1
|
Zhang Z, Shen Z, Xie S, Li J, Zhang Z, Zhang S, Peng B, Huang Q, Li M, Ma S, Huang Q. Rapamycin exerts neuroprotective effects by inhibiting FKBP12 instead of mTORC1 in the mouse model of Parkinson's disease. Neuropharmacology 2025; 275:110504. [PMID: 40345576 DOI: 10.1016/j.neuropharm.2025.110504] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2024] [Revised: 04/28/2025] [Accepted: 05/05/2025] [Indexed: 05/11/2025]
Abstract
Parkinson's disease (PD), characterized by the selective loss of nigral dopaminergic neurons, is a common neurodegenerative disorder for which effective disease-modifying therapies remain unavailable. Rapamycin, a clinical immunosuppressant used for decades, has demonstrated neuroprotective effects in various animal models of neurological diseases, including PD. These effects are believed to be mediated through the inhibition of mammalian target of rapamycin (mTOR) complex 1 (mTORC1) signaling, with rapamycin binding to FKBP12. However, recent studies have suggested that mTOR activation can be neuroprotective in degenerating dopaminergic neurons, presenting a paradox to the neuroprotective mechanism of rapamycin via mTORC1 inhibition. In this study, we showed that mTORC1 signaling was inactivated in nigral dopaminergic neurons in the 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) mouse model of PD. Notably, the optimal neuroprotective dose of rapamycin did not inhibit mTORC1 signaling nor restore autophagy defects in nigral dopaminergic neurons of MPTP-treated male C57BL/6 mice. Furthermore, acute Raptor knockout in dopaminergic neurons, which abolishes mTORC1 activity, did not diminish rapamycin's neuroprotective effects, suggesting that its protection is independent of mTORC1 inhibition. Importantly, rapamycin is also a potent inhibitor of FKBP12, a peptidyl-prolyl cis-trans isomerase highly expressed in the brain. Selective knockdown of FKBP12 in nigral dopaminergic neurons confers neuroprotective effects comparable to that of rapamycin, with no synergism observed when the two are combined. Collectively, our results indicate that rapamycin exerts neuroprotective effects in parkinsonian mice through inhibition of FKBP12 rather than mTORC1 signaling. These findings suggest that FKBP12 may serve as a novel target for disease-modifying therapies in PD.
Collapse
Affiliation(s)
- Zeyan Zhang
- Guangdong Provincial Key Laboratory of Brain Function and Disease, Department of Pharmacology, Zhongshan School of Medicine, Sun Yat-sen University, No. 74 Zhongshan 2nd Road, Guangzhou, 510080, China
| | - Ziyue Shen
- Guangdong Provincial Key Laboratory of Brain Function and Disease, Department of Pharmacology, Zhongshan School of Medicine, Sun Yat-sen University, No. 74 Zhongshan 2nd Road, Guangzhou, 510080, China
| | - Shiming Xie
- Guangdong Provincial Key Laboratory of Brain Function and Disease, Department of Pharmacology, Zhongshan School of Medicine, Sun Yat-sen University, No. 74 Zhongshan 2nd Road, Guangzhou, 510080, China
| | - Junyu Li
- Guangdong Provincial Key Laboratory of Brain Function and Disease, Department of Pharmacology, Zhongshan School of Medicine, Sun Yat-sen University, No. 74 Zhongshan 2nd Road, Guangzhou, 510080, China
| | - Zeyu Zhang
- Guangdong Provincial Key Laboratory of Brain Function and Disease, Department of Pharmacology, Zhongshan School of Medicine, Sun Yat-sen University, No. 74 Zhongshan 2nd Road, Guangzhou, 510080, China
| | - Sheng Zhang
- Department of Cell Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Bo Peng
- Institute for Translational Brain Research, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, MOE Innovative Center for New Drug Development of Immune Inflammatory Diseases, Fudan University, Shanghai, 200032, China
| | - Qianchu Huang
- Department of Psychology, University of Wisconsin-Madison, Madison, WI, 53706, USA
| | - Mingtao Li
- Guangdong Provincial Key Laboratory of Brain Function and Disease, Department of Pharmacology, Zhongshan School of Medicine, Sun Yat-sen University, No. 74 Zhongshan 2nd Road, Guangzhou, 510080, China
| | - Shanshan Ma
- Guangdong Provincial Key Laboratory of Brain Function and Disease, Department of Pharmacology, Zhongshan School of Medicine, Sun Yat-sen University, No. 74 Zhongshan 2nd Road, Guangzhou, 510080, China.
| | - Qiaoying Huang
- Guangdong Provincial Key Laboratory of Brain Function and Disease, Department of Pharmacology, Zhongshan School of Medicine, Sun Yat-sen University, No. 74 Zhongshan 2nd Road, Guangzhou, 510080, China.
| |
Collapse
|
2
|
Shao KY, Luo SD, Huang EY, Chang TM, Botcha L, Sehar M, Liu JF, Chuang PK. Acetylshikonin induces cell necroptosis via mediating mitochondrial function and oxidative stress-regulated signaling in human Oral Cancer cells. Bioorg Chem 2025; 159:108396. [PMID: 40168882 DOI: 10.1016/j.bioorg.2025.108396] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2025] [Revised: 02/23/2025] [Accepted: 03/17/2025] [Indexed: 04/03/2025]
Abstract
Human oral squamous cell carcinoma (OSCC) represents a significant global health challenge, with conventional treatments showing limited efficacy in improving patient survival rates. To investigate the therapeutic potential of acetylshikonin on OSCC, we conducted comprehensive analyses including cell viability assays, flow cytometry, and molecular pathway investigations. Our findings demonstrate that acetylshikonin significantly inhibits OSCC cell proliferation with IC50 values of 3.81 μM and 5.87 μM in HSC3 and SCC4 cells respectively. Flow cytometry analysis revealed that acetylshikonin treatment significantly increased reactive oxygen species (ROS) production and decreased mitochondrial membrane potential in OSCC cells. Additionally, Western blot analysis showed enhanced phosphorylation of RIPK1, RIPK3, and MLKL proteins, indicating activation of the necroptotic pathway. The critical role of necroptosis was further confirmed using specific inhibitors (GSK872, Necrostatin-1, and 7-CL-O Nec-1), which significantly attenuated acetylshikonin-induced cell death. Transmission electron microscopy revealed distinct ultrastructural changes in cellular organelles, while decreased GPX4 expression suggested potential cross-activation of ferroptotic pathways. These data demonstrate that acetylshikonin suppresses OSCC growth through selective activation of oxidative stress-mediated necroptosis and mitochondrial dysfunction, identifying it as a promising natural compound for OSCC therapy through its ability to activate alternative cell death pathways and overcome traditional therapy limitations.
