.2025.116951] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2024] [Revised: 03/18/2025] [Accepted: 04/14/2025] [Indexed: 04/25/2025]
Abstract
Due to changes in dietary structures, population aging, and the exacerbation of metabolic risk factors, the incidence of cardiovascular disease continues to rise annually, posing a significant health burden worldwide. Cell death plays a crucial role in the onset and progression of cardiovascular diseases. As a regulated endpoint encountered by cells under adverse stress conditions, the execution of necroptosis is regulated by classicalpathways, the calmodulin-dependent protein kinases (CaMK) pathway, and mitochondria-dependent pathways, and implicated in various cardiovascular diseases, including atherosclerosis, myocardial infarction, myocardial ischemia-reperfusion injury (IRI), heart failure, diabetic cardiomyopathy, dilated cardiomyopathy, hypertrophic cardiomyopathy, chemotherapy drug-induced cardiomyopathy, and abdominal aortic aneurysm (AAA). To further investigate potential therapeutic targets for cardiovascular diseases, we also analyzed the main molecules and their inhibitors involved in necroptosis in an effort to uncover insights for treatment.
Collapse
Affiliation(s)
- Yan-Wei Ji
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Xin-Yu Wen
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, China
| | - He-Peng Tang
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Wa-Ting Su
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Zhong-Yuan Xia
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Shao-Qing Lei
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, China.
| |
Collapse
|
9
|
Ros U, Martinez-Osorio V, Valiente PA, Abdelwahab Y, Gojkovic M, Shalaby R, Zanna S, Saggau J, Wachsmuth L, Nemade HN, Zoeller J, Lottermoser H, Chen YG, Ibrahim M, Kelepouras K, Vasilikos L, Bedoya P, Espiritu RA, Müller S, Altmannova V, Tieleman DP, Weir J, Langer J, Adam M, Walczak H, Wong WWL, Liccardi G, Mollenhauer M, Pasparakis M, Peltzer N, García-Sáez AJ. MLKL activity requires a splicing-regulated, druggable intramolecular interaction. Mol Cell 2025; 85:1589-1605.e12. [PMID: 40209701 DOI: 10.1016/j.molcel.2025.03.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2023] [Revised: 02/10/2025] [Accepted: 03/17/2025] [Indexed: 04/12/2025]
Abstract
Necroptosis is an inflammatory form of regulated cell death implicated in a range of human pathologies, whose execution depends on the poorly understood pseudokinase mixed lineage kinase domain-like (MLKL). Here, we report that splicing-dependent insertion of a short amino acid sequence in the C-terminal α-helix (Hc) of MLKL abolishes cell killing activity and creates an anti-necroptotic isoform that counteracts cell death induced by the necroptosis-proficient protein in mice and humans. We show that interaction of Hc with a previously unrecognized hydrophobic groove is essential for necroptosis, which we exploited in a strategy to identify small molecules that inhibit MLKL and substantially ameliorate disease in murine models of necroptosis-driven dermatitis and abdominal aortic aneurysm. Thus, alternative splicing of microexons controls the ability of MLKL to undergo an intramolecular rearrangement essential for necroptosis with potential to guide the development of allosteric MLKL inhibitors for the treatment of human disease.
Collapse
Affiliation(s)
- Uris Ros
- Institute of Genetics and Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Cologne 50931, Germany; Max Planck Institute of Biophysics, Frankfurt am Main 60439, Germany.
| | - Veronica Martinez-Osorio
- Institute of Genetics and Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Cologne 50931, Germany
| | - Pedro A Valiente
- Center for Protein Studies, Faculty of Biology, Havana University, Havana 10400, Cuba
| | - Yasmin Abdelwahab
- Institute of Genetics and Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Cologne 50931, Germany
| | - Milos Gojkovic
- Institute of Genetics and Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Cologne 50931, Germany
| | - Raed Shalaby
- Institute of Genetics and Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Cologne 50931, Germany
| | - Silvia Zanna
- Institute of Genetics and Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Cologne 50931, Germany
| | - Julia Saggau
- Institute of Biochemistry I, Centre for Biochemistry, Faculty of Medicine, University of Cologne, Cologne 50931, Germany; Center for Molecular Medicine Cologne, Cologne 50931, Germany
| | - Laurens Wachsmuth
- Institute of Genetics and Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Cologne 50931, Germany
| | - Harshal N Nemade
- Department for Experimental Cardiology, Faculty of Medicine, University of Cologne, and Clinic III for Internal Medicine, University Hospital Cologne, Cologne 50931, Germany
| | - Jonathan Zoeller
- Max Planck Institute of Biophysics, Frankfurt am Main 60439, Germany
| | - Hannah Lottermoser
- Department for Experimental Cardiology, Faculty of Medicine, University of Cologne, and Clinic III for Internal Medicine, University Hospital Cologne, Cologne 50931, Germany
| | - Yu-Guang Chen
- Centre for Cell Death, Cancer, and Inflammation (CCCI), UCL Cancer Institute, University College London, London WC1E 6DD, UK; Division of Hematology/Oncology, Department of Internal Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei 11490, Taiwan
| | - Mohamed Ibrahim
- Institute of Genetics and Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Cologne 50931, Germany; Center for Molecular Medicine Cologne, Cologne 50931, Germany
| | - Konstantinos Kelepouras
- Institute of Biochemistry I, Centre for Biochemistry, Faculty of Medicine, University of Cologne, Cologne 50931, Germany; Center for Molecular Medicine Cologne, Cologne 50931, Germany
| | - Lazaros Vasilikos
- Institute of Experimental Immunology, University of Zürich, Zürich 8057, Switzerland
| | - Paula Bedoya
- Institute of Genetics and Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Cologne 50931, Germany
| | - Rafael A Espiritu
- Interfaculty Institute of Biochemistry, University of Tübingen, Tübingen 72076, Germany
| | - Stefan Müller
- Institute of Genetics and Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Cologne 50931, Germany
| | - Veronika Altmannova
- Friedrich Miescher Laboratory and Max Planck Institute, Tübingen 72076, Germany
| | - D Peter Tieleman
- Centre for Molecular Simulation, Department of Biological Sciences, University of Calgary, Calgary T2N 1N4, Canada
| | - John Weir
- Friedrich Miescher Laboratory and Max Planck Institute, Tübingen 72076, Germany
| | - Julian Langer
- Max Planck Institute of Biophysics, Frankfurt am Main 60439, Germany
| | - Matti Adam
- Department for Experimental Cardiology, Faculty of Medicine, University of Cologne, and Clinic III for Internal Medicine, University Hospital Cologne, Cologne 50931, Germany
| | - Henning Walczak
- Institute of Genetics and Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Cologne 50931, Germany; Institute of Biochemistry I, Centre for Biochemistry, Faculty of Medicine, University of Cologne, Cologne 50931, Germany; Centre for Cell Death, Cancer, and Inflammation (CCCI), UCL Cancer Institute, University College London, London WC1E 6DD, UK
| | - W Wei-Lynn Wong
- Institute of Experimental Immunology, University of Zürich, Zürich 8057, Switzerland
| | - Gianmaria Liccardi
- Institute of Biochemistry I, Centre for Biochemistry, Faculty of Medicine, University of Cologne, Cologne 50931, Germany; Center for Molecular Medicine Cologne, Cologne 50931, Germany
| | - Martin Mollenhauer
- Department for Experimental Cardiology, Faculty of Medicine, University of Cologne, and Clinic III for Internal Medicine, University Hospital Cologne, Cologne 50931, Germany
| | - Manolis Pasparakis
- Institute of Genetics and Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Cologne 50931, Germany
| | - Nieves Peltzer
- Institute of Genetics and Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Cologne 50931, Germany; Center for Molecular Medicine Cologne, Cologne 50931, Germany; Department of Genome Editing, Institute of Biomedical Genetics (IBMG), University of Stuttgart, Stuttgart 70569, Germany
| | - Ana J García-Sáez
- Institute of Genetics and Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Cologne 50931, Germany; Max Planck Institute of Biophysics, Frankfurt am Main 60439, Germany.
| |
Collapse
|
10
|
Hu T, Lai X, Li L, Li Y, Wang M, Zhang H, Yang Y, Zhang C, Yan Y, Wang B. UVB-Induced necroptosis of the skin cells via RIPK3-MLKL activation independent of RIPK1 kinase activity. Cell Death Discov 2025; 11:167. [PMID: 40221399 PMCID: PMC11993685 DOI: 10.1038/s41420-025-02471-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2024] [Revised: 03/29/2025] [Accepted: 04/02/2025] [Indexed: 04/14/2025] Open
Abstract
Ultraviolet B (UVB) is recognized for inducing inflammation and death of keratinocytes through the activation of death receptors and pattern recognition receptors (PRRs). Receptor-interacting protein kinase 1 (RIPK1) and RIPK3 play pivotal roles in mediating necroptosis, which can be triggered by the activation of specific death receptors and PRRs. In this study, we observed a reduction of RIPK1 protein after UVB exposure which led to activation of Nuclear factor-kappa B (NF-κB) in HaCaT cells. This activation, in turn, promoted the production of IL-1β and TNF-α. However, RIPK1 kinase remained inactive and did not participate in cell death. Interestingly, UVB radiation triggered the activation of RIPK3 independently of RIPK1 kinase activity and subsequently induced phosphorylation of mixed-lineage kinase domain-like protein (MLKL), culminating in necroptosis and inflammation of the skin. At the same time, UVB-induced activation of RIPK3 also played a role in promoting the mitochondrial apoptotic pathway of Keratinocytes. In conclusion, UVB irradiation initiates an inflammatory response via RIPK1 pathway without necessitating its enzymatic activity. Simultaneously, RIPK3 can be activated by UVB exposure independently of RIPK1's activity, resulting in necroptosis and inflammation of the skin.
Collapse
Affiliation(s)
- Tairan Hu
- Department of Dermatology, Plastic Surgery Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xiaodong Lai
- Department of Dermatology, Plastic Surgery Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Li Li
- Department of Dermatology, Plastic Surgery Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yi Li
- Department of Dermatology, Plastic Surgery Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Meng Wang
- Department of Dermatology, Plastic Surgery Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Haini Zhang
- Department of Dermatology, Plastic Surgery Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yan Yang
- Department of Dermatology, Plastic Surgery Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Chong Zhang
- Department of Dermatology, Plastic Surgery Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yan Yan
- Department of Dermatology, Plastic Surgery Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
| | - Baoxi Wang
- Department of Dermatology, Plastic Surgery Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
| |
Collapse
|
11
|
Vecchio A, Colasuonno F, Occhigrossi L, Pitolli C, Bellanca V, Ciccarone F, D'Eletto M, Di Sano F, Pagliarini V, Sette C, Piacentini M, Rossin F. Epigenetic modulation of RIPK3 by transglutaminase 2-dependent serotonylation of H3K4me3 affects necroptosis. Cell Mol Life Sci 2025; 82:154. [PMID: 40208325 PMCID: PMC11985880 DOI: 10.1007/s00018-025-05640-w] [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: 11/11/2024] [Revised: 02/22/2025] [Accepted: 02/24/2025] [Indexed: 04/11/2025]
Abstract
The receptor interacting protein kinase 3 (RIPK3) is the main player in the activation of necroptosis, a pro-inflammatory regulated cell death modality induced by many different stimuli. RIPK3 is epigenetically regulated by DNA methylation and can be expressed when its promoter is associated with H3K4me3 histone. In this study, we show that Transglutaminase 2 protein (TG2) is necessary to induce necroptosis pathway allowing the expression of Ripk3 gene. Indeed, cells lacking TG2 show a strong downregulation of Ripk3 gene and are resistant to necroptotic stimuli. TG2 is known to promote the serotonylation of H3K4me3 histone (H3K4me3Q5ser) regulating in this way the target gene expression. Interestingly, we find that TG2 interacts with both histones H3K4me3 and H3K4me3Q5ser and these post-translational modifications are associated with the Ripk3 promoter only in presence of TG2. In addition, the absence of the H3K4me3Q5ser, in cells lacking TG2, is correlated with Ripk3 gene methylation. Altogether, these results indicate that RIPK3 expression requires TG2 mediated serotonylation of H3K4me3 to prevent Ripk3 promoter methylation, thus favouring its expression and necroptosis induction.
Collapse
Affiliation(s)
- Alessio Vecchio
- Department of Biology, University of Rome "Tor Vergata", Rome, Italy
| | - Fiorella Colasuonno
- Department of Biology, University of Rome "Tor Vergata", Rome, Italy
- Department of Experimental Medicine, University of Rome "Tor Vergata", Rome, Italy
| | - Luca Occhigrossi
- Department of Biology, University of Rome "Tor Vergata", Rome, Italy
| | - Consuelo Pitolli
- Department of Neuroscience, Section of Human Anatomy, Catholic University of the Sacred Heart, Rome, Italy
| | - Veronica Bellanca
- Department of Biology, University of Rome "Tor Vergata", Rome, Italy
- Department of Experimental Medicine, University of Rome "Tor Vergata", Rome, Italy
| | - Fabio Ciccarone
- Department of Biology, University of Rome "Tor Vergata", Rome, Italy
- IRCCS San Raffaele Roma, Rome, Italy
| | - Manuela D'Eletto
- Department of Biology, University of Rome "Tor Vergata", Rome, Italy
| | - Federica Di Sano
- Department of Biology, University of Rome "Tor Vergata", Rome, Italy
| | - Vittoria Pagliarini
- Department of Neuroscience, Section of Human Anatomy, Catholic University of the Sacred Heart, Rome, Italy
- GSTEP-Organoids Research Core Facility, IRCCS Fondazione Policlinico Universitario Agostino Gemelli, Rome, Italy
| | - Claudio Sette
- Department of Neuroscience, Section of Human Anatomy, Catholic University of the Sacred Heart, Rome, Italy
- GSTEP-Organoids Research Core Facility, IRCCS Fondazione Policlinico Universitario Agostino Gemelli, Rome, Italy
| | - Mauro Piacentini
- Department of Biology, University of Rome "Tor Vergata", Rome, Italy.
- Department of Epidemiology, Preclinical Research and Advanced Diagnostics, National Institute for Infectious Diseases IRCCS "Lazzaro Spallanzani", Rome, Italy.
- University of Rome "Tor Vergata", Via della Ricerca scientifica 1, Rome, 00133, Italy.
| | - Federica Rossin
- Department of Biology, University of Rome "Tor Vergata", Rome, Italy.
- University of Rome "Tor Vergata", Via della Ricerca scientifica 1, Rome, 00133, Italy.
| |
Collapse
|
12
|
Ma D, Hu S, Wang C, Ai J, Ma J, Gao T, Hong Y, Wu Z, Gu M, Tang X, Chang Y, Chen Q, Chen S, Yu Q, Yang J, Zhang C, Li C, Liu X, Shi J, Liu X, Liu Y, Liu M. Discovery of Potent and Balanced Dual RIPK2 and 3 Inhibitors as a New Strategy for the Treatment of Inflammatory Bowel Diseases. J Med Chem 2025; 68:7539-7559. [PMID: 40131099 DOI: 10.1021/acs.jmedchem.4c03226] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/26/2025]
Abstract
Receptor-interacting serine/threonine protein kinase 2 (RIPK2) and RIPK3 have been demonstrated to be promising targets for treating multiple inflammatory diseases, including inflammatory bowel diseases (IBDs). Due to the complexity of IBD pathogenesis, on the basis of synergy strategies, we herein describe the discovery and optimization of a series of N,7-diaryl-quinazolin-4-amine derivatives as dual RIPK2 and RIPK3 inhibitors. Based on a step-by-step process involving three rounds of structural modifications, compound 29 was identified as the most one, exhibiting balanced potency against RIPK2 (IC50 = 12 nM) and RIPK3 (IC50 = 18 nM), as well as demonstrating good selectivity over other kinase targets. Further biological evaluation confirmed that compound 29 could bind directly to RIPK2 and RIPK3, effectively suppressing NOD-induced cytokine production and cellular necroptosis. Notably, compound 29 displayed significant therapeutic effects in a DSS-induced colitis mouse model, with no detectable toxicity, indicating its promising therapeutic potential as RIPK2/RIPK3 dual inhibitors for treatment of IBD.