Collapse
Affiliation(s)
- Kung-Yu Shao
- Oral-Maxillofacial Surgery Division, Department of Dentistry, Shuang Ho Hospital, Ministry of Health and Welfare, New Taipei City, Taiwan
| | - Sheng-Dean Luo
- Department of Otolaryngology, Kaohsiung Chang Gung Memorial Hospital, Taiwan; School of Traditional Chinese Medicine, Chang Gung University College of Medicine, Taoyuan 33302, Taiwan; Graduate Institute of Clinical Medical Sciences, College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan; School of Medicine, College of Medicine, National SunYat-sen University, Kaohsiung, Taiwan
| | - Eng-Yen Huang
- Department of Otolaryngology, Kaohsiung Chang Gung Memorial Hospital, Taiwan; Department of Radiation Oncology & Proton and Radiation Therapy Center, Kaohsiung Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Kaohsiung City, Taiwan; Department of Radiation Oncology, Kaohsiung Chang Gung Memorial Hospital, School of Medicine, College of Medicine, National Sun Yat-sen University, Kaohsiung City, Taiwan
| | - Tsung-Ming Chang
- School of Dental Technology, College of Oral Medicine, Taipei Medical University, Taipei, Taiwan
| | - Lavanya Botcha
- Institute of Biomedical Sciences, College of Medicine, National Sun Yat-sen University, Kaohsiung, Taiwan
| | - Misbah Sehar
- Institute of Biomedical Sciences, College of Medicine, National Sun Yat-sen University, Kaohsiung, Taiwan
| | - Ju-Fang Liu
- School of Oral Hygiene, College of Oral Medicine, Taipei Medical University, Taipei, Taiwan.
| | - Po-Kai Chuang
- Institute of Biomedical Sciences, College of Medicine, National Sun Yat-sen University, Kaohsiung, Taiwan.
| |
Collapse
|
3
|
Ma K, Huang J, Zhang J, Tian Y, Hu J, Ma L, Wang C. Hydrogen Sulfide (H 2S) Mitigates Sepsis-Induced Adrenal Dysfunction via Inhibition of TNFα-Mediated Necroptosis. Pathogens 2025; 14:439. [PMID: 40430756 PMCID: PMC12113818 DOI: 10.3390/pathogens14050439] [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: 03/14/2025] [Revised: 04/10/2025] [Accepted: 04/18/2025] [Indexed: 05/29/2025] Open
Abstract
BACKGROUND Sepsis is a life-threatening condition that is characterized by systemic inflammation and organ dysfunction, with adrenal dysfunction being a significant complication. This study aimed to investigate the role of necroptosis and hydrogen sulfide (H2S) in sepsis-induced adrenal dysfunction. METHODS A cecal ligation and puncture (CLP)-induced sepsis mouse model was employed. Adrenocortical-specific mixed lineage kinase domain-like pseudokinase (MLKL) knockout (MLKL-KO) and cystathioneine β-synthase (CBS) knockout (CBS-KO) mice were generated using Cre-loxP technology and adrenocortical-specific Cre tool mice. In vitro experiments utilized TNFα-stimulated Y1 adrenocortical cells. The treatments included the H2S donor NaHS, TNFα inhibitor R-7050, necroptosis inhibitor NSA and CBS inhibitor AOAA. Pathological assessment involved hematoxylin-eosin (H&E) staining and a Western blot analysis of necroptosis markers (the phosphorylation of MLKL (p-MLKL) and phosphorylation of receptor-interacting protein kinases 1 (p-RIPK1)). RESULTS Sepsis induced adrenal congestion, elevated TNFα levels, and activated necroptosis (increased p-MLKL/p-RIPK1) in wild-type mice. H2S treatment attenuated adrenal damage, reduced TNFα, and suppressed necroptosis. MLKL knockout reduced septic adrenal dysfunction, whereas CBS knockout exacerbated septic adrenal dysfunction. In vitro, TNFα induced Y1 cell necroptosis, which was reversed by H2S or NSA. AOAA exacerbated TNFα-induced necroptosis in Y1 cells. CONCLUSIONS H2S inhibits TNFα-mediated necroptosis, thereby preserving adrenal integrity in sepsis. Targeting the TNFα-necroptosis axis and enhancing endogenous H2S production may represent novel therapeutic strategies for sepsis-associated adrenal dysfunction.
Collapse
Affiliation(s)
- Kai Ma
- Lab of Stress Injury, School of Life Sciences, Shanghai University, Shanghai 200444, China; (K.M.); (J.H.); (Y.T.)
| | - Jingwen Huang
- Lab of Stress Injury, School of Life Sciences, Shanghai University, Shanghai 200444, China; (K.M.); (J.H.); (Y.T.)
| | - Jin Zhang
- General Practice Department, Shanghai Pudong New District Kangqiao Community Health Service Center, Shanghai 201315, China;
| | - Yuan Tian
- Lab of Stress Injury, School of Life Sciences, Shanghai University, Shanghai 200444, China; (K.M.); (J.H.); (Y.T.)
| | - Jing Hu
- Lab of Stress Injury, School of Life Sciences, Shanghai University, Shanghai 200444, China; (K.M.); (J.H.); (Y.T.)
| | - Linhao Ma
- Department of Emergency Medicine, Shanghai Fourth People’s Hospital, School of Medicine, Tongji University, Shanghai 200081, China
| | - Changnan Wang
- Lab of Stress Injury, School of Life Sciences, Shanghai University, Shanghai 200444, China; (K.M.); (J.H.); (Y.T.)
| |
Collapse
|
4
|
Xiao H, Han Z, Xu M, Gao X, Qiu S, Ren N, Yi Y, Zhou C. The Role of Post-Translational Modifications in Necroptosis. Biomolecules 2025; 15:549. [PMID: 40305291 PMCID: PMC12024652 DOI: 10.3390/biom15040549] [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: 01/28/2025] [Revised: 03/05/2025] [Accepted: 03/06/2025] [Indexed: 05/02/2025] Open
Abstract
Necroptosis, a distinct form of regulated necrosis implicated in various human pathologies, is orchestrated through sophisticated signaling pathways. During this process, cells undergoing necroptosis exhibit characteristic necrotic morphology and provoke substantial inflammatory responses. Post-translational modifications (PTMs)-chemical alterations occurring after protein synthesis that critically regulate protein functionality-constitute essential regulatory components within these complex signaling cascades. This intricate crosstalk between necroptotic pathways and PTM networks presents promising therapeutic opportunities. Our comprehensive review systematically analyzes the molecular mechanisms underlying necroptosis, with particular emphasis on the regulatory roles of PTMs in signal transduction. Through systematic evaluation of key modifications including ubiquitination, phosphorylation, glycosylation, methylation, acetylation, disulfide bond formation, caspase cleavage, nitrosylation, and SUMOylation, we examine potential therapeutic applications targeting necroptosis in disease pathogenesis. Furthermore, we synthesize current pharmacological strategies for manipulating PTM-regulated necroptosis, offering novel perspectives on clinical target development and therapeutic intervention.
Collapse
Affiliation(s)
- Hao Xiao
- Department of Liver Surgery and Transplantation, Zhongshan Hospital, Fudan University, Shanghai 200032, China; (H.X.); (Z.H.)
- Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai 200032, China
- Key Laboratory of Carcinogenesis and Cancer Invasion, Fudan University, Ministry of Education, Shanghai 200032, China
| | - Zeping Han
- Department of Liver Surgery and Transplantation, Zhongshan Hospital, Fudan University, Shanghai 200032, China; (H.X.); (Z.H.)
- Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai 200032, China
- Key Laboratory of Carcinogenesis and Cancer Invasion, Fudan University, Ministry of Education, Shanghai 200032, China
| | - Min Xu
- Department of Liver Surgery and Transplantation, Zhongshan Hospital, Fudan University, Shanghai 200032, China; (H.X.); (Z.H.)
- Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai 200032, China
- Key Laboratory of Carcinogenesis and Cancer Invasion, Fudan University, Ministry of Education, Shanghai 200032, China
| | - Xukang Gao
- Department of Liver Surgery and Transplantation, Zhongshan Hospital, Fudan University, Shanghai 200032, China; (H.X.); (Z.H.)
- Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai 200032, China
- Key Laboratory of Carcinogenesis and Cancer Invasion, Fudan University, Ministry of Education, Shanghai 200032, China
| | - Shuangjian Qiu
- Department of Liver Surgery and Transplantation, Zhongshan Hospital, Fudan University, Shanghai 200032, China; (H.X.); (Z.H.)
- Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai 200032, China
- Key Laboratory of Carcinogenesis and Cancer Invasion, Fudan University, Ministry of Education, Shanghai 200032, China
| | - Ning Ren
- Department of Liver Surgery and Transplantation, Zhongshan Hospital, Fudan University, Shanghai 200032, China; (H.X.); (Z.H.)
- Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai 200032, China
- Key Laboratory of Carcinogenesis and Cancer Invasion, Fudan University, Ministry of Education, Shanghai 200032, China
| | - Yong Yi
- Department of Liver Surgery and Transplantation, Zhongshan Hospital, Fudan University, Shanghai 200032, China; (H.X.); (Z.H.)
- Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai 200032, China
- Key Laboratory of Carcinogenesis and Cancer Invasion, Fudan University, Ministry of Education, Shanghai 200032, China
| | - Chenhao Zhou
- Department of Liver Surgery and Transplantation, Zhongshan Hospital, Fudan University, Shanghai 200032, China; (H.X.); (Z.H.)
- Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai 200032, China
- Key Laboratory of Carcinogenesis and Cancer Invasion, Fudan University, Ministry of Education, Shanghai 200032, China
| |
Collapse
|
5
|
He J, Huang Z, Zou R. Andrographolide ameliorates sepsis-induced acute liver injury by attenuating endoplasmic reticulum stress through the FKBP1A-mediated NOTCH1/AK2 pathway. Cell Biol Toxicol 2025; 41:56. [PMID: 40053226 PMCID: PMC11889056 DOI: 10.1007/s10565-025-10007-9] [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: 11/18/2024] [Accepted: 02/20/2025] [Indexed: 03/10/2025]
Abstract
Andrographolide (AP) has been shown to possess anti-inflammatory activities. In this study, the impact of AP in sepsis-induced acute liver injury (ALI) and the molecules involved were dissected. FKBP1A was predicted to be the sole target protein of AP that was also differentially expressed in the GSE166868 dataset. AP induced the protein expression of FKBP1A and suppressed that of NOTCH1 in a dose-dependent manner. AP ameliorated ALI in mice induced by D-galactosamine and LPS and inhibited LPS-induced liver parenchymal cell injury in vitro. By contrast, the protective effect of AP was significantly lost after the knockdown of FKBP1A. As a positive control, the therapeutic effect of dexamethasone on ALI may be related to NOTCH1, which was not related to FKBP1A. NOTCH1 promoted AK2 transcription in liver parenchymal cells, and FKBP1A inhibited endoplasmic reticulum (ER) stress by impairing NOTCH1/AK2 signaling. Restoration of NOTCH1 significantly reversed the hepatoprotective effect of AP in ALI mice and LPS-induced liver parenchymal cell injury by activating the ER stress pathway. Therefore, AP-promoted FKBP1A expression inhibits ALI progression by blocking the NOTCH1/AK2-mediated ER pathway.