Collapse
Affiliation(s)
- Duo Ma
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, The Key Laboratory of Antiinflammatory and Immune Medicine, Ministry of Education, School of Pharmacy, Anhui Medical University, Hefei 230032, China
| | - Shuang Hu
- Department of Pharmacy, Eye & ENT Hospital of Fudan University, Shanghai 200031, China
| | - Chun Wang
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, The Key Laboratory of Antiinflammatory and Immune Medicine, Ministry of Education, School of Pharmacy, Anhui Medical University, Hefei 230032, China
| | - Jiaxin Ai
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, The Key Laboratory of Antiinflammatory and Immune Medicine, Ministry of Education, School of Pharmacy, Anhui Medical University, Hefei 230032, China
| | - Jiahai Ma
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, The Key Laboratory of Antiinflammatory and Immune Medicine, Ministry of Education, School of Pharmacy, Anhui Medical University, Hefei 230032, China
| | - Tianwen Gao
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, The Key Laboratory of Antiinflammatory and Immune Medicine, Ministry of Education, School of Pharmacy, Anhui Medical University, Hefei 230032, China
- Department of Pharmacy, Fuyang Hospital of Anhui Medical University, Fuyang 236112, China
| | - Yaling Hong
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, The Key Laboratory of Antiinflammatory and Immune Medicine, Ministry of Education, School of Pharmacy, Anhui Medical University, Hefei 230032, China
| | - Zhengxing Wu
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, The Key Laboratory of Antiinflammatory and Immune Medicine, Ministry of Education, School of Pharmacy, Anhui Medical University, Hefei 230032, China
| | - Mingzhen Gu
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, The Key Laboratory of Antiinflammatory and Immune Medicine, Ministry of Education, School of Pharmacy, Anhui Medical University, Hefei 230032, China
| | - XiaoXin Tang
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, The Key Laboratory of Antiinflammatory and Immune Medicine, Ministry of Education, School of Pharmacy, Anhui Medical University, Hefei 230032, China
| | - YanTai Chang
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, The Key Laboratory of Antiinflammatory and Immune Medicine, Ministry of Education, School of Pharmacy, Anhui Medical University, Hefei 230032, China
| | - QiHang Chen
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, The Key Laboratory of Antiinflammatory and Immune Medicine, Ministry of Education, School of Pharmacy, Anhui Medical University, Hefei 230032, China
| | - Shuo Chen
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, The Key Laboratory of Antiinflammatory and Immune Medicine, Ministry of Education, School of Pharmacy, Anhui Medical University, Hefei 230032, China
| | - Qing Yu
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, The Key Laboratory of Antiinflammatory and Immune Medicine, Ministry of Education, School of Pharmacy, Anhui Medical University, Hefei 230032, China
| | - JunJie Yang
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, The Key Laboratory of Antiinflammatory and Immune Medicine, Ministry of Education, School of Pharmacy, Anhui Medical University, Hefei 230032, China
| | - Chen Zhang
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, The Key Laboratory of Antiinflammatory and Immune Medicine, Ministry of Education, School of Pharmacy, Anhui Medical University, Hefei 230032, China
| | - Chong Li
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, The Key Laboratory of Antiinflammatory and Immune Medicine, Ministry of Education, School of Pharmacy, Anhui Medical University, Hefei 230032, China
| | - Xuesong Liu
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, The Key Laboratory of Antiinflammatory and Immune Medicine, Ministry of Education, School of Pharmacy, Anhui Medical University, Hefei 230032, China
| | - Jingbo Shi
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, The Key Laboratory of Antiinflammatory and Immune Medicine, Ministry of Education, School of Pharmacy, Anhui Medical University, Hefei 230032, China
| | - Xinhua Liu
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, The Key Laboratory of Antiinflammatory and Immune Medicine, Ministry of Education, School of Pharmacy, Anhui Medical University, Hefei 230032, China
| | - Yuhai Liu
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, The Key Laboratory of Antiinflammatory and Immune Medicine, Ministry of Education, School of Pharmacy, Anhui Medical University, Hefei 230032, China
- Department of Gastrointestinal Surgery, The Third Affiliated Hospital of Anhui Medical University, The First People's Hospital of Hefei, Binhu Hospital District, Hefei 230041, China
| | - Mingming Liu
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, The Key Laboratory of Antiinflammatory and Immune Medicine, Ministry of Education, School of Pharmacy, Anhui Medical University, Hefei 230032, China
| |
Collapse
|
13
|
Zhang H, Li X, Lin L. Biological Functions and Clinical Implications of CFLAR: From Cell Death Mechanisms to Therapeutic Targeting in Immune Regulation. J Inflamm Res 2025; 18:4911-4928. [PMID: 40224389 PMCID: PMC11994107 DOI: 10.2147/jir.s519885] [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: 01/28/2025] [Accepted: 04/01/2025] [Indexed: 04/15/2025] Open
Abstract
Since its initial functional characterization in the late 1990s, CASP-8 and FADD-like apoptosis regulator (CFLAR) has been recognized as a crucial regulator of both apoptosis and immune responses. CFLAR inhibits caspase-8 activation by forming heterodimers with procaspase-8 at the death-inducing signaling complex (DISC), thereby preventing its proteolytic maturation. In addition to its role in cell death, CFLAR is integral to immune regulation, modulating NF-κB-dependent cytokine production (eg, IL-1β, TNF-α) and effector functions of T cells and macrophages. Recent studies underscore the pathological significance of dysregulated CFLAR expression in a variety of diseases, including cancers and inflammatory conditions. Within the tumor microenvironment, elevated CFLAR expression confers resistance to therapy, while in infectious and inflammatory diseases, its expression levels modulate the magnitude and direction of the immune response. This review provides an in-depth exploration of CFLAR's structural and functional properties, focusing on its involvement in apoptosis, autophagy, and immune modulation. Moreover, we examine its translational potential as a therapeutic target, evidenced by ongoing preclinical studies targeting CFLAR isoforms in cancer immunotherapy. By synthesizing recent advances in CFLAR's dual roles in cell death and immune surveillance, this review highlights actionable targets for overcoming therapy resistance and immune dysregulation.
Collapse
Affiliation(s)
- Haiyang Zhang
- Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, People’s Republic of China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Sichuan University, Ministry of Education, Chengdu, People’s Republic of China
- NHC Key Laboratory of Chronobiology, Sichuan University, Chengdu, People’s Republic of China
| | - Xin Li
- Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, People’s Republic of China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Sichuan University, Ministry of Education, Chengdu, People’s Republic of China
- NHC Key Laboratory of Chronobiology, Sichuan University, Chengdu, People’s Republic of China
| | - Liangkang Lin
- Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, People’s Republic of China
- Department of Pediatrics, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen, People’s Republic of China
| |
Collapse
|
14
|
Ying YT, Yang J, Ye HW, Chen MY, Liu X, Chen W, Xu JX, Tan X. Staphylococcus aureus reprograms CASP8 (caspase 8) signaling to evade cell death and Xenophagy. Autophagy 2025:1-14. [PMID: 40143428 DOI: 10.1080/15548627.2025.2483887] [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: 11/06/2024] [Revised: 03/18/2025] [Accepted: 03/20/2025] [Indexed: 03/28/2025] Open
Abstract
Regulated cell death and xenophagy constitute fundamental cellular mechanisms against invading microorganisms. Staphylococcus aureus, a notorious pathogen, can invade and persist within host cells for extended periods. Here, we describe a novel mechanism by which S. aureus subverts these host defenses through the manipulation of the CASP8 (caspase 8) signaling pathway. Upon invasion, S. aureus triggers the assembly of a RIPK3 (receptor interacting serine/threonine kinase 3) complex to induce CASP8 autoprocessing. However, the bacterium inhibits CUL3 (cullin 3)-dependent K63-linked ubiquitination, leading to an atypical activation of CASP8. This non-canonical activation does not initiate the CASP8-CASP3 cascade but instead suppresses RIPK3-dependent necroptosis, a regulated cell death pathway typically activated when apoptosis fails. The resulting non-apoptotic, cleaved CASP8 redirects its enzymatic activity toward cleaving SQSTM1/p62, a selective macroautophagy/autophagy receptor, thus enabling S. aureus to evade antimicrobial xenophagy. The results of this study suggest that S. aureus reprograms the CASP8 signaling pathway from inducing cell death to preserving cell survival and inhibiting xenophagy, a critical strategy that supports its stealthy replication and persistence within host cells.Abbreviations: CASP3: caspase 3; CASP8: caspase 8; CFU: colony-forming units; CUL3: cullin 3; DUB: deubiquitinating enzyme; MAP1LC3B-II/LC3B-II: microtubule associated protein 1 light chain 3 beta-II; MOI: multiplicity of infection; RIPK1: receptor interacting protein kinase 1; RIPK3: receptor interacting protein kinase 3; S. aureus: Staphylococcus aureus.
Collapse
Affiliation(s)
- Yi-Tian Ying
- Department of Veterinary Medicine, Zhejiang University, Hangzhou, China
- Veterinary Medical Center, Zhejiang University, Hangzhou, China
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China
| | - Jing Yang
- Department of Veterinary Medicine, Zhejiang University, Hangzhou, China
- Veterinary Medical Center, Zhejiang University, Hangzhou, China
| | - Hui-Wen Ye
- Department of Veterinary Medicine, Zhejiang University, Hangzhou, China
- Veterinary Medical Center, Zhejiang University, Hangzhou, China
| | - Mei-Yi Chen
- Department of Veterinary Medicine, Zhejiang University, Hangzhou, China
- Veterinary Medical Center, Zhejiang University, Hangzhou, China
| | - Xia Liu
- Department of Veterinary Medicine, Zhejiang University, Hangzhou, China
- Veterinary Medical Center, Zhejiang University, Hangzhou, China
| | - Wei Chen
- Department of Veterinary Medicine, Zhejiang University, Hangzhou, China
- Veterinary Medical Center, Zhejiang University, Hangzhou, China
| | - Jin-Xin Xu
- Department of Veterinary Medicine, Zhejiang University, Hangzhou, China
- Veterinary Medical Center, Zhejiang University, Hangzhou, China
| | - Xun Tan
- Department of Veterinary Medicine, Zhejiang University, Hangzhou, China
- Veterinary Medical Center, Zhejiang University, Hangzhou, China
| |
Collapse
|
15
|
Oda H, Annibaldi A, Kastner DL, Aksentijevich I. Genetic Regulation of Cell Death: Insights from Autoinflammatory Diseases. Annu Rev Immunol 2025; 43:313-342. [PMID: 40279314 DOI: 10.1146/annurev-immunol-090222-105848] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/27/2025]
Abstract
Metazoans have evolved innate antimicrobial defenses that promote cellular survival and proliferation. Countering the inevitable molecular mechanisms by which microbes sabotage these pathways, multicellular organisms rely on an alternative, perhaps more ancient, strategy that is the immune equivalent of suicide bombing: Infection triggers cell death programs that summon localized or even systemic inflammation. The study of human genetics has now unveiled a level of complexity that refutes the naive view that cell death is merely a blunt instrument or an evolutionary afterthought. To the contrary, findings from patients with rare diseases teach us that cell death-induced inflammation is a sophisticated, tightly choreographed process. We herein review the emerging body of evidence describing a group of illnesses-inborn errors of cell death, which define many of the molecular building blocks and regulatory elements controlling cell death-induced inflammation in humans-and provide a possible road map to countering this process across the spectrum of rare and common illnesses.
Collapse
Affiliation(s)
- Hirotsugu Oda
- Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Cologne, Germany;
- Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | | | - Daniel L Kastner
- National Human Genome Research Institute (NHGRI), National Institutes of Health (NIH), Bethesda, Maryland, USA;
| | - Ivona Aksentijevich
- National Human Genome Research Institute (NHGRI), National Institutes of Health (NIH), Bethesda, Maryland, USA;
| |
Collapse
|
16
|
Kofman SB, Chu LH, Ames JM, Chavarria SD, Lichauco K, Daniels BP, Oberst A. RIPK3 coordinates RHIM domain-dependent antiviral inflammatory transcription in neurons. Sci Signal 2025; 18:eado9745. [PMID: 40168465 PMCID: PMC12042699 DOI: 10.1126/scisignal.ado9745] [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: 02/29/2024] [Accepted: 02/28/2025] [Indexed: 04/03/2025]
Abstract
Neurons are postmitotic, nonregenerative cells that have evolved fine-tuned immunological responses to maintain life-long cellular integrity, including resistance to common programmed cell death pathways such as necroptosis. We previously demonstrated a necroptosis-independent role for the key necroptotic kinase RIPK3 in host defense against neurotropic flavivirus infection. Here, we show that RIPK3 activation had distinct outcomes in primary cortical neurons when compared with mouse embryonic fibroblasts (MEFs) during Zika virus (ZIKV) infection or after sterile activation. We found that RIPK3 activation did not induce neuronal death but instead drove antiviral gene transcription after ZIKV infection. Although RIPK3 activation in MEFs induced cell death, ablation of downstream cell death effectors unveiled a RIPK3-dependent transcriptional program that largely overlapped with that observed in ZIKV-infected neurons. In death-resistant MEFs, RIPK3-dependent transcription relied on interactions with the RHIM domain-containing proteins RIPK1 and TRIF, similar to the requirements for the RIPK3-dependent antiviral transcriptional signature in ZIKV-infected neurons. These findings suggest that the pleotropic functions of RIPK3 are largely context dependent and that in cells that are resistant to cell death, RIPK3 acts as a mediator of inflammatory transcription.
Collapse
Affiliation(s)
- Sigal B. Kofman
- Department of Immunology, University of Washington, Seattle, WA 98109, USA
| | - Lan H. Chu
- Department of Immunology, University of Washington, Seattle, WA 98109, USA
| | - Joshua M. Ames
- Department of Immunology, University of Washington, Seattle, WA 98109, USA
| | | | - Katrina Lichauco
- Department of Immunology, University of Washington, Seattle, WA 98109, USA
| | - Brian P. Daniels
- Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ, 08854, USA
| | - Andrew Oberst
- Department of Immunology, University of Washington, Seattle, WA 98109, USA
| |
Collapse
|
17
|
Sawant P, Mathew A, Bensalel J, Gallego-Delgado J, Mandal P. Cancer Drug Bortezomib, a Proteasomal Inhibitor, Triggers Cytotoxicity in Microvascular Endothelial Cells via Multi-Organelle Stress. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.25.645304. [PMID: 40196627 PMCID: PMC11974872 DOI: 10.1101/2025.03.25.645304] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/09/2025]
Abstract
Proteasomes maintain cellular homeostasis by degrading abnormal proteins, while cancer cells exploit them for survival, becoming a key chemotherapeutic target. Bortezomib (BTZ), a reversible proteasomal inhibitor, is a front-line treatment for multiple myeloma, mantle cell lymphoma, and non-small cell lung cancer. However, its efficacy is limited by severe side effects, including neurotoxicity and cardiovascular distress, with its toxicity mechanisms largely unexplored. Here, we discover that Bortezomib (BTZ), is cytotoxic to non-cancerous cells distinctly from Carfilzomib (CFZ), the second-line irreversible PI. BTZ or CFZ is administered intravenously, impacting blood vessel (vascular) endothelial cells. We used human pulmonary microvascular endothelial cells (HPMECs) to demonstrate that BTZ but not CFZ elicits endoplasmic reticulum (ER) stress, mitochondrial membrane compromise, mitochondrial reactive oxygen species (ROS) accumulation, and Caspase (CASP)9 activation (mediator of Intrinsic apoptosis) within fifteen hours of treatment. By twenty-four hours, BTZ-treated cells display cleavage of CASP8 (mediator of extrinsic apoptosis), activation of CASP3 (terminal executioner of apoptosis), cell-death and vascular barrier loss. Pan-caspase inhibitor zVAD significantly rescues BTZ-treated cells from cytotoxicity. Both BTZ and CFZ effectively kill MM cells. These findings reveal novel insights into fundamental signaling of regular cells where reversible inhibition of the proteasome dictates a unique cascade of stress distinct from irreversible inhibition. These harmful effects of BTZ emphasize the need to re- evaluate its use as a frontline chemotherapy for MM. Highlights Reversible proteasomal inhibitor Bortezomib is cytotoxic to non-cancerous, microvascular endothelial cellsIn endothelial cells, Bortezomib, but not irreversible inhibitor Carfilzomib, activates temporal cascade of caspases (Caspase-9, Caspase-8, Caspase-3) triggering apoptosisCaspase activation results from ER stress (via the IRE1α-CHOP) pathway and mitochondrial stress (ROS accumulation) independently from contribution from extrinsic signal via TNFBortezomib-dependent cytotoxicity compromises endothelial barrier potential.