Collapse
Affiliation(s)
- Jiaqi He
- Department of General Surgery, Huadong Hospital Affiliated to Fudan University, Shanghai, 200040, P.R. China
| | - Zepeng Huang
- Department of Hepatobiliary Surgery, Hainan Cancer Hospital, Haikou, 570000, Hainan, P.R. China
| | - Rui Zou
- Department of Hepatobiliary Surgery, Hainan Cancer Hospital, Haikou, 570000, Hainan, P.R. China.
- Hainan Clinical Research Center for Liver Disease and Liver Critical Illness, Haikou, 570000, Hainan, P.R. China.
| |
Collapse
|
6
|
Eskander G, Abdelhamid SG, Wahdan SA, Radwan SM. Insights on the crosstalk among different cell death mechanisms. Cell Death Discov 2025; 11:56. [PMID: 39929794 PMCID: PMC11811070 DOI: 10.1038/s41420-025-02328-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Revised: 12/23/2024] [Accepted: 01/27/2025] [Indexed: 02/13/2025] Open
Abstract
The phenomenon of cell death has garnered significant scientific attention in recent years, emerging as a pivotal area of research. Recently, novel modalities of cellular death and the intricate interplay between them have been unveiled, offering insights into the pathogenesis of various diseases. This comprehensive review delves into the intricate molecular mechanisms, inducers, and inhibitors of the underlying prevalent forms of cell death, including apoptosis, autophagy, ferroptosis, necroptosis, mitophagy, and pyroptosis. Moreover, it elucidates the crosstalk and interconnection among the key pathways or molecular entities associated with these pathways, thereby paving the way for the identification of novel therapeutic targets, disease management strategies, and drug repurposing.
Collapse
Affiliation(s)
- Georgette Eskander
- Postgraduate program, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt
| | | | - Sara A Wahdan
- Pharmacology and toxicology Department, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt
| | - Sara M Radwan
- Biochemistry Department, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt.
| |
Collapse
|
7
|
He X, Jiang X, Guo J, Sun H, Yang J. PANoptosis in Bacterial Infections: A Double-Edged Sword Balancing Host Immunity and Pathogenesis. Pathogens 2025; 14:43. [PMID: 39861004 PMCID: PMC11768250 DOI: 10.3390/pathogens14010043] [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: 12/10/2024] [Revised: 12/29/2024] [Accepted: 01/03/2025] [Indexed: 01/27/2025] Open
Abstract
PANoptosis is a newly identified programmed cell death pathway that integrates characteristics of apoptosis, pyroptosis, and necroptosis. It plays a dual role in the host immune response to bacterial infections. On one hand, PANoptosis acts as a protective mechanism by inducing the death of infected cells to eliminate pathogens and releasing pro-inflammatory cytokines to amplify the immune response. On the other hand, bacteria can exploit PANoptosis to evade host immune defenses. This dual nature underscores the potential of PANoptosis as a target for developing novel therapies against bacterial infections. This review summarizes the molecular mechanisms of PANoptosis, along with the crosstalk and integration of different cell death pathways in response to various bacterial pathogens. We also discuss the dual roles of PANoptosis in bacterial infectious diseases, including sepsis, pulmonary infections, and intestinal infections. Elucidating the molecular mechanisms underlying PANoptosis and how bacteria manipulate this pathway offers critical insights into host-pathogen interactions. These insights provide a foundation for designing targeted antibacterial strategies, modulating inflammation, and advancing precision medicine to improve clinical outcomes.
Collapse
Affiliation(s)
| | | | | | | | - Jing Yang
- Cuiying Biomedical Research Center, The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou 730030, China; (X.H.); (X.J.); (J.G.); (H.S.)
| |
Collapse
|
8
|
Xu Q, Gong H, Zhou M, Guo J, Chen S, Xiao K, Liu Y. Necroptosis contributes to deoxynivalenol-induced liver injury and inflammation in weaned piglets. J Anim Sci Biotechnol 2024; 15:160. [PMID: 39623511 PMCID: PMC11613918 DOI: 10.1186/s40104-024-01117-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2024] [Accepted: 10/21/2024] [Indexed: 12/06/2024] Open
Abstract
BACKGROUND The aim of this study was to investigate the role of necroptosis in deoxynivalenol (DON)-induced liver injury and inflammation in weaned piglets. METHODS In Exp. 1, 12 weaned piglets were divided into 2 groups including pigs fed basal diet and pigs fed diet contaminated with 4 mg/kg DON for 21 d. In Exp. 2, 12 weaned piglets were divided into 2 groups including control piglets and piglets given a gavage of 2 mg/kg body weight (BW) DON. In Exp. 3, 24 weaned piglets were used in a 2 × 2 factorial design and the main factors including necrostatin-1 (Nec-1) (DMSO or 0.5 mg/kg BW Nec-1) and DON challenge (saline or 2 mg/kg BW DON gavage). On 21 d in Exp. 1, or at 6 h post DON gavage in Exp. 2 and 3, pigs were killed for blood samples and liver tissues. Liver histology, blood biochemical indicators, and liver inflammation and necroptosis signals were tested. RESULTS Dietary or oral gavage with DON caused liver morphological damage in piglets. Dietary DON led to hepatocyte damage indicated by increased aspartate transaminase (AST) activity and AST/alanine aminotransferase (ALT) ratio, and DON gavage also caused hepatocyte damage and cholestasis indicated by increased AST and alkaline phosphatase (AKP) activities. Dietary DON caused liver necroptosis indicated by increased protein abundance of total receptor interacting protein kinase 3 (t-RIP3) and total mixed lineage kinase domain-like protein (t-MLKL). Moreover, DON gavage increased mRNA expression of interleukin (IL)-6 and IL-1β in liver. DON gavage also induced liver necroptosis demonstrated by increased protein abundance of t-RIP3, phosphorylated-RIP3 (p-RIP3), t-MLKL and p-MLKL. However, pretreatment with Nec-1, a specific inhibitor of necroptosis, inhibited liver necroptosis indicated by decreased protein expression of t-RIP3, p-RIP3, t-MLKL and p-MLKL. Nec-1 pretreatment reduced liver morphological damage after DON gavage. Pretreatment with Nec-1 also attenuated liver damage induced by DON indicated by decreased activities of AST and AKP. Furthermore, Nec-1 pretreatment inhibited liver mRNA expression of IL-6 and IL-1β after DON challenge. CONCLUSIONS Our data demonstrate for the first time that necroptosis contributes to DON-induced liver injury and inflammation in piglets.
Collapse
Affiliation(s)
- Qilong Xu
- Hubei Key Laboratory of Animal Nutrition and Feed Science, Wuhan Polytechnic University, Wuhan, 430023, China
| | - Hanqiu Gong
- Hubei Key Laboratory of Animal Nutrition and Feed Science, Wuhan Polytechnic University, Wuhan, 430023, China
| | - Mohan Zhou
- Hubei Key Laboratory of Animal Nutrition and Feed Science, Wuhan Polytechnic University, Wuhan, 430023, China
| | - Junjie Guo
- Hubei Key Laboratory of Animal Nutrition and Feed Science, Wuhan Polytechnic University, Wuhan, 430023, China
| | - Shaokui Chen
- Hubei Key Laboratory of Animal Nutrition and Feed Science, Wuhan Polytechnic University, Wuhan, 430023, China
| | - Kan Xiao
- Hubei Key Laboratory of Animal Nutrition and Feed Science, Wuhan Polytechnic University, Wuhan, 430023, China.
| | - Yulan Liu
- Hubei Key Laboratory of Animal Nutrition and Feed Science, Wuhan Polytechnic University, Wuhan, 430023, China.
| |
Collapse
|
9
|
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
|
10
|
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
|
11
|
Baral B, Saini V, Kandpal M, Kundu P, Dixit AK, Parmar HS, Meena AK, Trivedi P, Jha HC. The interplay of co-infections in shaping COVID-19 severity: Expanding the scope beyond SARS-CoV-2. J Infect Public Health 2024; 17:102486. [PMID: 39002466 DOI: 10.1016/j.jiph.2024.102486] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2023] [Revised: 06/17/2024] [Accepted: 06/23/2024] [Indexed: 07/15/2024] Open
Abstract
High mortality has been reported in severe cases of COVID-19. Emerging reports suggested that the severity is not only due to SARS-CoV-2 infection, but also due to coinfections by other pathogens exhibiting symptoms like COVID-19. During the COVID-19 pandemic, simultaneous respiratory coinfections with various viral (Retroviridae, Flaviviridae, Orthomyxoviridae, and Picoviridae) and bacterial (Mycobacteriaceae, Mycoplasmataceae, Enterobacteriaceae and Helicobacteraceae) families have been observed. These pathogens intensify disease severity by potentially augmenting SARSCoV-2 replication, inflammation, and modulation of signaling pathways. Coinfection emerges as a critical determinant of COVID-19 severity, principally instigated by heightened pro-inflammatory cytokine levels, as cytokine storm. Thereby, in co-infection scenario, the severity is also driven by the modulation of inflammatory signaling pathways by both pathogens possibly associated with interleukin, interferon, and cell death exacerbating the severity. In the current review, we attempt to understand the role of co- infections by other pathogens and their involvement in the severity of COVID-19.
Collapse
Affiliation(s)
- Budhadev Baral
- Infection Bioengineering Group, Department of Biosciences and Biomedical Engineering, Indian Institute of Technology Indore, Simrol, Indore 453552, Madhya Pradesh, India
| | - Vaishali Saini
- Infection Bioengineering Group, Department of Biosciences and Biomedical Engineering, Indian Institute of Technology Indore, Simrol, Indore 453552, Madhya Pradesh, India
| | - Meenakshi Kandpal
- Infection Bioengineering Group, Department of Biosciences and Biomedical Engineering, Indian Institute of Technology Indore, Simrol, Indore 453552, Madhya Pradesh, India
| | - Pratik Kundu
- Infection Bioengineering Group, Department of Biosciences and Biomedical Engineering, Indian Institute of Technology Indore, Simrol, Indore 453552, Madhya Pradesh, India
| | - Amit Kumar Dixit
- Central Ayurveda Research Institute, 4-CN Block, Sector -V, Bidhannagar, Kolkata 700 091, India
| | - Hamendra Singh Parmar
- School of Biotechnology, Devi Ahilya Vishwavidyalaya, Takshashila Campus, Indore, Madhya Pradesh 452001, India
| | - Ajay Kumar Meena
- Regional Ayurveda Research Institute, Gwalior, Amkhoh, Gwalior, Madhya Pradesh 474001, India
| | - Pankaj Trivedi
- Department of Experimental Medicine, Sapienza University, Rome, Italy
| | - Hem Chandra Jha
- Infection Bioengineering Group, Department of Biosciences and Biomedical Engineering, Indian Institute of Technology Indore, Simrol, Indore 453552, Madhya Pradesh, India; Centre for Rural Development and Technology, Indian Institute of Technology Indore, Simrol, Indore 453552, Madhya Pradesh, India.