Collapse
|
18
|
Cai L, Fan Q, Pang R, Chen C, Zhang Y, Xie H, Huang J, Wang Y, Li P, Huang D, Jin X, Zhou Y, Li Y. Microglia programmed cell death in neurodegenerative diseases and CNS injury. Apoptosis 2025; 30:446-465. [PMID: 39656359 PMCID: PMC11799081 DOI: 10.1007/s10495-024-02041-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/03/2024] [Indexed: 02/06/2025]
Abstract
Programmed cell death (PCD) has emerged as a critical regulatory mechanism in the initiation and progression of various pathological conditions. PCD in microglia, including necroptosis, pyroptosis, apoptosis, ferroptosis, and autophagy, occurs in a variety of central nervous system (CNS) diseases. Dysregulation of microglia can lead to excessive tissue damage or neuronal death in CNS injury. Various injury stimuli trigger aberrant activation of the PCD pathway of microglia, which then further leads to inflammatory cascades that exacerbates CNS pathology in a vicious cycle. Therefore, targeting PCD in microglia is considered an important avenue for the treatment of various neurodegenerative diseases and CNS injury. In this review, we summarize the major and recent findings focusing on the mechanisms of PCD in microglia modulating functions in neurodegenerative diseases and CNS injury and provide a systematic overview of the current inhibitors targeting various PCD pathways, which may provide important therapeutic targets that merit further investigation.
Collapse
Affiliation(s)
- Ling Cai
- Department of Anesthesiology, Key Laboratory of the Ministry of Education, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Qiuyue Fan
- Department of Anesthesiology, Key Laboratory of the Ministry of Education, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Rui Pang
- Department of Anesthesiology, Key Laboratory of the Ministry of Education, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Chen Chen
- Department of Anesthesiology, Key Laboratory of the Ministry of Education, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yueman Zhang
- Department of Anesthesiology, Key Laboratory of the Ministry of Education, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Haiyi Xie
- Department of Anesthesiology, Key Laboratory of the Ministry of Education, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jingyi Huang
- Department of Anesthesiology, Key Laboratory of the Ministry of Education, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yu Wang
- Clinical Research Center, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Peiying Li
- Department of Anesthesiology, Key Laboratory of the Ministry of Education, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Clinical Research Center, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Dan Huang
- Department of Anesthesiology, Key Laboratory of the Ministry of Education, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xia Jin
- Department of Anesthesiology, Key Laboratory of the Ministry of Education, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Yuxi Zhou
- Department of Anesthesiology, Key Laboratory of the Ministry of Education, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Yan Li
- Department of Anesthesiology, Key Laboratory of the Ministry of Education, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| |
Collapse
|
19
|
Hoblos H, Cawthorne W, Samson AL, Murphy JM. Protein shapeshifting in necroptotic cell death signaling. Trends Biochem Sci 2025; 50:92-105. [PMID: 39730228 DOI: 10.1016/j.tibs.2024.11.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2024] [Revised: 11/16/2024] [Accepted: 11/22/2024] [Indexed: 12/29/2024]
Abstract
Necroptosis is a mode of programmed cell death executed by the mixed lineage kinase domain-like (MLKL) pseudokinase following its activation by the upstream receptor-interacting protein kinase-3 (RIPK3), subsequent to activation of death, Toll-like, and pathogen receptors. The pathway originates in innate immunity, although interest has surged in therapeutically targeting necroptosis owing to its dysregulation in inflammatory diseases. Here, we explore how protein conformation and higher order assembly of the pathway effectors - Z-DNA-binding protein-1 (ZBP1), RIPK1, RIPK3, and MLKL - can be modulated by post-translational modifications, such as phosphorylation, ubiquitylation, and lipidation, and intermolecular interactions to tune activities and modulate necroptotic signaling flux. As molecular level knowledge of cell death signaling grows, we anticipate targeting the conformations of key necrosomal effector proteins will emerge as new avenues for drug development.
Collapse
Affiliation(s)
- Hanadi Hoblos
- Walter and Eliza Hall Institute of Medical Research, 1G Royal Parade, Parkville, VIC 3052, Australia; Department of Medical Biology, University of Melbourne, Parkville, VIC 3052, Australia
| | - Wayne Cawthorne
- Walter and Eliza Hall Institute of Medical Research, 1G Royal Parade, Parkville, VIC 3052, Australia; Department of Medical Biology, University of Melbourne, Parkville, VIC 3052, Australia
| | - André L Samson
- Walter and Eliza Hall Institute of Medical Research, 1G Royal Parade, Parkville, VIC 3052, Australia; Department of Medical Biology, University of Melbourne, Parkville, VIC 3052, Australia
| | - James M Murphy
- Walter and Eliza Hall Institute of Medical Research, 1G Royal Parade, Parkville, VIC 3052, Australia; Department of Medical Biology, University of Melbourne, Parkville, VIC 3052, Australia; Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC 3052, Australia.
| |
Collapse
|
20
|
Fay EJ, Isterabadi K, Rezanka CM, Le J, Daugherty MD. Evolutionary and functional analyses reveal a role for the RHIM in tuning RIPK3 activity across vertebrates. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.05.09.593370. [PMID: 39149247 PMCID: PMC11326134 DOI: 10.1101/2024.05.09.593370] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 08/17/2024]
Abstract
Receptor interacting protein kinases (RIPK) RIPK1 and RIPK3 play important roles in diverse innate immune pathways. Despite this, some RIPK1/3-associated proteins are absent in specific vertebrate lineages, suggesting that some RIPK1/3 functions are conserved while others are more evolutionarily labile. Here, we perform comparative evolutionary analyses of RIPK1-5 and associated proteins in vertebrates to identify lineage-specific rapid evolution of RIPK3 and RIPK1 and recurrent loss of RIPK3-associated proteins. Despite this, diverse vertebrate RIPK3 proteins are able to activate NF-κB and cell death in human cells. Additional analyses revealed a striking conservation of the RIP homotypic interaction motif (RHIM) in RIPK3, as well as other human RHIM-containing proteins. Interestingly, diversity in the RIPK3 RHIM can tune activation of NF-κB while retaining the ability to activate cell death. Altogether, these data suggest that NF-κB activation is a core, conserved function of RIPK3, and the RHIM can tailor RIPK3 function to specific needs within and between species.
Collapse
Affiliation(s)
- Elizabeth J. Fay
- Department of Molecular Biology, School of Biological Sciences, University of California, San Diego, La Jolla, CA, 92093
| | - Kolya Isterabadi
- Department of Molecular Biology, School of Biological Sciences, University of California, San Diego, La Jolla, CA, 92093
| | - Charles M. Rezanka
- Department of Molecular Biology, School of Biological Sciences, University of California, San Diego, La Jolla, CA, 92093
| | - Jessica Le
- Department of Molecular Biology, School of Biological Sciences, University of California, San Diego, La Jolla, CA, 92093
| | - Matthew D. Daugherty
- Department of Molecular Biology, School of Biological Sciences, University of California, San Diego, La Jolla, CA, 92093
| |
Collapse
|
21
|
Pefanis A, Bongoni AK, McRae JL, Salvaris EJ, Fisicaro N, Murphy JM, Ierino FL, Cowan PJ. Inhibition of RIPK1 or RIPK3 kinase activity post ischemia-reperfusion reduces the development of chronic kidney injury. Biochem J 2025; 482:73-86. [PMID: 39705008 DOI: 10.1042/bcj20240569] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2024] [Revised: 12/19/2024] [Accepted: 12/20/2024] [Indexed: 12/21/2024]
Abstract
Ischemia-reperfusion injury (IRI) occurs when the blood supply to an organ is temporarily reduced and then restored. Kidney IRI is a form of acute kidney injury (AKI), which often progresses to kidney fibrosis. Necroptosis is a regulated necrosis pathway that has been implicated in kidney IRI. Necroptotic cell death involves the recruitment of the RIPK1 and RIPK3 kinases and the activation of the terminal effector, the mixed lineage kinase domain-like (MLKL) pseudokinase. Phosphorylated MLKL causes cell death by plasma membrane rupture, driving 'necroinflammation'. Owing to their apical role in the pathway, RIPK1 and RIPK3 have been implicated in the development of kidney fibrosis. Here, we used a mouse model of unilateral kidney IRI to assess whether the inhibition of RIPK1 or RIPK3 kinase activity reduces AKI and the progression to kidney fibrosis. Mice treated with the RIPK1 inhibitor Nec-1s, either before or after IR, showed reduced kidney injury at 24 hr compared with controls, whereas no protection was offered by the RIPK3 inhibitor GSK´872. In contrast, treatment with either inhibitor from days 3 to 9 post-IR reduced the degree of kidney fibrosis at day 28. These findings further support the role of necroptosis in IRI and provide important validation for the contribution of both RIPK1 and RIPK3 catalytic activities in the progression of kidney fibrosis. Targeting the necroptosis pathway could be a promising therapeutic strategy to mitigate kidney disease following IR.
Collapse
Affiliation(s)
- Aspasia Pefanis
- Immunology Research Centre, St Vincent's Hospital, Melbourne, Victoria, Australia
- Department of Medicine, The University of Melbourne, Melbourne, Victoria, Australia
- Department of Nephrology, St Vincent's Hospital, Melbourne, Victoria, Australia
| | - Anjan K Bongoni
- Immunology Research Centre, St Vincent's Hospital, Melbourne, Victoria, Australia
| | - Jennifer L McRae
- Immunology Research Centre, St Vincent's Hospital, Melbourne, Victoria, Australia
| | - Evelyn J Salvaris
- Immunology Research Centre, St Vincent's Hospital, Melbourne, Victoria, Australia
| | - Nella Fisicaro
- Immunology Research Centre, St Vincent's Hospital, Melbourne, Victoria, Australia
| | - James M Murphy
- Walter and Eliza Hall Institute of Medical Research, Parkville, Melbourne, Australia
- Department of Medical Biology, The University of Melbourne, Melbourne, Victoria, Australia
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia
| | - Francesco L Ierino
- Department of Medicine, The University of Melbourne, Melbourne, Victoria, Australia
- Department of Nephrology, St Vincent's Hospital, Melbourne, Victoria, Australia
| | - Peter J Cowan
- Immunology Research Centre, St Vincent's Hospital, Melbourne, Victoria, Australia
- Department of Medicine, The University of Melbourne, Melbourne, Victoria, Australia
| |
Collapse
|
22
|
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
|
23
|
Park SE, Kwon SJ, Kim SJ, Jeong JB, Kim MJ, Choi SJ, Oh SY, Ryu GH, Jeon HB, Chang JW. Anti-necroptotic effects of human Wharton's jelly-derived mesenchymal stem cells in skeletal muscle cell death model via secretion of GRO-α. PLoS One 2024; 19:e0313693. [PMID: 39621655 PMCID: PMC11611217 DOI: 10.1371/journal.pone.0313693] [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: 04/04/2024] [Accepted: 10/29/2024] [Indexed: 01/06/2025] Open
Abstract
Human mesenchymal stem cells (hMSCs) have therapeutic applications and potential for use in regenerative medicine. However, the use of hMSCs in research and clinical medicine is limited by a lack of information pertaining to their donor-specific functional attributes. In this study, we compared the characteristics of same-donor derived placenta (PL) and Wharton's jelly (WJ)-derived hMSCs, we also compared their mechanism of action in a skeletal muscle disease in vitro model. The same-donor-derived hWJ- and hPL-MSCs exhibited typical hMSC characteristics. However, GRO-α was differentially expressed in hWJ- and hPL-MSCs. hWJ-MSCs, which secreted a high amount of GRO-α, displayed a higher ability to inhibit necroptosis in skeletal muscle cells than hPL-MSCs. This demonstrates the anti-necroptotic therapeutic effect of GRO-α in the skeletal muscle cell death model. Furthermore, GRO-α also exhibited the anti-necroptotic effect in a Duchenne muscular dystrophy (DMD) mouse model. Considering their potential to inhibit necroptosis in skeletal muscle cells, hWJ-MSCs and the derived GRO-α are novel treatment options for skeletal muscle diseases such as DMD.
Collapse
Affiliation(s)
- Sang Eon Park
- Cell and Gene Therapy Institute, ENCell Co. Ltd, Seoul, Republic of Korea
- Cell and Gene Therapy Institute, Samsung Medical Center, Seoul, Republic of Korea
| | - Soo Jin Kwon
- Cell and Gene Therapy Institute, ENCell Co. Ltd, Seoul, Republic of Korea
- Cell and Gene Therapy Institute, Samsung Medical Center, Seoul, Republic of Korea
| | - Sun Jeong Kim
- Cell and Gene Therapy Institute, ENCell Co. Ltd, Seoul, Republic of Korea
- Cell and Gene Therapy Institute, Samsung Medical Center, Seoul, Republic of Korea
| | - Jang Bin Jeong
- Cell and Gene Therapy Institute, ENCell Co. Ltd, Seoul, Republic of Korea
- Cell and Gene Therapy Institute, Samsung Medical Center, Seoul, Republic of Korea
| | - Min-Jeong Kim
- Cell and Gene Therapy Institute, Samsung Medical Center, Seoul, Republic of Korea
| | - Suk-joo Choi
- Department of Obstetrics and Gynecology, Samsung Medical Center, Seoul, Republic of Korea
| | - Soo-young Oh
- Department of Obstetrics and Gynecology, Samsung Medical Center, Seoul, Republic of Korea
| | - Gyu Ha Ryu
- Department of Medical Device Management and Research, Samsung Advanced Institute for Health Sciences & Technology, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
- The Office of R&D Strategy & Planning, Samsung Medical Center, Seoul, Republic of Korea
| | - Hong Bae Jeon
- Cell and Gene Therapy Institute, ENCell Co. Ltd, Seoul, Republic of Korea
| | - Jong Wook Chang
- Cell and Gene Therapy Institute, ENCell Co. Ltd, Seoul, Republic of Korea
- Cell and Gene Therapy Institute, Samsung Medical Center, Seoul, Republic of Korea
- Department of Health Sciences and Technology, Samsung Advanced Institute for Health Sciences & Technology, Sungkyunkwan University, Seoul, Korea
| |
Collapse
|
24
|
Pang BPS, Iu ECY, Hang M, Chan WS, Tse MCL, Yeung CTY, Wang M, Siu PMF, Lee CW, Ye K, So H, Chan CB. Deficiency of muscle-generated brain-derived neurotrophic factor causes inflammatory myopathy through reactive oxygen species-mediated necroptosis and pyroptosis. Redox Biol 2024; 78:103418. [PMID: 39531828 PMCID: PMC11602578 DOI: 10.1016/j.redox.2024.103418] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2024] [Revised: 10/21/2024] [Accepted: 11/02/2024] [Indexed: 11/16/2024] Open
Abstract
Idiopathic inflammatory myopathy (commonly known as myositis) is a group of immune-related diseases characterized by muscle damage, weakness, and fatigue with unknown causes. Although overactivated innate immunity is a widely believed cause of myositis onset, the mechanism that provokes and maintains a high immune response in myositis patients is still unclear. This study aims to test if brain-derived neurotrophic factor (BDNF) deficiency per se is sufficient to cause myositis and determine its underlying mechanism. We found that ablating BDNF production in skeletal muscle is sufficient to trigger myositis development in mice. Muscle-specific Bdnf knockout (MBKO) mice displayed extensive myocyte necrosis, mononuclear cell infiltration, and myophagocytosis. In association with these damages, elevated production of pro-inflammatory cytokines such as interleukin (IL) 23, IL-1β, IL-18, and tumor necrosis factor α (TNFα) was found in the muscle of MBKO mice. Disruption of sarcolemma integrity was also detected in MBKO mice, which is a result of necroptosis executioner Mixed lineage kinase domain-like protein (MLKL) and pyroptosis executioner Gasdermin D (GSDMD) activation. Mechanistically, diminishing BDNF synthesis in myotubes enhances the accumulation of mitochondrial reactive oxygen species (mtROS), which sensitizes the cells towards TNFα-induced receptor-interacting protein kinase (RIPs) activation and promotes the formation of NLR family pyrin domain containing 3 (NLRP3)-containing inflammasome. BDNF deficiency-induced cell death could be alleviated by scavenging mtROS, suppressing the activity of GSDMD, or inhibiting receptor-interacting kinase 3 (RIP3). Similarly, supplementation of BDNF mimetics, suppression of RIP3 activity, increasing the intramyocellular antioxidant, or enhancing mitophagy ameliorated the myopathies of MBKO mice and improved their muscle strength. Together, our study demonstrates that insufficient BDNF production in mouse muscle causes the development of pathological features of myositis via enhancing oxidative stress, necroptosis, and pyroptosis in myofibers.