| |
Collapse
|
12
|
Khaleque MA, Kim JH, Hwang BJ, Kang JK, Quan M, Kim YY. Role of Necroptosis in Intervertebral Disc Degeneration. Int J Mol Sci 2023; 24:15292. [PMID: 37894970 PMCID: PMC10607531 DOI: 10.3390/ijms242015292] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Revised: 10/06/2023] [Accepted: 10/11/2023] [Indexed: 10/29/2023] Open
Abstract
Apoptosis has historically been considered the primary form of programmed cell death (PCD) and is responsible for regulating cellular processes during development, homeostasis, and disease. Conversely, necrosis was considered uncontrolled and unregulated. However, recent evidence has unveiled the significance of necroptosis, a regulated form of necrosis, as an important mechanism of PCD alongside apoptosis. The activation of necroptosis leads to cellular membrane disruption, inflammation, and vascularization. This process is crucial in various pathological conditions, including intervertebral disc degeneration (IVDD), neurodegeneration, inflammatory diseases, multiple cancers, and kidney injury. In recent years, extensive research efforts have shed light on the molecular regulation of the necroptotic pathway. Various stimuli trigger necroptosis, and its regulation involves the activation of specific proteins such as receptor-interacting protein kinase 1 (RIPK1), RIPK3, and the mixed lineage kinase domain-like (MLKL) pseudokinase. Understanding the intricate mechanisms governing necroptosis holds great promise for developing novel therapeutic interventions targeting necroptosis-associated IVDD. The objective of this review is to contribute to the growing body of scientific knowledge in this area by providing a comprehensive overview of necroptosis and its association with IVDD. Ultimately, these understandings will allow the development of innovative drugs that can modulate the necroptotic pathway, offering new therapeutic avenues for individuals suffering from necroptosis.
Collapse
Affiliation(s)
| | | | | | | | | | - Young-Yul Kim
- Department of Orthopedic Surgery, Daejeon St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Daejeon 34943, Republic of Korea; (M.A.K.); (J.-H.K.); (B.-J.H.); (J.-K.K.); (M.Q.)
| |
Collapse
|
13
|
Park SH, Lee HC, Jeong HM, Lee JS, Cha HJ, Kim CH, Kim J, Song KS. Inhibition of Urban Particulate Matter-Induced Airway Inflammation by RIPK3 through the Regulation of Tight Junction Protein Production. Int J Mol Sci 2023; 24:13320. [PMID: 37686124 PMCID: PMC10487650 DOI: 10.3390/ijms241713320] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Revised: 08/24/2023] [Accepted: 08/24/2023] [Indexed: 09/10/2023] Open
Abstract
Urban particulate matter (UPM) is a high-hazard cause of various diseases in humans, including in the respiratory tract, skin, heart, and even brain. Unfortunately, there is no established treatment for the damage caused by UPM in the respiratory epithelium. In addition, although RIPK3 is known to induce necroptosis, its intracellular role as a negative regulator in human lungs and bronchial epithelia remains unclear. Here, the endogenous expression of RIPK3 was significantly decreased 6 h after exposure to UPM. In RIPK3-ovexpressed cells, RIPK3 was not moved to the cytoplasm from the nucleus. Interestingly, the overexpression of RIPK3 dramatically decreased TEER and F-actin formation. Its overexpression also decreased the expression of genes for pro-inflammatory cytokines (IL-6 and IL-8) and tight junctions (ZO-1, -2, -3, E-cadherin, and claudin) during UPM-induced airway inflammation. Importantly, overexpression of RIPK3 inhibited the UPM-induced ROS production by inhibiting the activation of iNOS and eNOS and by regulating mitochondrial fission processing. In addition, UPM-induced activation of the iκB and NF-κB signaling pathways was dramatically decreased by RIPK3, and the expression of pro-inflammatory cytokines was decreased by inhibiting the iκB signaling pathway. Our data indicated that RIPK3 is essential for the UPM-induced inflammatory microenvironment to maintain homeostasis. Therefore, we suggest that RIPK3 is a potential therapeutic candidate for UPM-induced pulmonary inflammation.
Collapse
Affiliation(s)
- Sun-Hee Park
- Department of Medical Science, Kosin University College of Medicine, Seo-gu, Busan 49267, Republic of Korea; (S.-H.P.); (H.-C.L.); (H.M.J.)
| | - Hyun-Chae Lee
- Department of Medical Science, Kosin University College of Medicine, Seo-gu, Busan 49267, Republic of Korea; (S.-H.P.); (H.-C.L.); (H.M.J.)
| | - Hye Min Jeong
- Department of Medical Science, Kosin University College of Medicine, Seo-gu, Busan 49267, Republic of Korea; (S.-H.P.); (H.-C.L.); (H.M.J.)
| | - Jeong-Sang Lee
- Department of Functional Foods and Biotechnology, College of Medical Sciences, Jeonju University, 303 Cheonjam-ro, Jeonju 55069, Republic of Korea;
| | - Hee-Jae Cha
- Department of Genetics, Kosin University College of Medicine, Seo-gu, Busan 49267, Republic of Korea;
| | - Cheol Hong Kim
- Department of Pediatrics, Myongji Hospital, Hanyang University College of Medicine, Goyang 15588, Republic of Korea;
| | - Jeongtae Kim
- Department of Anatomy, Kosin University College of Medicine, Seo-gu, Busan 49267, Republic of Korea;
| | - Kyoung Seob Song
- Department of Medical Science, Kosin University College of Medicine, Seo-gu, Busan 49267, Republic of Korea; (S.-H.P.); (H.-C.L.); (H.M.J.)
| |
Collapse
|
14
|
Yu H, Chen L, Du B. Necroptosis in the pathophysiology of preeclampsia. Cell Cycle 2023; 22:1713-1725. [PMID: 37365800 PMCID: PMC10446795 DOI: 10.1080/15384101.2023.2229138] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Revised: 03/30/2023] [Accepted: 06/19/2023] [Indexed: 06/28/2023] Open
Abstract
Necroptosis is a newly-identified form of gene-regulated cell necrosis that is increasingly considered to be a pathway associated with human pathophysiological conditions. Cells undergoing necroptosis exhibit necrotic phenotypes, including disruption of the plasma membrane integrity, organelle swelling, and cytolysis. Accumulating evidence suggests that trophoblast necroptosis plays a complex role in preeclampsia (PE). However, the exact pathogenesis remains unclear. Its unique mechanisms of action in various diseases are expected to provide prospects for the treatment of PE. Therefore, it is necessary to further explore its molecular mechanism in PE in order to identify potential therapeutic options. This review examines the current knowledge regarding the role and mechanisms of necroptosis in PE and provides a theoretical basis for new therapeutic targets for PE.
Collapse
Affiliation(s)
- Hongbiao Yu
- Department of Obstetrics and Gynecology, the Second Clinical College of North Sichuan Medical College, Nanchong Central Hospital, Nanchong, Sichuan, China
| | - Ling Chen
- Department of Oncology, the Second Clinical College of North Sichuan Medical College, Nanchong Central Hospital, Nanchong, Sichuan, China
| | - Boyu Du
- Department of Obstetrics and Gynecology, the Second Clinical College of North Sichuan Medical College, Nanchong Central Hospital, Nanchong, Sichuan, China
| |
Collapse
|
15
|
Shiau JP, Chuang YT, Tang JY, Yang KH, Chang FR, Hou MF, Yen CY, Chang HW. The Impact of Oxidative Stress and AKT Pathway on Cancer Cell Functions and Its Application to Natural Products. Antioxidants (Basel) 2022; 11:1845. [PMID: 36139919 PMCID: PMC9495789 DOI: 10.3390/antiox11091845] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Revised: 09/14/2022] [Accepted: 09/16/2022] [Indexed: 01/10/2023] Open
Abstract
Oxidative stress and AKT serine-threonine kinase (AKT) are responsible for regulating several cell functions of cancer cells. Several natural products modulate both oxidative stress and AKT for anticancer effects. However, the impact of natural product-modulating oxidative stress and AKT on cell functions lacks systemic understanding. Notably, the contribution of regulating cell functions by AKT downstream effectors is not yet well integrated. This review explores the role of oxidative stress and AKT pathway (AKT/AKT effectors) on ten cell functions, including apoptosis, autophagy, endoplasmic reticulum stress, mitochondrial morphogenesis, ferroptosis, necroptosis, DNA damage response, senescence, migration, and cell-cycle progression. The impact of oxidative stress and AKT are connected to these cell functions through cell function mediators. Moreover, the AKT effectors related to cell functions are integrated. Based on this rationale, natural products with the modulating abilities for oxidative stress and AKT pathway exhibit the potential to regulate these cell functions, but some were rarely reported, particularly for AKT effectors. This review sheds light on understanding the roles of oxidative stress and AKT pathway in regulating cell functions, providing future directions for natural products in cancer treatment.
Collapse
Affiliation(s)
- Jun-Ping Shiau
- Division of Breast Oncology and Surgery, Department of Surgery, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 80708, Taiwan or
| | - Ya-Ting Chuang
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
| | - Jen-Yang Tang
- School of Post-Baccalaureate Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
- Department of Radiation Oncology, Kaohsiung Medical University Hospital, Kaoshiung Medical University, Kaohsiung 80708, Taiwan
| | - Kun-Han Yang
- Graduate Institute of Natural Products, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
| | - Fang-Rong Chang
- Graduate Institute of Natural Products, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
| | - Ming-Feng Hou
- Division of Breast Oncology and Surgery, Department of Surgery, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 80708, Taiwan or
- Department of Biomedical Science and Environmental Biology, College of Life Science, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
| | - Ching-Yu Yen
- Department of Oral and Maxillofacial Surgery, Chi-Mei Medical Center, Tainan 71004, Taiwan
- School of Dentistry, Taipei Medical University, Taipei 11031, Taiwan
| | - Hsueh-Wei Chang
- Department of Biomedical Science and Environmental Biology, College of Life Science, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
- Center for Cancer Research, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
| |
Collapse
|
16
|
Liu X, Xie X, Ren Y, Shao Z, Zhang N, Li L, Ding X, Zhang L. The role of necroptosis in disease and treatment. MedComm (Beijing) 2021; 2:730-755. [PMID: 34977874 PMCID: PMC8706757 DOI: 10.1002/mco2.108] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2021] [Revised: 11/23/2021] [Accepted: 11/24/2021] [Indexed: 12/11/2022] Open
Abstract
Necroptosis, a distinctive type of programmed cell death different from apoptosis or necrosis, triggered by a series of death receptors such as tumor necrosis factor receptor 1 (TNFR1), TNFR2, and Fas. In case that apoptosis process is blocked, necroptosis pathway is initiated with the activation of three key downstream mediators which are receptor-interacting serine/threonine protein kinase 1 (RIPK1), RIPK3, and mixed lineage kinase domain-like protein (MLKL). The whole process eventually leads to destruction of the cell membrane integrity, swelling of organelles, and severe inflammation. Over the past decade, necroptosis has been found widely involved in life process of human beings and animals. In this review, we attempt to explore the therapeutic prospects of necroptosis regulators by describing its molecular mechanism and the role it played in pathological condition and tissue homeostasis, and to summarize the research and clinical applications of corresponding regulators including small molecule inhibitors, chemicals, Chinese herbal extracts, and biological agents in the treatment of various diseases.