Collapse
Affiliation(s)
- Brian Pak Shing Pang
- School of Biological Sciences, Faculty of Science, The University of Hong Kong, Hong Kong Special Administrative Region
| | - Elsie Chit Yu Iu
- School of Biological Sciences, Faculty of Science, The University of Hong Kong, Hong Kong Special Administrative Region
| | - Miaojia Hang
- School of Biological Sciences, Faculty of Science, The University of Hong Kong, Hong Kong Special Administrative Region
| | - Wing Suen Chan
- School of Biological Sciences, Faculty of Science, The University of Hong Kong, Hong Kong Special Administrative Region
| | - Margaret Chui Ling Tse
- School of Biological Sciences, Faculty of Science, The University of Hong Kong, Hong Kong Special Administrative Region
| | - Connie Tsz Ying Yeung
- School of Biological Sciences, Faculty of Science, The University of Hong Kong, Hong Kong Special Administrative Region
| | - Mingfu Wang
- Shenzhen Key Laboratory of Food Nutrition and Health, Institute for Advanced Study, Shenzhen University, Shenzhen, 518060, Hong Kong Special Administrative Region
| | - Parco Ming Fai Siu
- Division of Kinesiology, School of Public Health, The University of Hong Kong, Pok Fu Lam, Hong Kong Special Administrative Region
| | - Chi Wai Lee
- Department of Biology, Hong Kong Baptist University, Hong Kong Special Administrative Region
| | - Keqiang Ye
- Faculty of Life and Health Sciences, and Brain Cognition and Brain Disease Institute (BCBDI), Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Hong Kong Special Administrative Region
| | - Ho So
- Department of Medicine & Therapeutics, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong Special Administrative Region
| | - Chi Bun Chan
- School of Biological Sciences, Faculty of Science, The University of Hong Kong, Hong Kong Special Administrative Region; State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, Hong Kong Special Administrative Region.
| |
Collapse
|
25
|
Ge Q, Zhang T, Yu J, Lu X, Xiao S, Zhang T, Qing T, Xiao Z, Zeng L, Luo L. A new perspective on targeting pulmonary arterial hypertension: Programmed cell death pathways (Autophagy, Pyroptosis, Ferroptosis). Biomed Pharmacother 2024; 181:117706. [PMID: 39581144 DOI: 10.1016/j.biopha.2024.117706] [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: 05/05/2024] [Revised: 11/10/2024] [Accepted: 11/19/2024] [Indexed: 11/26/2024] Open
Abstract
Pulmonary arterial hypertension (PAH) is a severe cardiovascular disease characterized by elevated pulmonary vascular resistance, progressive increases in pulmonary artery pressures, ultimately leading to right-sided heart failure, and potentially mortality. Pulmonary vascular remodeling is pivotal in PAH onset and progression. While targeted drug therapies have notably ameliorated PAH prognosis, current medications primarily focus on vascular vasodilation, with limited ability to reverse pulmonary vascular remodeling fundamentally, resulting in suboptimal patient prognoses. Cellular death in pulmonary vasculature, once thought to be confined to apoptosis and necrosis, has evolved with the identification of pyroptosis, autophagy, and ferroptosis, revealing their association with vascular injury in PAH. These novel forms of regulated cellular death impact reactive oxygen species (ROS) generation, calcium stress, and inflammatory cascades, leading to pulmonary vascular cell loss, exacerbating vascular injury, and mediating adverse remodeling, inflammation, immune anomalies, and current emerging mechanisms (such as endothelial-mesenchymal transition, abnormal energy metabolism, and epigenetic regulation) in the pathogenesis of PAH. This review comprehensively delineates the roles of autophagy, pyroptosis, and ferroptosis in PAH, elucidating recent advances in their involvement and regulation of vascular injury. It juxtaposes their distinct functions in PAH and discusses the interplay of these programmed cell deaths in pulmonary vascular injury, highlighting the benefits of combined targeted therapies in mitigating pulmonary arterial hypertension-induced vascular injury, providing novel insights into targeted treatments for pulmonary arterial hypertension.
Collapse
Affiliation(s)
- Qingliang Ge
- Department of Cardiology, Changde Hospital, Xiangya School of Medicine, Central South University (The first people's hospital of Changde city), Changde City, China
| | - Tianqing Zhang
- Department of Cardiology, Changde Hospital, Xiangya School of Medicine, Central South University (The first people's hospital of Changde city), Changde City, China
| | - Jiangbiao Yu
- Department of Cardiology, Changde Hospital, Xiangya School of Medicine, Central South University (The first people's hospital of Changde city), Changde City, China
| | - Xuelin Lu
- Department of Cardiology, Changde Hospital, Xiangya School of Medicine, Central South University (The first people's hospital of Changde city), Changde City, China
| | - Sijie Xiao
- Department of Ultrasound, Changde Hospital, Xiangya School of Medicine, Central South University (The first people's hospital of Changde city), Changde City, China
| | - Ting Zhang
- Department of Cardiology, Changde Hospital, Xiangya School of Medicine, Central South University (The first people's hospital of Changde city), Changde City, China
| | - Tao Qing
- Department of Cardiology, Changde Hospital, Xiangya School of Medicine, Central South University (The first people's hospital of Changde city), Changde City, China
| | - Zhenni Xiao
- Department of Cardiology, Changde Hospital, Xiangya School of Medicine, Central South University (The first people's hospital of Changde city), Changde City, China
| | - Liuting Zeng
- Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Nanjing, China
| | - Li Luo
- Department of Cardiology, Changde Hospital, Xiangya School of Medicine, Central South University (The first people's hospital of Changde city), Changde City, China.
| |
Collapse
|
26
|
Xu N, Du Q, Cheng Y, Nie L, Ma P, Feng D, Huang Y, Tong D. Porcine parvovirus infection induces necroptosis of porcine placental trophoblast cells via a ZBP1-mediated pathway. Vet Res 2024; 55:156. [PMID: 39614405 DOI: 10.1186/s13567-024-01410-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Accepted: 09/06/2024] [Indexed: 12/01/2024] Open
Abstract
Porcine parvovirus (PPV) infection induces germ cell death, leading to reproductive disorders in first-pregnant sows. Porcine placental trophoblast cells (PTCs) are the major target of PPV, and we have previously found that PPV infection leads to the death of PTCs by a non-apoptotic process, which may be related to PPV pathogenicity. The Z-nucleic acid-binding protein 1 (ZBP1) is often activated after virus invasion and mediates subsequent cell death. Here, we found that PPV infection induced ZBP1-mediated necroptosis of porcine PTCs in the presence of the apoptosis inhibitor, AC-DEVD-CHO. ZBP1 expression was upregulated during PPV infection, and ZBP1 knockout or RNA interference significantly reduced its expression along with the PPV-induced necroptosis. We discovered that RIPK3 and MLKL, but not Caspase-8, were involved in downstream signaling of ZBP1 during PPV infection; the phosphorylation levels of RIPK3 and MLKL were enhanced, but Caspase-8 was not significantly cleaved. The knockout of RIPK3 and MLKL significantly reduced the PPV infection-induced necroptosis of porcine PTCs. RIPK3 knockout did not affect the PPV infection-induced upregulation of ZBP1 expression, but blocked the activation of MLKL. MLKL knockout did not affect the upregulation of ZBP1 expression and RIPK3 phosphorylation during PPV infection. UV-inactivated PPV induced significantly less necroptosis of porcine PTCs than non-irradiated PPV. A PPV strain with a mutation in the translation initiation codon was still able to induce necroptosis of PTCs through the ZBP1/RIPK3/MLKL pathway. These results provide new insights into the pathogenic mechanism of PPV infection and suggest that PPV infection of porcine PTCs induces necroptosis through the viral DNA-dependent ZBP1/RIPK3/MLKL pathway.
Collapse
Affiliation(s)
- Ning Xu
- College of Veterinary Medicine, Northwest A&F University, Yangling, China
| | - Qian Du
- College of Veterinary Medicine, Northwest A&F University, Yangling, China
- Engineering Research Center of Efficient New Vaccines for Animals, Ministry of Education, Yangling, China
- Key Laboratory of Ruminant Disease Prevention and Control (West), Ministry of Agriculture and Rural Affairs, Yangling, China
- Engineering Research Center of Efficient New Vaccines for Animals, Universities of Shaanxi Province, Yangling, China
| | - Yijiao Cheng
- College of Veterinary Medicine, Northwest A&F University, Yangling, China
| | - Lichen Nie
- College of Veterinary Medicine, Northwest A&F University, Yangling, China
| | - Peipei Ma
- College of Veterinary Medicine, Northwest A&F University, Yangling, China
| | - Dingwen Feng
- College of Veterinary Medicine, Northwest A&F University, Yangling, China
| | - Yong Huang
- College of Veterinary Medicine, Northwest A&F University, Yangling, China
- Engineering Research Center of Efficient New Vaccines for Animals, Ministry of Education, Yangling, China
- Key Laboratory of Ruminant Disease Prevention and Control (West), Ministry of Agriculture and Rural Affairs, Yangling, China
- Engineering Research Center of Efficient New Vaccines for Animals, Universities of Shaanxi Province, Yangling, China
| | - Dewen Tong
- College of Veterinary Medicine, Northwest A&F University, Yangling, China.
- Engineering Research Center of Efficient New Vaccines for Animals, Ministry of Education, Yangling, China.
- Key Laboratory of Ruminant Disease Prevention and Control (West), Ministry of Agriculture and Rural Affairs, Yangling, China.
- Engineering Research Center of Efficient New Vaccines for Animals, Universities of Shaanxi Province, Yangling, China.
| |
Collapse
|
27
|
Mocarski ES. Cytomegalovirus Biology Viewed Through a Cell Death Suppression Lens. Viruses 2024; 16:1820. [PMID: 39772130 PMCID: PMC11680106 DOI: 10.3390/v16121820] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2024] [Revised: 11/22/2024] [Accepted: 11/22/2024] [Indexed: 01/11/2025] Open
Abstract
Cytomegaloviruses, species-specific members of the betaherpesviruses, encode an impressive array of immune evasion strategies committed to the manipulation of the host immune system enabling these viruses to remain for life in a stand-off with host innate and adaptive immune mechanisms. Even though they are species-restricted, cytomegaloviruses are distributed across a wide range of different mammalian species in which they cause systemic infection involving many different cell types. Regulated, or programmed cell death has a recognized potential to eliminate infected cells prior to completion of viral replication and release of progeny. Cell death also naturally terminates replication during the final stages of replication. Over the past two decades, the host defense potential of known programmed cell death pathways (apoptosis, necroptosis, and pyroptosis), as well as a novel mitochondrial serine protease pathway have been defined through studies of cytomegalovirus-encoded cell death suppressors. Such virus-encoded inhibitors prevent virus-induced, cytokine-induced, and stress-induced death of infected cells while also moderating inflammation. By evading cell death and consequent inflammation as well as innate and adaptive immune clearance, cytomegaloviruses represent successful pathogens that become a critical disease threat when the host immune system is compromised. This review will discuss cell death programs acquired for mammalian host defense against cytomegaloviruses and enumerate the range of modulatory strategies this type of virus employs to balance host defense in favor of lifelong persistence.
Collapse
Affiliation(s)
- Edward S. Mocarski
- Department of Microbiology & Immunology, Stanford Medical School, Stanford University, Stanford, CA 94305, USA;
- Department of Microbiology & Immunology, Emory Medical School, Emory Vaccine Center, Emory University, Atlanta, GA 30322, USA
| |
Collapse
|
28
|
Li B, Ling Z, Wang Y, Xing Y. Receptor-Interacting Protein Kinase 3 Augments Neuroinflammation by Facilitating Neutrophil Infiltration during an Ischemic Stroke. J Vasc Res 2024; 62:51-62. [PMID: 39571563 DOI: 10.1159/000542571] [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: 02/03/2024] [Accepted: 10/22/2024] [Indexed: 01/11/2025] Open
Abstract
INTRODUCTION Neutrophil infiltration is responsible for the neuroinflammation during an ischemic stroke. Here, we explored the role of receptor-interacting protein kinase 3 (RIP3) in neutrophil infiltration during an ischemic stroke. METHODS The rat middle cerebral artery occlusion (MCAO) model was utilized to identify pivotal proteins involved in neutrophil infiltration during an ischemic stroke. Neutrophils were isolated from the peripheral blood of mice, and a co-immunoprecipitation (co-IP) assay was performed to identify the proteins that interact with RIP3. RESULTS The rat MCAO model was successfully established. Myeloperoxidase (MPO) was significantly upregulated in the MCAO group, indicating the presence of neutrophil infiltration. RIP3 protein level exhibited a similar trend to MPO protein level, suggesting that neuroinflammation might be partly activated by RIP3 through the promotion of neutrophil infiltration. Co-IP and mass spectrometry analyses suggested that RIP3 facilitated neutrophil infiltration partly by affecting protein kinases (Rock1 and Prkaca) downstream of RIP3, and the interaction between RIP3 and Rock1 or Prkaca was validated by IF and co-IP assays. CONCLUSION In this study, it was observed that RIP3 affects neutrophil infiltration, a critical phenomenon associated with neuronal injury during ischemic stroke, partly by the modulation of downstream proteins such as Rock1 and Prkaca.
Collapse
Affiliation(s)
- Baiyu Li
- Department of Neurology Cadre Ward, Gansu Provincial Hospital, Lanzhou, China
| | - Zexia Ling
- Department of Gastroenterology Cadre Ward, Gansu Provincial Hospital, Lanzhou, China
| | - Yanyan Wang
- Department of Neurology Cadre Ward, Gansu Provincial Hospital, Lanzhou, China
| | - Yinhua Xing
- Department of Gastroenterology Cadre Ward, Gansu Provincial Hospital, Lanzhou, China
| |
Collapse
|
29
|
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
|
30
|
Park JS, Leem YH, Kim DY, Park JM, Kim SE, Kim HS. Neuroprotective and anti-inflammatory effects of the RIPK3 inhibitor GSK872 in an MPTP-induced mouse model of Parkinson's disease. Neurochem Int 2024; 181:105896. [PMID: 39491747 DOI: 10.1016/j.neuint.2024.105896] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Revised: 10/28/2024] [Accepted: 10/31/2024] [Indexed: 11/05/2024]
Abstract
Parkinson's disease (PD) is a neurodegenerative disorder triggered by the loss of dopaminergic neurons in the substantia nigra (SN). Recent studies have demonstrated that necroptosis is involved in dopaminergic neuronal cell death and the resulting neuroinflammation. During the process of necroptosis, a necrosome complex is formed consisting of the proteins receptor-interacting protein kinase 1 (RIPK1), RIPK3, and mixed lineage kinase domain-like protein (MLKL). Although the neuroprotective effects of the RIPK1-specific inhibitor necrostatin-1, as well as RIPK3 and MLKL knockout in mice, have been described, the effects of RIPK3 pharmacological inhibitors have not yet been reported in animal models of PD. In the present study, we investigated the neuroprotective effects of GSK872, a specific RIPK3 inhibitor, in a 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-induced mouse model of PD. GSK872 rescued MPTP-induced motor impairment and inhibited tyrosine hydroxylase-positive dopaminergic cell death in the SN and striatum. Additionally, GSK872 inhibited the MPTP-induced increase in the expression of p-RIPK3 and p-MLKL in both the dopaminergic neurons and microglia, as assessed by biochemical and histological analyses. GSK872 further inhibited microglial activation and the expression of inflammatory mediators including NLRP3, interleukin (IL)-1β, IL-6, tumor necrosis factor-alpha, and inducible nitric oxide synthase in the SN region of MPTP mice. Using in vitro experiments, we validated the effects of GSK872 on necroptosis in SH-SY5Y neuronal and BV2 microglial cells. Overall, our results suggest that GSK872 exerts neuroprotective and anti-inflammatory effects, and may thus have therapeutic potential for PD.
Collapse
Affiliation(s)
- Jin-Sun Park
- Department of Molecular Medicine, Inflammation-Cancer Microenvironment Research Center, School of Medicine, Ewha Womans University, Seoul, South Korea
| | - Yea-Hyun Leem
- Department of Molecular Medicine, Inflammation-Cancer Microenvironment Research Center, School of Medicine, Ewha Womans University, Seoul, South Korea
| | - Do-Yeon Kim
- Department of Molecular Medicine, Inflammation-Cancer Microenvironment Research Center, School of Medicine, Ewha Womans University, Seoul, South Korea
| | - Jae-Min Park
- Department of Molecular Medicine, Inflammation-Cancer Microenvironment Research Center, School of Medicine, Ewha Womans University, Seoul, South Korea
| | - Seong-Eun Kim
- Department of Molecular Medicine, Inflammation-Cancer Microenvironment Research Center, School of Medicine, Ewha Womans University, Seoul, South Korea
| | - Hee-Sun Kim
- Department of Molecular Medicine, Inflammation-Cancer Microenvironment Research Center, School of Medicine, Ewha Womans University, Seoul, South Korea; Department of Brain & Cognitive Sciences, Ewha Womans University, Seoul, South Korea.
| |
Collapse
|
31
|
Davies KA, Czabotar PE, Murphy JM. Death at a funeral: Activation of the dead enzyme, MLKL, to kill cells by necroptosis. Curr Opin Struct Biol 2024; 88:102891. [PMID: 39059047 DOI: 10.1016/j.sbi.2024.102891] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Revised: 06/25/2024] [Accepted: 07/05/2024] [Indexed: 07/28/2024]
Abstract
Necroptosis is a lytic form of programmed cell death implicated in inflammatory pathologies, leading to intense interest in the underlying mechanisms and therapeutic prospects. Here, we review our current structural understanding of how the terminal executioner of the pathway, the dead kinase, mixed lineage kinase domain-like (MLKL), is converted from a dormant to killer form by the upstream regulatory kinase, RIPK3. RIPK3-mediated phosphorylation of MLKL's pseudokinase domain toggles a molecular switch that induces dissociation from a cytoplasmic platform, assembly of MLKL oligomers, and trafficking to the plasma membrane, where activated MLKL accumulates and permeabilises the lipid bilayer to induce cell death. We highlight gaps in mechanistic knowledge of MLKL's activation, how mechanisms diverge between species, and the power of modelling in advancing structural insights.
Collapse
Affiliation(s)
- Katherine A Davies
- Walter and Eliza Hall Institute of Medical Research, 1G Royal Parade, Parkville, VIC 3052, Australia; Department of Medical Biology, University of Melbourne, Parkville, VIC 3052, Australia.
| | - Peter E Czabotar
- Walter and Eliza Hall Institute of Medical Research, 1G Royal Parade, Parkville, VIC 3052, Australia; Department of Medical Biology, University of Melbourne, Parkville, VIC 3052, Australia.
| | - James M Murphy
- Walter and Eliza Hall Institute of Medical Research, 1G Royal Parade, Parkville, VIC 3052, Australia; Department of Medical Biology, University of Melbourne, Parkville, VIC 3052, Australia; Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC 3052, Australia.
| |
Collapse
|
32
|
Wen W, Hu X, Liu J, Zeng F, Xu Y, Yuan Y, Gao C, Sun X, Cheng B, Wang J, Hu X, Xiao RP, Chen X, Zhang X. RIP3 regulates doxorubicin-induced intestinal mucositis via FUT2-mediated α-1,2-fucosylation. Inflamm Res 2024; 73:1781-1801. [PMID: 39180691 DOI: 10.1007/s00011-024-01932-2] [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: 06/03/2024] [Revised: 07/24/2024] [Accepted: 08/05/2024] [Indexed: 08/26/2024] Open
Abstract
OBJECTIVE Intestinal mucositis is one of the common side effects of anti-cancer chemotherapy. However, the molecular mechanisms involved in mucositis development remain incompletely understood. In this study, we investigated the function of receptor-interacting protein kinase 3 (RIP3/RIPK3) in regulating doxorubicin-induced intestinal mucositis and its potential mechanisms. METHODS Intestinal mucositis animal models were induced in mice for in vivo studies. Rat intestinal cell line IEC-6 was used for in vitro studies. RNA‑seq was used to explore the transcriptomic changes in doxorubicin-induced intestinal mucositis. Intact glycopeptide characterization using mass spectrometry was applied to identify α-1,2-fucosylated proteins associated with mucositis. RESULTS Doxorubicin treatment increased RIP3 expression in the intestine and caused severe intestinal mucositis in the mice, depletion of RIP3 abolished doxorubicin-induced intestinal mucositis. RIP3-mediated doxorubicin-induced mucositis did not depend on mixed lineage kinase domain-like (MLKL) but on α-1,2-fucosyltransferase 2 (FUT2)-catalyzed α-1,2-fucosylation on inflammation-related proteins. Deficiency of MLKL did not affect intestinal mucositis, whereas inhibition of α-1,2-fucosylation by 2-deoxy-D-galactose (2dGal) profoundly attenuated doxorubicin-induced inflammation and mucositis. CONCLUSIONS RIP3-FUT2 pathway is a central node in doxorubicin-induced intestinal mucositis. Targeting intestinal RIP3 and/or FUT2-mediated α-1,2-fucosylation may provide potential targets for preventing chemotherapy-induced intestinal mucositis.
Collapse
Affiliation(s)
- Wei Wen
- Institute of Molecular Medicine, College of Future Technology, Peking University, Beijing, 100871, China
- Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Peking University, Beijing, 100871, China
- PKU-Nanjing Institute of Translational Medicine, Nanjing, 211800, China
| | - Xiaomin Hu
- Institute of Molecular Medicine, College of Future Technology, Peking University, Beijing, 100871, China
- Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Peking University, Beijing, 100871, China
- State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100730, China
| | - Jialin Liu
- College of Chemistry and Molecular Engineering, Peking University, Beijing, 100871, China
- Beijing National Laboratory for Molecular Sciences, Peking University, Beijing, 100871, China
| | - Fanxin Zeng
- Institute of Molecular Medicine, College of Future Technology, Peking University, Beijing, 100871, China
- Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Peking University, Beijing, 100871, China
- Department of Clinical Research Center, Dazhou Central Hospital, Dazhou, 635000, China
| | - Yihua Xu
- Institute of Molecular Medicine, College of Future Technology, Peking University, Beijing, 100871, China
- Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Peking University, Beijing, 100871, China
| | - Ye Yuan
- Institute of Molecular Medicine, College of Future Technology, Peking University, Beijing, 100871, China
- Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Peking University, Beijing, 100871, China
| | - Chunyan Gao
- Institute of Molecular Medicine, College of Future Technology, Peking University, Beijing, 100871, China
- Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Peking University, Beijing, 100871, China
| | - Xueting Sun
- Institute of Molecular Medicine, College of Future Technology, Peking University, Beijing, 100871, China
- Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Peking University, Beijing, 100871, China
| | - Bo Cheng
- College of Chemistry and Molecular Engineering, Peking University, Beijing, 100871, China
- Beijing National Laboratory for Molecular Sciences, Peking University, Beijing, 100871, China
| | - Jue Wang
- Institute of Molecular Medicine, College of Future Technology, Peking University, Beijing, 100871, China
- Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Peking University, Beijing, 100871, China
| | - Xinli Hu
- Institute of Molecular Medicine, College of Future Technology, Peking University, Beijing, 100871, China
- Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Peking University, Beijing, 100871, China
| | - Rui-Ping Xiao
- Institute of Molecular Medicine, College of Future Technology, Peking University, Beijing, 100871, China.
- Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Peking University, Beijing, 100871, China.
- PKU-Nanjing Institute of Translational Medicine, Nanjing, 211800, China.
- State Key Laboratory of Biomembrane and Membrane Biotechnology, Peking University, Beijing, 100871, China.
- Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, 100871, China.
| | - Xing Chen
- College of Chemistry and Molecular Engineering, Peking University, Beijing, 100871, China.
- Beijing National Laboratory for Molecular Sciences, Peking University, Beijing, 100871, China.
- Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, 100871, China.
- Synthetic and Functional Biomolecules Center, Peking University, Beijing, 100871, China.
- Key Laboratory of Bioorganic Chemistry and Molecular Engineering of Ministry of Education, Peking University, Beijing, 100871, China.
| | - Xiuqin Zhang
- Institute of Molecular Medicine, College of Future Technology, Peking University, Beijing, 100871, China.
- Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Peking University, Beijing, 100871, China.
- PKU-Nanjing Institute of Translational Medicine, Nanjing, 211800, China.
- National Biomedical Imaging Center, School of Future Technology, Peking University, Beijing, 100871, China.
| |
Collapse
|
33
|
Khaleque MA, Kim JH, Tanvir MAH, Park JB, Kim YY. Significance of Necroptosis in Cartilage Degeneration. Biomolecules 2024; 14:1192. [PMID: 39334958 PMCID: PMC11429838 DOI: 10.3390/biom14091192] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Revised: 09/09/2024] [Accepted: 09/18/2024] [Indexed: 09/30/2024] Open
Abstract
Cartilage, a critical tissue for joint function, often degenerates due to osteoarthritis (OA), rheumatoid arthritis (RA), and trauma. Recent research underscores necroptosis, a regulated form of necrosis, as a key player in cartilage degradation. Unlike apoptosis, necroptosis triggers robust inflammatory responses, exacerbating tissue damage. Key mediators such as receptor-interacting serine/threonine-protein kinase-1 (RIPK1), receptor-interacting serine/threonine-protein kinase-3(RIPK3), and mixed lineage kinase domain-like (MLKL) are pivotal in this process. Studies reveal necroptosis contributes significantly to OA and RA pathophysiology, where elevated RIPK3 and associated proteins drive cartilage degradation. Targeting necroptotic pathways shows promise; inhibitors like Necrostatin-1 (Nec-1), GSK'872, and Necrosulfonamide (NSA) reduce necroptotic cell death, offering potential therapeutic avenues. Additionally, autophagy's role in mitigating necroptosis-induced damage highlights the need for comprehensive strategies addressing multiple pathways. Despite these insights, further research is essential to fully understand necroptosis' mechanisms and develop effective treatments. This review synthesizes current knowledge on necroptosis in cartilage degeneration, aiming to inform novel therapeutic approaches for OA, RA, and trauma.
Collapse
Affiliation(s)
- Md Abdul Khaleque
- Department of Orthopedic Surgery, Daejeon St. Mary's Hospital, The Catholic University of Korea, Seoul 06591, Republic of Korea
| | - Jea-Hoon Kim
- Department of Orthopedic Surgery, Daejeon St. Mary's Hospital, The Catholic University of Korea, Seoul 06591, Republic of Korea
| | - Md Amit Hasan Tanvir
- Department of Orthopedic Surgery, Daejeon St. Mary's Hospital, The Catholic University of Korea, Seoul 06591, Republic of Korea
| | - Jong-Beom Park
- Department of Orthopedic Surgery, Uijeongbu Saint Mary's Hospital, The Catholic University of Korea, Seoul 06591, Republic of Korea
| | - Young-Yul Kim
- Department of Orthopedic Surgery, Daejeon St. Mary's Hospital, The Catholic University of Korea, Seoul 06591, Republic of Korea
| |
Collapse
|
34
|
Ran R, Zhang SB, Shi YQ, Dong H, Song W, Dong YB, Zhou KS, Zhang HH. Spotlight on necroptosis: Role in pathogenesis and therapeutic potential of intervertebral disc degeneration. Int Immunopharmacol 2024; 138:112616. [PMID: 38959544 DOI: 10.1016/j.intimp.2024.112616] [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: 05/16/2024] [Revised: 06/19/2024] [Accepted: 06/30/2024] [Indexed: 07/05/2024]
Abstract
Intervertebral disc degeneration (IDD) is the leading cause of low back pain, which is one of the major factors leading to disability and severe economic burden. Necroptosis is an important form of programmed cell death (PCD), a highly regulated caspase-independent type of cell death that is regulated by receptor-interacting protein kinase 1 (RIPK1), RIPK3 and mixed lineage kinase domain-like protein (MLKL)-mediated, play a key role in the pathophysiology of various inflammatory, infectious and degenerative diseases. Recent studies have shown that necroptosis plays an important role in the occurrence and development of IDD. In this review, we provide an overview of the initiation and execution of necroptosis and explore in depth its potential mechanisms of action in IDD. The analysis focuses on the connection between NP cell necroptosis and mitochondrial dysfunction-oxidative stress pathway, inflammation, endoplasmic reticulum stress, apoptosis, and autophagy. Finally, we evaluated the possibility of treating IDD by inhibiting necroptosis, and believed that targeting necroptosis may be a new strategy to alleviate the symptoms of IDD.
Collapse
Affiliation(s)
- Rui Ran
- Lanzhou University Second Hospital, 82 Cuiying Men, Lanzhou 730000, PR China; Orthopedics Key Laboratory of Gansu Province, Lanzhou 730000, PR China
| | - Shun-Bai Zhang
- Lanzhou University Second Hospital, 82 Cuiying Men, Lanzhou 730000, PR China; Orthopedics Key Laboratory of Gansu Province, Lanzhou 730000, PR China
| | - Yong-Qiang Shi
- Lanzhou University Second Hospital, 82 Cuiying Men, Lanzhou 730000, PR China; Orthopedics Key Laboratory of Gansu Province, Lanzhou 730000, PR China
| | - Hao Dong
- Lanzhou University Second Hospital, 82 Cuiying Men, Lanzhou 730000, PR China; Orthopedics Key Laboratory of Gansu Province, Lanzhou 730000, PR China
| | - Wei Song
- Lanzhou University Second Hospital, 82 Cuiying Men, Lanzhou 730000, PR China; Orthopedics Key Laboratory of Gansu Province, Lanzhou 730000, PR China
| | - Yan-Bo Dong
- Lanzhou University Second Hospital, 82 Cuiying Men, Lanzhou 730000, PR China; Orthopedics Key Laboratory of Gansu Province, Lanzhou 730000, PR China
| | - Kai-Sheng Zhou
- Lanzhou University Second Hospital, 82 Cuiying Men, Lanzhou 730000, PR China; Orthopedics Key Laboratory of Gansu Province, Lanzhou 730000, PR China
| | - Hai-Hong Zhang
- Lanzhou University Second Hospital, 82 Cuiying Men, Lanzhou 730000, PR China; Orthopedics Key Laboratory of Gansu Province, Lanzhou 730000, PR China.
| |
Collapse
|
35
|
Garnish SE, Horne CR, Meng Y, Young SN, Jacobsen AV, Hildebrand JM, Murphy JM. Inhibitors identify an auxiliary role for mTOR signalling in necroptosis execution downstream of MLKL activation. Biochem J 2024; 481:1125-1142. [PMID: 39136677 PMCID: PMC11555701 DOI: 10.1042/bcj20240255] [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: 05/27/2024] [Revised: 08/11/2024] [Accepted: 08/13/2024] [Indexed: 08/28/2024]
Abstract
Necroptosis is a lytic and pro-inflammatory form of programmed cell death executed by the terminal effector, the MLKL (mixed lineage kinase domain-like) pseudokinase. Downstream of death and Toll-like receptor stimulation, MLKL is trafficked to the plasma membrane via the Golgi-, actin- and microtubule-machinery, where activated MLKL accumulates until a critical lytic threshold is exceeded and cell death ensues. Mechanistically, MLKL's lytic function relies on disengagement of the N-terminal membrane-permeabilising four-helix bundle domain from the central autoinhibitory brace helix: a process that can be experimentally mimicked by introducing the R30E MLKL mutation to induce stimulus-independent cell death. Here, we screened a library of 429 kinase inhibitors for their capacity to block R30E MLKL-mediated cell death, to identify co-effectors in the terminal steps of necroptotic signalling. We identified 13 compounds - ABT-578, AR-A014418, AZD1480, AZD5363, Idelalisib, Ipatasertib, LJI308, PHA-793887, Rapamycin, Ridaforolimus, SMI-4a, Temsirolimus and Tideglusib - each of which inhibits mammalian target of rapamycin (mTOR) signalling or regulators thereof, and blocked constitutive cell death executed by R30E MLKL. Our study implicates mTOR signalling as an auxiliary factor in promoting the transport of activated MLKL oligomers to the plasma membrane, where they accumulate into hotspots that permeabilise the lipid bilayer to cause cell death.
Collapse
Affiliation(s)
- Sarah E. Garnish
- Walter and Eliza Hall Institute of Medical Research, 1G Royal Parade, Parkville, Victoria 3052, Australia
- Department of Medical Biology, University of Melbourne, Parkville, Victoria 3052, Australia
| | - Christopher R. Horne
- Walter and Eliza Hall Institute of Medical Research, 1G Royal Parade, Parkville, Victoria 3052, Australia
- Department of Medical Biology, University of Melbourne, Parkville, Victoria 3052, Australia
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria 3052, Australia
| | - Yanxiang Meng
- Walter and Eliza Hall Institute of Medical Research, 1G Royal Parade, Parkville, Victoria 3052, Australia
- Department of Medical Biology, University of Melbourne, Parkville, Victoria 3052, Australia
| | - Samuel N. Young
- Walter and Eliza Hall Institute of Medical Research, 1G Royal Parade, Parkville, Victoria 3052, Australia
| | - Annette V. Jacobsen
- Walter and Eliza Hall Institute of Medical Research, 1G Royal Parade, Parkville, Victoria 3052, Australia
- Department of Medical Biology, University of Melbourne, Parkville, Victoria 3052, Australia
| | - Joanne M. Hildebrand
- Walter and Eliza Hall Institute of Medical Research, 1G Royal Parade, Parkville, Victoria 3052, Australia
- Department of Medical Biology, University of Melbourne, Parkville, Victoria 3052, Australia
| | - James M. Murphy
- Walter and Eliza Hall Institute of Medical Research, 1G Royal Parade, Parkville, Victoria 3052, Australia
- Department of Medical Biology, University of Melbourne, Parkville, Victoria 3052, Australia
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria 3052, Australia
| |
Collapse
|
36
|
Zhu T, Wu BW. Recognition of necroptosis: From molecular mechanisms to detection methods. Biomed Pharmacother 2024; 178:117196. [PMID: 39053418 DOI: 10.1016/j.biopha.2024.117196] [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: 05/11/2024] [Revised: 07/05/2024] [Accepted: 07/22/2024] [Indexed: 07/27/2024] Open
Abstract
Necroptosis is a crucial modality of programmed cell death characterized by distinct morphological and biochemical hallmarks, including cell membrane rupture, organelle swelling, cytoplasmic and nuclear disintegration, cellular contents leakage, and release of damage-associated molecular patterns (DAMPs), accompanied by the inflammatory responses. Studies have shown that necroptosis is involved in the etiology and evolution of a variety of pathologies including organ damage, inflammation disorders, and cancer. Despite its significance, the field of necroptosis research grapples with the challenge of non-standardized detection methodologies. In this review, we introduce the fundamental concepts and molecular mechanisms of necroptosis and critically appraise the principles, merits, and inherent limitations of current detection technologies. This endeavor seeks to establish a methodological framework for necroptosis detection, thereby propelling deeper insights into the research of cell necroptosis.
Collapse
Affiliation(s)
- Ting Zhu
- Department of pharmacy, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang 441000, China
| | - Bo-Wen Wu
- School of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450001, China.
| |
Collapse
|
37
|
Deng M, Tang C, Yin L, Yang J, Chen Z, Jiang Y, Huang Y, Chen C. Screening chondrocyte necroptosis-related genes in the diagnosis and treatment of osteoarthritis. Heliyon 2024; 10:e35263. [PMID: 39170298 PMCID: PMC11336430 DOI: 10.1016/j.heliyon.2024.e35263] [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: 04/24/2024] [Revised: 07/18/2024] [Accepted: 07/25/2024] [Indexed: 08/23/2024] Open
Abstract
Background Osteoarthritis (OA) is the most common form of joint diseases, with hallmark of cartilage degeneration. Recent studies have shown that the pathogenesis of OA is associated with chondrocyte necroptosis. Methods In this study, we used single-cell RNA sequencing (scRNA-seq) and bulk RNA sequencing data to analyze necroptosis regulation in OA chondrocytes. We performed enrichment analysis, carried out experimental validation, constructed machine learning models, and docked drug molecules. Results After least absolute shrinkage and selection operator (LASSO) algorithm screening, 4 hub genes (RIPK3, CYBB, HSP90AB1, and TRAF5) with diagnostic characteristics were obtained. Following the comparison of multiple models, the Bayesian model with an average area under curve (AUC) value of 0.944 was finally selected. We found that nimesulide exhibited strong binding affinity to CYBB and HSP90AB1, and experimentally verified that nimesulide reduced the expression of RIPK3 and CYBB, suggesting its potential as an inhibitor of chondrocyte necroptosis. Furthermore, scRNA-seq results showed that necroptosis in OA was significantly upregulated on regulatory chondrocytes (RegC) compared to other chondrocyte subtypes. Conclusions The results indicate that nimesulide might be used to treat OA by inhibiting chondrocyte necroptosis through down-regulation of RIK3 and CYBB genes. This study reveals the role of chondrocyte necroptosis in OA, and suggests a potential therapeutic strategy by regulating necroptosis with nimesulide.
Collapse
Affiliation(s)
- Muhai Deng
- College of Medical Informatics, Chongqing Medical University, Chongqing, 400016, China
| | - Cong Tang
- College of Medical Informatics, Chongqing Medical University, Chongqing, 400016, China
| | - Li Yin
- Department of Orthopaedics, General Hospital of Western Theater Command, Chengdu, 610083, China
| | - Junjun Yang
- Key Laboratory of Biorheological Science and Technology, Ministry of Education College of Bioengineering, Chongqing University, Chongqing, 400044, China
| | - Zhiyu Chen
- Orthopedic Laboratory of Chongqing Medical University, Chongqing, 400016, China
- Department of Orthopedics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Yunsheng Jiang
- College of Medical Informatics, Chongqing Medical University, Chongqing, 400016, China
| | - Yang Huang
- State Key Laboratory of Trauma, Burns and Combined Injury, Department of Wound Infection and Drug, Daping Hospital, Army Medical University, Chongqing, 400042, China
| | - Cheng Chen
- College of Medical Informatics, Chongqing Medical University, Chongqing, 400016, China
| |
Collapse
|
38
|
Yin J, Yu Y, Huang X, Chan FKM. Necroptosis in immunity, tissue homeostasis, and cancer. Curr Opin Immunol 2024; 89:102455. [PMID: 39167896 DOI: 10.1016/j.coi.2024.102455] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Accepted: 08/06/2024] [Indexed: 08/23/2024]
Abstract
Immune and tissue homeostasis is achieved through balancing signals that regulate cell survival, proliferation, and cell death. Recent studies indicate that certain cell death programs can stimulate inflammation and are often referred as 'immunogenic cell death' (ICD). ICD is a double-edged sword that can confer protection against pathogen infection but also cause tissue damage. Necroptosis is a key ICD module that has been shown to participate in host defense against pathogen infection, tissue homeostasis, and cancer response to immunotherapy. Here, we will review recent findings on the regulation of necroptosis signaling and its role in pathogen infection, tissue homeostasis, and cancer.
Collapse
Affiliation(s)
| | - Yuqiang Yu
- Department of Cardiology of the Second Affiliated Hospital, China; State Key Laboratory of Transvascular Implantation Devices, Heart Regeneration and Repair Key Laboratory of Zhejiang Province, China
| | | | - Francis K-M Chan
- Department of Cardiology of the Second Affiliated Hospital, China; State Key Laboratory of Transvascular Implantation Devices, Heart Regeneration and Repair Key Laboratory of Zhejiang Province, China; Liangzhu Laboratory, China; Zhejiang University School of Medicine, 1369 West Wenyi Road, Hangzhou 311121, China.
| |
Collapse
|
39
|
Lyu P, Wen J, Zhang W, Liu N, Stolzer I, Gießl A, Jia Y, Mauro D, Zhang F, Ciccia F, Soulat D, Günther C, Schett G, Bozec A. Expression of HIF1α in intestinal epithelium restricts arthritis inflammation by inhibiting RIPK3-induced cell death machinery. Ann Rheum Dis 2024; 83:984-997. [PMID: 38503474 PMCID: PMC11287550 DOI: 10.1136/ard-2023-224491] [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: 06/06/2023] [Accepted: 02/28/2024] [Indexed: 03/21/2024]
Abstract
OBJECTIVES To investigate the mechanism by which intestinal epithelial cell (IEC) death induces arthritis. METHODS IEC death was assessed by staining for necroptosis and apoptosis markers and fluorescence in situ hybridisation at different time points during collagen-induced arthritis (CIA). During the development of CIA, messenger RNA (mRNA) sequencing was performed, followed by Gene Ontology enrichment analysis of differentially expressed genes. Mice deficient for hypoxia-inducible factor 1α (Hif1a) in IECs (Hif1a ∆IEC) were generated and induced for arthritis. mRNA sequencing, chromatin immunoprecipitated (ChIP) DNA sequencing and ChIP-qualitative PCR were performed on IECs from Hif1a ∆IEC mice and littermate controls. Effects of HIF1α stabilisation by inhibition of prolyl hydroxylase domain-containing enzymes and treatment with the inhibitor of receptor-interacting protein kinase-3 (RIPK3) were tested in intestinal organoids and in CIA. RESULTS IEC underwent apoptotic and necroptotic cell death at the onset of arthritis, leading to impaired gut barrier function. HIF1α was identified as one of the most upregulated genes in IECs during the onset of arthritis. Deletion of Hif1a in IEC enhanced IEC necroptosis, triggered intestinal inflammation and exacerbated arthritis. HIF1α was found to be a key transcriptional repressor for the necroptosis-inducing factor RIPK3. Enhanced RIPK3 expression, indicating necroptosis, was also found in the intestinal epithelium of patients with new-onset rheumatoid arthritis. Therapeutic stabilisation of HIF1α as well as small-molecule-based RIPK3 inhibition rescued intestinal necroptosis in vitro and in vivo and suppressed the development of arthritis. CONCLUSION Our results identify IEC necroptosis as a critical link between the gut and the development of arthritis.
Collapse
Affiliation(s)
- Pang Lyu
- Department of Internal Medicine 3, Rheumatology and Immunology, Friedrich-Alexander University (FAU) Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany
- Deutsches Zentrum Immuntherapie, Friedrich-Alexander University (FAU) Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Jinming Wen
- Department of Internal Medicine 3, Rheumatology and Immunology, Friedrich-Alexander University (FAU) Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany
- Deutsches Zentrum Immuntherapie, Friedrich-Alexander University (FAU) Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Wenshuo Zhang
- Department of Internal Medicine 3, Rheumatology and Immunology, Friedrich-Alexander University (FAU) Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany
- Deutsches Zentrum Immuntherapie, Friedrich-Alexander University (FAU) Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Ning Liu
- Department of Internal Medicine 3, Rheumatology and Immunology, Friedrich-Alexander University (FAU) Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany
- Deutsches Zentrum Immuntherapie, Friedrich-Alexander University (FAU) Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Iris Stolzer
- Deutsches Zentrum Immuntherapie, Friedrich-Alexander University (FAU) Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany
- Department of Internal Medicine 1, Friedrich-Alexander University (FAU) Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Bayern, Germany
| | - Andreas Gießl
- Department of Opthalmology, Friedrich-Alexander University (FAU) Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Yewei Jia
- Department of Internal Medicine 3, Rheumatology and Immunology, Friedrich-Alexander University (FAU) Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany
- Deutsches Zentrum Immuntherapie, Friedrich-Alexander University (FAU) Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Daniele Mauro
- Precision Medicine, Università degli Studi della Campania Luigi Vanvitelli, Napoli, Campania, Italy
| | - Fulin Zhang
- Department of Internal Medicine 3, Rheumatology and Immunology, Friedrich-Alexander University (FAU) Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany
- Deutsches Zentrum Immuntherapie, Friedrich-Alexander University (FAU) Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Francesco Ciccia
- Precision Medicine, Università degli Studi della Campania Luigi Vanvitelli, Napoli, Campania, Italy
| | - Didier Soulat
- Microbiology Institute, Friedrich-Alexander University (FAU) Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Claudia Günther
- Deutsches Zentrum Immuntherapie, Friedrich-Alexander University (FAU) Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany
- Department of Internal Medicine 1, Friedrich-Alexander University (FAU) Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Bayern, Germany
| | - Georg Schett
- Department of Internal Medicine 3, Rheumatology and Immunology, Friedrich-Alexander University (FAU) Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany
- Deutsches Zentrum Immuntherapie, Friedrich-Alexander University (FAU) Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Aline Bozec
- Department of Internal Medicine 3, Rheumatology and Immunology, Friedrich-Alexander University (FAU) Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany
- Deutsches Zentrum Immuntherapie, Friedrich-Alexander University (FAU) Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany
| |
Collapse
|
40
|
Liu X, Yao Y, Zhu Y, Lu F, Chen X. Inhibition of Adipocyte Necroptosis Alleviates Fat Necrosis and Fibrosis After Grafting in a Murine Model. Aesthet Surg J 2024; 44:NP585-NP605. [PMID: 38796831 DOI: 10.1093/asj/sjae108] [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: 01/16/2024] [Revised: 04/24/2024] [Accepted: 04/24/2024] [Indexed: 05/29/2024] Open
Abstract
BACKGROUND Because of the delicate structure of the adipose tissue, fat necrosis accounts for 43.7% of all complications after autologous fat grafting; however, its regulation remains unclear. OBJECTIVES The purpose of this study was to examine the role of necroptosis in fat graft remodeling after grafting. METHODS Clinical fat graft necrosis samples were collected, and the expression levels of the necroptosis marker phosphorylated(p)-MLKL were analyzed. Transcriptome analysis was performed on fat grafts before and 1 week after transplantation in C57BL/6 mouse fat grafting models. Additionally, the in vivo effects of RIPK1 inhibitor Nec-1s or RIPK3 inhibitor GSK'872 on the fat grafting complications, including fat necrosis and fibrosis, were investigated. RESULTS Necroptosis markers were observed and associated with higher occurrence of fibrosis in clinical fat graft necrosis samples compared to normal fat tissue. Amplification and RNA-Seq were conducted on RNA isolated from fat grafts before and after grafting. MLKL, RIPK1, and RIPK3's expression levels were significantly upregulated in comparison to controls. Higher expression levels of necroptotic RNAs were associated with higher levels of DAMPs, including Cxcl2, HMGB1, S100a8, S100a9, Nlrp3, and IL33, and activated proinflammatory signaling pathways, including the TNF, NF-kappa B, and chemokine signaling pathways. Necroptotic inhibitor Nec-1s and GSK'872 robustly suppressed the p-MLKL expression level and significantly inhibited necroptotic cell death, especially in adipocytes. Moreover, administration of Nec-1s and GSK'872 significantly alleviated fat necrosis and subsequent fibrosis in fat grafts. CONCLUSIONS Collectively, our study findings highlight the potential therapeutic applications of necroptosis inhibitors in preventing fat necrosis and fibrosis after grafting. LEVEL OF EVIDENCE: 4
Collapse
|
41
|
Osbron CA, Lawson C, Hanna N, Koehler HS, Goodman AG. Caspase-8 activity mediates TNFα production and restricts Coxiella burnetii replication during murine macrophage infection. Infect Immun 2024; 92:e0005324. [PMID: 38837340 PMCID: PMC11238558 DOI: 10.1128/iai.00053-24] [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: 01/31/2024] [Accepted: 05/14/2024] [Indexed: 06/07/2024] Open
Abstract
Coxiella burnetii is an obligate intracellular bacteria that causes the global zoonotic disease Q Fever. Treatment options for chronic infection are limited, and the development of novel therapeutic strategies requires a greater understanding of how C. burnetii interacts with immune signaling. Cell death responses are known to be manipulated by C. burnetii, but the role of caspase-8, a central regulator of multiple cell death pathways, has not been investigated. In this research, we studied bacterial manipulation of caspase-8 signaling and the significance of caspase-8 to C. burnetii infection, examining bacterial replication, cell death induction, and cytokine signaling. We measured caspase, RIPK, and MLKL activation in C. burnetii-infected tumor necrosis factor alpha (TNFα)/cycloheximide-treated THP-1 macrophage-like cells and TNFα/ZVAD-treated L929 cells to assess apoptosis and necroptosis signaling. Additionally, we measured C. burnetii replication, cell death, and TNFα induction over 12 days in RIPK1-kinase-dead, RIPK3-kinase-dead, or RIPK3-kinase-dead-caspase-8-/- bone marrow-derived macrophages (BMDMs) to understand the significance of caspase-8 and RIPK1/3 during infection. We found that caspase-8 is inhibited by C. burnetii, coinciding with inhibition of apoptosis and increased susceptibility to necroptosis. Furthermore, C. burnetii replication was increased in BMDMs lacking caspase-8, but not in those lacking RIPK1/3 kinase activity, corresponding with decreased TNFα production and reduced cell death. As TNFα is associated with the control of C. burnetii, this lack of a TNFα response may allow for the unchecked bacterial growth we saw in caspase-8-/- BMDMs. This research identifies and explores caspase-8 as a key regulator of C. burnetii infection, opening novel therapeutic doors.
Collapse
Affiliation(s)
- Chelsea A. Osbron
- School of Molecular Biosciences, College of Veterinary Medicine, Washington State University, Pullman, Washington, USA
| | - Crystal Lawson
- School of Molecular Biosciences, College of Veterinary Medicine, Washington State University, Pullman, Washington, USA
| | - Nolan Hanna
- School of Molecular Biosciences, College of Veterinary Medicine, Washington State University, Pullman, Washington, USA
| | - Heather S. Koehler
- School of Molecular Biosciences, College of Veterinary Medicine, Washington State University, Pullman, Washington, USA
| | - Alan G. Goodman
- School of Molecular Biosciences, College of Veterinary Medicine, Washington State University, Pullman, Washington, USA
- Paul G. Allen School for Global Health, College of Veterinary Medicine, Washington State University, Pullman, Washington, USA
| |
Collapse
|
42
|
Imai T, Lin J, Kaya GG, Ju E, Kondylis V, Kelepouras K, Liccardi G, Kim C, Pasparakis M. The RIPK1 death domain restrains ZBP1- and TRIF-mediated cell death and inflammation. Immunity 2024; 57:1497-1513.e6. [PMID: 38744293 DOI: 10.1016/j.immuni.2024.04.016] [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] [Revised: 02/05/2024] [Accepted: 04/17/2024] [Indexed: 05/16/2024]
Abstract
RIPK1 is a multi-functional kinase that regulates cell death and inflammation and has been implicated in the pathogenesis of inflammatory diseases. RIPK1 acts in a kinase-dependent and kinase-independent manner to promote or suppress apoptosis and necroptosis, but the underlying mechanisms remain poorly understood. Here, we show that a mutation (R588E) disrupting the RIPK1 death domain (DD) caused perinatal lethality induced by ZBP1-mediated necroptosis. Additionally, these mice developed postnatal inflammatory pathology, which was mediated by necroptosis-independent TNFR1, TRADD, and TRIF signaling, partially requiring RIPK3. Our biochemical mechanistic studies revealed that ZBP1- and TRIF-mediated activation of RIPK3 required RIPK1 kinase activity in wild-type cells but not in Ripk1R588E/R588E cells, suggesting that DD-dependent oligomerization of RIPK1 and its interaction with FADD determine the mechanisms of RIPK3 activation by ZBP1 and TRIF. Collectively, these findings revealed a critical physiological role of DD-dependent RIPK1 signaling that is important for the regulation of tissue homeostasis and inflammation.
Collapse
Affiliation(s)
- Takashi Imai
- Institute for Genetics, University of Cologne, 50674 Cologne, Germany; Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, 50931 Cologne, Germany
| | - Juan Lin
- Institute for Genetics, University of Cologne, 50674 Cologne, Germany; Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, 50931 Cologne, Germany; Research Unit of Cellular Stress of Chinese Academy of Medical Sciences, Cancer Research Center of Xiamen University, School of Medicine, Xiamen University, Xiamen, China.
| | - Göksu Gökberk Kaya
- Institute for Genetics, University of Cologne, 50674 Cologne, Germany; Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, 50931 Cologne, Germany
| | - Eunjin Ju
- Institute for Genetics, University of Cologne, 50674 Cologne, Germany; Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, 50931 Cologne, Germany
| | - Vangelis Kondylis
- Center for Molecular Medicine (CMMC), University of Cologne, 50931 Cologne, Germany; Institute of Pathology, Faculty of Medicine and University Hospital Cologne, 50931 Cologne, Germany
| | - Konstantinos Kelepouras
- Institute of Biochemistry I, Center for Biochemistry, Faculty of Medicine, University of Cologne, Joseph-Stelzmann-Str. 52, 50931 Cologne, Germany
| | - Gianmaria Liccardi
- Center for Molecular Medicine (CMMC), University of Cologne, 50931 Cologne, Germany; Institute of Biochemistry I, Center for Biochemistry, Faculty of Medicine, University of Cologne, Joseph-Stelzmann-Str. 52, 50931 Cologne, Germany
| | - Chun Kim
- Institute for Genetics, University of Cologne, 50674 Cologne, Germany; Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, 50931 Cologne, Germany; Department of Medicinal and Life Sciences, Hanyang University (ERICA Campus), Ansan 15588, Republic of Korea
| | - Manolis Pasparakis
- Institute for Genetics, University of Cologne, 50674 Cologne, Germany; Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, 50931 Cologne, Germany; Center for Molecular Medicine (CMMC), University of Cologne, 50931 Cologne, Germany.
| |
Collapse
|
43
|
Min Y, Yu ZQ. GSK'872 Improves Prognosis of Traumatic Brain Injury by Switching Receptor-Interacting Serine/Threonine-Protein Kinase 3-dependent Necroptosis to Cysteinyl Aspartate Specific Proteinase-8-Dependent Apoptosis. World Neurosurg 2024; 187:e136-e147. [PMID: 38636634 DOI: 10.1016/j.wneu.2024.04.051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Revised: 04/10/2024] [Accepted: 04/11/2024] [Indexed: 04/20/2024]
Abstract
BACKGROUND Traumatic brain injury (TBI) is an important health concern in the society. Previous studies have suggested that necroptosis occurs following TBI. However, the underlying mechanisms and roles of necroptosis are not well understood. In this study, we aimed to assess the role of receptor-interacting serine/threonine-protein kinase 3 (RIP3)-mediated necroptosis after TBI both in vitro and in vivo. METHODS We established a cell-stretching injury and mouse TBI model by applying a cell injury controller and controlled cortical impactor to evaluate the relationships among necroptosis, apotosis, inflammation, and TBI both in vitro and in vivo. RESULTS The results revealed that necroptosis mediated by RIP1, RIP3, and mixed lineage kinase domain-like protein was involved in secondary TBI. Additionally, protein kinase B (Akt), phosphorylated Akt, mammalian target of rapamycin (mTOR), and phosphorylated mTOR potentially contribute to necroptosis. The inhibition of RIP3 by GSK'872 (a specific inhibitor) blocked necroptosis and reduced the activity of Akt/mTOR, leading to the alleviation of inflammation by reducing the levels of NOD-, LRR- and pyrin domain-containing protein 3. Moreover, the inhibition of RIP3 by GSK'872 promoted the activity of cysteinyl aspartate specific proteinase-8, an enzyme involved in apoptosis and inflammation. CONCLUSIONS These data demonstrate that RIP3 inhibition could improve the prognosis of TBI, based on the attenuation of inflammation by switching RIP3-dependent necroptosis to cysteinyl aspartate specific proteinase-8-dependent apoptosis.
Collapse
Affiliation(s)
- Yue Min
- Department of Neurosurgery, West China Hospital, Sichuan University/West China School of Nursing, Sichuan University, Chengdu, Sichuan, China
| | - Ze-Qi Yu
- Department of Neurosurgery, Armed Police Force Hospital of Sichuan, Leshan, Sichuan, China.
| |
Collapse
|
44
|
Oh TJ, Krishnamurthy V, Han JW, Zhu J, Beg Z, Mehfooz A, Gworek B, Shapiro DJ, Zhang K. Spatiotemporal Control of Inflammatory Lytic Cell Death Through Optogenetic Induction of RIPK3 Oligomerization. J Mol Biol 2024; 436:168628. [PMID: 38797430 PMCID: PMC11234905 DOI: 10.1016/j.jmb.2024.168628] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Revised: 04/21/2024] [Accepted: 05/21/2024] [Indexed: 05/29/2024]
Abstract
Necroptosis is a programmed lytic cell death involving active cytokine production and plasma membrane rupture through distinct signaling cascades. However, it remains challenging to delineate this inflammatory cell death pathway at specific signaling nodes with spatiotemporal accuracy. To address this challenge, we developed an optogenetic system, termed Light-activatable Receptor-Interacting Protein Kinase 3 or La-RIPK3, to enable ligand-free, optical induction of RIPK3 oligomerization. La-RIPK3 activation dissects RIPK3-centric lytic cell death through the induction of RIPK3-containing necrosome, which mediates cytokine production and plasma membrane rupture. Bulk RNA-Seq analysis reveals that RIPK3 oligomerization results in partially overlapped gene expression compared to pharmacological induction of necroptosis. Additionally, La-RIPK3 activates separated groups of genes regulated by RIPK3 kinase-dependent and -independent processes. Using patterned light stimulation delivered by a spatial light modulator, we demonstrate precise spatiotemporal control of necroptosis in La-RIPK3-transduced HT-29 cells. Optogenetic control of proinflammatory lytic cell death could lead to the development of innovative experimental strategies to finetune the immune landscape for disease intervention.
Collapse
Affiliation(s)
- Teak-Jung Oh
- Department of Biochemistry, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - Vishnu Krishnamurthy
- High-throughput Screening Center, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - Jeong Won Han
- Department of Biochemistry, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - Junyao Zhu
- Department of Biochemistry, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - Zayn Beg
- Department of Biochemistry, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - Amna Mehfooz
- Department of Biochemistry, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - Bryan Gworek
- Center for Biophysics and Quantitative Biology, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - David J Shapiro
- Department of Biochemistry, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - Kai Zhang
- Department of Biochemistry, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA; Center for Biophysics and Quantitative Biology, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA; Cancer Center at Illinois, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA; NSF Science and Technology Center for Quantitative Cell Biology, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA.
| |
Collapse
|
45
|
Koerner L, Wachsmuth L, Kumari S, Schwarzer R, Wagner T, Jiao H, Pasparakis M. ZBP1 causes inflammation by inducing RIPK3-mediated necroptosis and RIPK1 kinase activity-independent apoptosis. Cell Death Differ 2024; 31:938-953. [PMID: 38849574 PMCID: PMC11239871 DOI: 10.1038/s41418-024-01321-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Revised: 05/24/2024] [Accepted: 05/29/2024] [Indexed: 06/09/2024] Open
Abstract
Z-DNA binding protein 1 (ZBP1) has important functions in anti-viral immunity and in the regulation of inflammatory responses. ZBP1 induces necroptosis by directly engaging and activating RIPK3, however, the mechanisms by which ZBP1 induces inflammation and in particular the role of RIPK1 and the contribution of cell death-independent signaling remain elusive. Here we show that ZBP1 causes skin inflammation by inducing RIPK3-mediated necroptosis and RIPK1-caspase-8-mediated apoptosis in keratinocytes. ZBP1 induced TNFR1-independent skin inflammation in mice with epidermis-specific ablation of FADD by triggering keratinocyte necroptosis. Moreover, transgenic expression of C-terminally truncated constitutively active ZBP1 (ZBP1ca) in mouse epidermis caused skin inflammation that was only partially inhibited by abrogation of RIPK3-MLKL-dependent necroptosis and fully prevented by combined deficiency in MLKL and caspase-8. Importantly, ZBP1ca induced caspase-8-mediated skin inflammation by RHIM-dependent but kinase activity-independent RIPK1 signaling. Furthermore, ZBP1ca-induced inflammatory cytokine production in the skin was completely prevented by combined inhibition of apoptosis and necroptosis arguing against a cell death-independent pro-inflammatory function of ZBP1. Collectively, these results showed that ZBP1 induces inflammation by activating necroptosis and RIPK1 kinase activity-independent apoptosis.
Collapse
Affiliation(s)
- Lioba Koerner
- Institute for Genetics, University of Cologne, 50674, Cologne, Germany
- Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, 50931, Cologne, Germany
| | - Laurens Wachsmuth
- Institute for Genetics, University of Cologne, 50674, Cologne, Germany
- Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, 50931, Cologne, Germany
| | - Snehlata Kumari
- Institute for Genetics, University of Cologne, 50674, Cologne, Germany
- Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, 50931, Cologne, Germany
- Frazer Institute, The University of Queensland, Faculty of Medicine, Brisbane, QLD, Australia
| | - Robin Schwarzer
- Institute for Genetics, University of Cologne, 50674, Cologne, Germany
- Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, 50931, Cologne, Germany
- Genentech Inc, South San Francisco, CA, USA
| | - Theresa Wagner
- Institute for Genetics, University of Cologne, 50674, Cologne, Germany
- Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, 50931, Cologne, Germany
| | - Huipeng Jiao
- Institute for Genetics, University of Cologne, 50674, Cologne, Germany
- Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, 50931, Cologne, Germany
- Life Sciences Institute, Zhejiang University, Hangzhou, China
| | - Manolis Pasparakis
- Institute for Genetics, University of Cologne, 50674, Cologne, Germany.
- Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, 50931, Cologne, Germany.
- Center for Molecular Medicine (CMMC), Medical Faculty and University Hospital Cologne, University of Cologne, 50931, Cologne, Germany.
| |
Collapse
|
46
|
Kelepouras K, Saggau J, Varanda AB, Zrilic M, Kiefer C, Rakhsh-Khorshid H, Lisewski I, Uranga-Murillo I, Arias M, Pardo J, Tonnus W, Linkermann A, Annibaldi A, Walczak H, Liccardi G. The importance of murine phospho-MLKL-S345 in situ detection for necroptosis assessment in vivo. Cell Death Differ 2024; 31:897-909. [PMID: 38783091 PMCID: PMC11239901 DOI: 10.1038/s41418-024-01313-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Revised: 05/02/2024] [Accepted: 05/07/2024] [Indexed: 05/25/2024] Open
Abstract
Necroptosis is a caspase-independent modality of cell death implicated in many inflammatory pathologies. The execution of this pathway requires the formation of a cytosolic platform that comprises RIPK1 and RIPK3 which, in turn, mediates the phosphorylation of the pseudokinase MLKL (S345 in mouse). The activation of this executioner is followed by its oligomerisation and accumulation at the plasma-membrane where it leads to cell death via plasma-membrane destabilisation and consequent permeabilisation. While the biochemical and cellular characterisation of these events have been amply investigated, the study of necroptosis involvement in vivo in animal models is currently limited to the use of Mlkl-/- or Ripk3-/- mice. Yet, even in many of the models in which the involvement of necroptosis in disease aetiology has been genetically demonstrated, the fundamental in vivo characterisation regarding the question as to which tissue(s) and specific cell type(s) therein is/are affected by the pathogenic necroptotic death are missing. Here, we describe and validate an immunohistochemistry and immunofluorescence-based method to reliably detect the phosphorylation of mouse MLKL at serine 345 (pMLKL-S345). We first validate the method using tissues derived from mice in which Caspase-8 (Casp8) or FADD are specifically deleted from keratinocytes, or intestinal epithelial cells, respectively. We next demonstrate the presence of necroptotic activation in the lungs of SARS-CoV-infected mice and in the skin and spleen of mice bearing a Sharpin inactivating mutation. Finally, we exclude necroptosis occurrence in the intestines of mice subjected to TNF-induced septic shock. Importantly, by directly comparing the staining of pMLKL-345 with that of cleaved Caspase-3 staining in some of these models, we identify spatio-temporal and functional differences between necroptosis and apoptosis supporting a role of RIPK3 in inflammation independently of MLKL versus the role of RIPK3 in activation of necroptosis.
Collapse
Grants
- Wellcome Trust
- G.L. is funded by the Center for Biochemistry, Univeristy of Cologne - 956400, Köln Fortune, CANcer TARgeting (CANTAR) project NW21-062A, two collaborative research center grants: SFB1399-413326622 Project C06, SFB1530-455784452 Project A03 both funded by the Deutsche Forschungsgemeinschaft (DFG)) and associated to the collaborative SFB1403 also funded by the DFG
- H.W. is funded by the Alexander von Humboldt Foundation, a Wellcome Trust Investigator Award (214342/Z/18/Z), a Medical Research Council Grant (MR/S00811X/1), a Cancer Research UK Programme Grant (A27323) and three collaborative research center grants (SFB1399, Project C06, SFB1530-455784452, Project A03 and SFB1403–414786233) funded by the Deutsche Forschungsgemeinschaft (DFG) and CANcer TARgeting (CANTAR) funded by Netzwerke 2021.
- AA is funded by the Center for Molecular Medine Cologne (CMMC) Junior Research Group program, Deutsche Forschungsgemeinschaft (DFG) (project number AN1717/1-1), the Jürgen Manchot Stiftung foundation, the collaborative research center SFB1530 (Project A5, ID: 455784452)
- JP is funded by FEDER (Fondo Europeo de Desarrollo Regional), Gobierno de Aragón (Group B29_23R), CIBERINFEC (CB21/13/00087), Ministerio de Ciencia, Innovación y Universidades (MCNU)/Agencia Estatal de Investigación (PID2020-113963RBI00)
- MA is funded by a Postdoctoral Juan de la Cierva Contract.
- Work in the Linkermann Lab was funded by the German Research Foundation SFB-TRR205, SFB-TRR 127, SPP2306, and a Heisenberg-Professorship to A.L., project number 324141047, and the international research training group (IRTG) 2251. It was further supported by the BMBF (FERROPath consortium), the TU Dresden / Kings College London transcampus initiative and the DFG-Sachbeihilfe LI 2107/10-1.
Collapse
Affiliation(s)
- Konstantinos Kelepouras
- Genome Instability, Inflammation and Cell Death Laboratory, Institute of Biochemistry I, Centre for Biochemistry, Faculty of Medicine, University of Cologne, 50931, Cologne, Germany
- Center for Molecular Medicine Cologne (CMMC), University of Cologne, 50931, Cologne, Germany
| | - Julia Saggau
- Genome Instability, Inflammation and Cell Death Laboratory, Institute of Biochemistry I, Centre for Biochemistry, Faculty of Medicine, University of Cologne, 50931, Cologne, Germany
- Center for Molecular Medicine Cologne (CMMC), University of Cologne, 50931, Cologne, Germany
- Cell Death, Inflammation and Immunity Laboratory, CECAD Cluster of Excellence, University of Cologne, 50931, Cologne, Germany
- Cell Death, Inflammation and Immunity Laboratory, Institute of Biochemistry I, Centre for Biochemistry, Faculty of Medicine, University of Cologne, 50931, Cologne, Germany
| | - Ana Beatriz Varanda
- Cell Death, Inflammation and Immunity Laboratory, CECAD Cluster of Excellence, University of Cologne, 50931, Cologne, Germany
- Cell Death, Inflammation and Immunity Laboratory, Institute of Biochemistry I, Centre for Biochemistry, Faculty of Medicine, University of Cologne, 50931, Cologne, Germany
| | - Matea Zrilic
- Center for Molecular Medicine Cologne (CMMC), University of Cologne, 50931, Cologne, Germany
| | - Christine Kiefer
- Genome Instability, Inflammation and Cell Death Laboratory, Institute of Biochemistry I, Centre for Biochemistry, Faculty of Medicine, University of Cologne, 50931, Cologne, Germany
- Center for Molecular Medicine Cologne (CMMC), University of Cologne, 50931, Cologne, Germany
| | - Hassan Rakhsh-Khorshid
- Genome Instability, Inflammation and Cell Death Laboratory, Institute of Biochemistry I, Centre for Biochemistry, Faculty of Medicine, University of Cologne, 50931, Cologne, Germany
- Center for Molecular Medicine Cologne (CMMC), University of Cologne, 50931, Cologne, Germany
| | - Ina Lisewski
- Center for Molecular Medicine Cologne (CMMC), University of Cologne, 50931, Cologne, Germany
| | - Iratxe Uranga-Murillo
- Department of Microbiology, Radiology, Paediatry and Public Heath, Faculty of Medicine, University of Zaragoza/IIS, Aragon, Spain
- Centro de Investigacion Biomedica en Red de Enfermedades infecciosas (CIBERINFEC), Instituto de Salud Carlos III, Madrid, Spain
| | - Maykel Arias
- Department of Microbiology, Radiology, Paediatry and Public Heath, Faculty of Medicine, University of Zaragoza/IIS, Aragon, Spain
- Centro de Investigacion Biomedica en Red de Enfermedades infecciosas (CIBERINFEC), Instituto de Salud Carlos III, Madrid, Spain
| | - Julian Pardo
- Department of Microbiology, Radiology, Paediatry and Public Heath, Faculty of Medicine, University of Zaragoza/IIS, Aragon, Spain
- Centro de Investigacion Biomedica en Red de Enfermedades infecciosas (CIBERINFEC), Instituto de Salud Carlos III, Madrid, Spain
| | - Wulf Tonnus
- Division of Nephrology, Department of Internal Medicine 3, University Hospital Carl Gustav Carus at the Technische Universität Dresden, Dresden, Germany
| | - Andreas Linkermann
- Division of Nephrology, Department of Internal Medicine 3, University Hospital Carl Gustav Carus at the Technische Universität Dresden, Dresden, Germany
- Division of Nephrology, Department of Medicine, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Alessandro Annibaldi
- Center for Molecular Medicine Cologne (CMMC), University of Cologne, 50931, Cologne, Germany
| | - Henning Walczak
- Cell Death, Inflammation and Immunity Laboratory, CECAD Cluster of Excellence, University of Cologne, 50931, Cologne, Germany
- Cell Death, Inflammation and Immunity Laboratory, Institute of Biochemistry I, Centre for Biochemistry, Faculty of Medicine, University of Cologne, 50931, Cologne, Germany
- Centre for Cell Death, Cancer and Inflammation, UCL Cancer Institute, University College London, WC1E 6BT, London, UK
| | - Gianmaria Liccardi
- Genome Instability, Inflammation and Cell Death Laboratory, Institute of Biochemistry I, Centre for Biochemistry, Faculty of Medicine, University of Cologne, 50931, Cologne, Germany.
- Center for Molecular Medicine Cologne (CMMC), University of Cologne, 50931, Cologne, Germany.
| |
Collapse
|
47
|
Rodriguez DA, Tummers B, Shaw JJP, Quarato G, Weinlich R, Cripps J, Fitzgerald P, Janke LJ, Pelletier S, Crawford JC, Green DR. The interaction between RIPK1 and FADD controls perinatal lethality and inflammation. Cell Rep 2024; 43:114335. [PMID: 38850531 PMCID: PMC11256114 DOI: 10.1016/j.celrep.2024.114335] [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: 07/06/2023] [Revised: 04/15/2024] [Accepted: 05/23/2024] [Indexed: 06/10/2024] Open
Abstract
Perturbation of the apoptosis and necroptosis pathways critically influences embryogenesis. Receptor-associated protein kinase-1 (RIPK1) interacts with Fas-associated via death domain (FADD)-caspase-8-cellular Flice-like inhibitory protein long (cFLIPL) to regulate both extrinsic apoptosis and necroptosis. Here, we describe Ripk1-mutant animals (Ripk1R588E [RE]) in which the interaction between FADD and RIPK1 is disrupted, leading to embryonic lethality. This lethality is not prevented by further removal of the kinase activity of Ripk1 (Ripk1R588E K45A [REKA]). Both Ripk1RE and Ripk1REKA animals survive to adulthood upon ablation of Ripk3. While embryonic lethality of Ripk1RE mice is prevented by ablation of the necroptosis effector mixed lineage kinase-like (MLKL), animals succumb to inflammation after birth. In contrast, Mlkl ablation does not prevent the death of Ripk1REKA embryos, but animals reach adulthood when both MLKL and caspase-8 are removed. Ablation of the nucleic acid sensor Zbp1 largely prevents lethality in both Ripk1RE and Ripk1REKA embryos. Thus, the RIPK1-FADD interaction prevents Z-DNA binding protein-1 (ZBP1)-induced, RIPK3-caspase-8-mediated embryonic lethality, affected by the kinase activity of RIPK1.
Collapse
Affiliation(s)
- Diego A Rodriguez
- Department of Immunology, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN 38105, USA
| | - Bart Tummers
- Centre for Inflammation Biology & Cancer Immunology (CIBCI), Department of Inflammation Biology, School of Immunology & Microbial Sciences, King's College London, London SE1 1UL, UK.
| | - Jeremy J P Shaw
- Department of Immunology, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN 38105, USA
| | - Giovanni Quarato
- Department of Immunology, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN 38105, USA; Treeline Biosciences, San Diego, CA 92121, USA
| | | | - James Cripps
- Center for Cancer Immunology and Immunotherapy, Brown Cancer Center, University of Louisville School of Medicine, Louisville, KY, USA
| | - Patrick Fitzgerald
- Department of Immunology, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN 38105, USA
| | - Laura J Janke
- Department of Pathology, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN 38105, USA
| | - Stephane Pelletier
- Department of Medical and Molecular Genetics, Indiana University Genome Editing Center, Indiana University School of Medicine, Indiana University, Indianapolis, IA 46902, USA
| | - Jeremy Chase Crawford
- Department of Immunology, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN 38105, USA
| | - Douglas R Green
- Department of Immunology, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN 38105, USA.
| |
Collapse
|
48
|
Suzuki T, Uchida H. Induction of necroptosis in multinucleated giant cells induced by conditionally replicating syncytial oHSV in co-cultures of cancer cells and non-cancerous cells. MOLECULAR THERAPY. ONCOLOGY 2024; 32:200803. [PMID: 38706990 PMCID: PMC11067338 DOI: 10.1016/j.omton.2024.200803] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Accepted: 04/11/2024] [Indexed: 05/07/2024]
Abstract
Viral modifications enabling syncytium formation in infected cells can augment lysis by oncolytic herpes simplex viruses (oHSVs) which selectively kill cancer cells. In the case of receptor-retargeted oHSVs (RR-oHSVs) that exclusively enter and spread to cancer cells, anti-tumor effects can be enhanced in a magnitude of >100,000-fold by modifying the virus to a syncytial type (RRsyn-oHSV). However, when syncytia containing non-cancerous cells are induced by conditionally replicating syncytial oHSV (CRsyn-oHSV), syncytial death occurs at an early stage. This results in limited anti-tumor effects of the CRsyn-oHSV. Here, we investigated whether necroptosis is involved in death of the syncytia formed by the fusion of cancer cells and non-cancerous cells. Mixed-lineage kinase domain-like (MLKL), a molecule executing necroptosis, was expressed in all murine cancer cell lines examined, while receptor-interacting protein kinase 3 (RIPK3), which phosphorylates MLKL, was absent from most cell lines. In contrast, RIPK3 was expressed in non-cancerous murine fibroblast cell lines. When a CRsyn-oHSV-infected RIPK3-deficient cancer cell line was co-cultured with the fibroblast cell line, but not with the cancer cells themselves, MLKL was phosphorylated and syncytial death was induced. These results indicate that early necroptosis is induced in multinucleated giant cells formed by CRsyn-oHSV when they also contain non-cancerous cells.
Collapse
Affiliation(s)
- Takuma Suzuki
- Tokyo University of Pharmacy and Life Sciences, Tokyo 192-0392, Japan
- Project Division of Cancer Biomolecular Therapy, The Institute of Medical Science, The University of Tokyo, Tokyo 108-8639, Japan
| | - Hiroaki Uchida
- Tokyo University of Pharmacy and Life Sciences, Tokyo 192-0392, Japan
- Project Division of Cancer Biomolecular Therapy, The Institute of Medical Science, The University of Tokyo, Tokyo 108-8639, Japan
| |
Collapse
|
49
|
Bonnet MC, Sun L. Editorial: Necroptosis: from bench to bedside. Front Immunol 2024; 15:1441901. [PMID: 38962001 PMCID: PMC11220229 DOI: 10.3389/fimmu.2024.1441901] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Accepted: 06/10/2024] [Indexed: 07/05/2024] Open
Affiliation(s)
- Marion C. Bonnet
- Division of Infection and Immunity, Cardiff University School of Medicine, Cardiff, United Kingdom
| | - Liming Sun
- State Key Laboratory in Cell Biology Centre for Excellence in Molecular Cell Science Chinese Academy of Sciences, Shanghai, China
| |
Collapse
|
50
|
Qian Z, Xiong W, Mao X, Li J. Macrophage Perspectives in Liver Diseases: Programmed Death, Related Biomarkers, and Targeted Therapy. Biomolecules 2024; 14:700. [PMID: 38927103 PMCID: PMC11202214 DOI: 10.3390/biom14060700] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2024] [Revised: 06/06/2024] [Accepted: 06/10/2024] [Indexed: 06/28/2024] Open
Abstract
Macrophages, as important immune cells of the organism, are involved in maintaining intrahepatic microenvironmental homeostasis and can undergo rapid phenotypic changes in the injured or recovering liver. In recent years, the crucial role of macrophage-programmed cell death in the development and regression of liver diseases has become a research hotspot. Moreover, macrophage-targeted therapeutic strategies are emerging in both preclinical and clinical studies. Given the macrophages' vital role in complex organismal environments, there is tremendous academic interest in developing novel therapeutic strategies that target these cells. This review provides an overview of the characteristics and interactions between macrophage polarization, programmed cell death, related biomarkers, and macrophage-targeted therapies. It aims to deepen the understanding of macrophage immunomodulation and molecular mechanisms and to provide a basis for the treatment of macrophage-associated liver diseases.
Collapse
Affiliation(s)
- Zibing Qian
- The First Clinical Medical College of Lanzhou University, Lanzhou 730000, China; (Z.Q.); (W.X.)
| | - Wanyuan Xiong
- The First Clinical Medical College of Lanzhou University, Lanzhou 730000, China; (Z.Q.); (W.X.)
| | - Xiaorong Mao
- The First Clinical Medical College of Lanzhou University, Lanzhou 730000, China; (Z.Q.); (W.X.)
- Department of Infectious Disease, The First Hospital of Lanzhou University, Lanzhou 730000, China
| | - Junfeng Li
- The First Clinical Medical College of Lanzhou University, Lanzhou 730000, China; (Z.Q.); (W.X.)
- Institute of Infectious Diseases, The First Hospital of Lanzhou University, Lanzhou 730000, China
- Department of Hepatology, The First Hospital of Lanzhou University, Lanzhou 730000, China
| |
Collapse
|