Collapse
Affiliation(s)
- Xiaoxiao Liu
- Department of Radiation OncologyAffiliated Hospital of Xuzhou Medical UniversityXuzhou Medical UniversityXuzhouJiangsu ProvinceP. R. China
- Jiangsu Center for the Collaboration and Innovation of Cancer BiotherapyCancer InstituteXuzhou Medical UniversityXuzhouJiangsu ProvinceP. R. China
| | - Xin Xie
- Department of Radiation OncologyAffiliated Hospital of Xuzhou Medical UniversityXuzhou Medical UniversityXuzhouJiangsu ProvinceP. R. China
- Jiangsu Center for the Collaboration and Innovation of Cancer BiotherapyCancer InstituteXuzhou Medical UniversityXuzhouJiangsu ProvinceP. R. China
| | - Yuanyuan Ren
- Department of Radiation OncologyAffiliated Hospital of Xuzhou Medical UniversityXuzhou Medical UniversityXuzhouJiangsu ProvinceP. R. China
- Jiangsu Center for the Collaboration and Innovation of Cancer BiotherapyCancer InstituteXuzhou Medical UniversityXuzhouJiangsu ProvinceP. R. China
| | - Zhiying Shao
- Department of Radiation OncologyAffiliated Hospital of Xuzhou Medical UniversityXuzhou Medical UniversityXuzhouJiangsu ProvinceP. R. China
- Jiangsu Center for the Collaboration and Innovation of Cancer BiotherapyCancer InstituteXuzhou Medical UniversityXuzhouJiangsu ProvinceP. R. China
- Cancer InstituteXuzhou Medical UniversityXuzhouJiangsu ProvinceP. R. China
| | - Nie Zhang
- Department of Radiation OncologyAffiliated Hospital of Xuzhou Medical UniversityXuzhou Medical UniversityXuzhouJiangsu ProvinceP. R. China
- Jiangsu Center for the Collaboration and Innovation of Cancer BiotherapyCancer InstituteXuzhou Medical UniversityXuzhouJiangsu ProvinceP. R. China
| | - Liantao Li
- Department of Radiation OncologyAffiliated Hospital of Xuzhou Medical UniversityXuzhou Medical UniversityXuzhouJiangsu ProvinceP. R. China
- Jiangsu Center for the Collaboration and Innovation of Cancer BiotherapyCancer InstituteXuzhou Medical UniversityXuzhouJiangsu ProvinceP. R. China
| | - Xin Ding
- Department of Radiation OncologyAffiliated Hospital of Xuzhou Medical UniversityXuzhou Medical UniversityXuzhouJiangsu ProvinceP. R. China
- Jiangsu Center for the Collaboration and Innovation of Cancer BiotherapyCancer InstituteXuzhou Medical UniversityXuzhouJiangsu ProvinceP. R. China
| | - Longzhen Zhang
- Department of Radiation OncologyAffiliated Hospital of Xuzhou Medical UniversityXuzhou Medical UniversityXuzhouJiangsu ProvinceP. R. China
- Jiangsu Center for the Collaboration and Innovation of Cancer BiotherapyCancer InstituteXuzhou Medical UniversityXuzhouJiangsu ProvinceP. R. China
| |
Collapse
|
17
|
Manes NP, Nita-Lazar A. Molecular Mechanisms of the Toll-Like Receptor, STING, MAVS, Inflammasome, and Interferon Pathways. mSystems 2021; 6:e0033621. [PMID: 34184910 PMCID: PMC8269223 DOI: 10.1128/msystems.00336-21] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Pattern recognition receptors (PRRs) form the front line of defense against pathogens. Many of the molecular mechanisms that facilitate PRR signaling have been characterized in detail, which is critical for the development of accurate PRR pathway models at the molecular interaction level. These models could support the development of therapeutics for numerous diseases, including sepsis and COVID-19. This review describes the molecular mechanisms of the principal signaling interactions of the Toll-like receptor, STING, MAVS, and inflammasome pathways. A detailed molecular mechanism network is included as Data Set S1 in the supplemental material.
Collapse
Affiliation(s)
- Nathan P. Manes
- Functional Cellular Networks Section, Laboratory of Immune System Biology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Aleksandra Nita-Lazar
- Functional Cellular Networks Section, Laboratory of Immune System Biology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| |
Collapse
|
18
|
Guo W, Vandoorne T, Steyaert J, Staats KA, Van Den Bosch L. The multifaceted role of kinases in amyotrophic lateral sclerosis: genetic, pathological and therapeutic implications. Brain 2021; 143:1651-1673. [PMID: 32206784 PMCID: PMC7296858 DOI: 10.1093/brain/awaa022] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2019] [Revised: 11/23/2019] [Accepted: 12/11/2019] [Indexed: 02/07/2023] Open
Abstract
Amyotrophic lateral sclerosis is the most common degenerative disorder of motor neurons in adults. As there is no cure, thousands of individuals who are alive at present will succumb to the disease. In recent years, numerous causative genes and risk factors for amyotrophic lateral sclerosis have been identified. Several of the recently identified genes encode kinases. In addition, the hypothesis that (de)phosphorylation processes drive the disease process resulting in selective motor neuron degeneration in different disease variants has been postulated. We re-evaluate the evidence for this hypothesis based on recent findings and discuss the multiple roles of kinases in amyotrophic lateral sclerosis pathogenesis. We propose that kinases could represent promising therapeutic targets. Mainly due to the comprehensive regulation of kinases, however, a better understanding of the disturbances in the kinome network in amyotrophic lateral sclerosis is needed to properly target specific kinases in the clinic.
Collapse
Affiliation(s)
- Wenting Guo
- KU Leuven-University of Leuven, Department of Neurosciences, Experimental Neurology, and Leuven Brain Institute (LBI), Leuven, Belgium.,VIB, Center for Brain & Disease Research, Laboratory of Neurobiology, Leuven, Belgium.,KU Leuven-Stem Cell Institute (SCIL), Leuven, Belgium
| | - Tijs Vandoorne
- KU Leuven-University of Leuven, Department of Neurosciences, Experimental Neurology, and Leuven Brain Institute (LBI), Leuven, Belgium.,VIB, Center for Brain & Disease Research, Laboratory of Neurobiology, Leuven, Belgium
| | - Jolien Steyaert
- KU Leuven-University of Leuven, Department of Neurosciences, Experimental Neurology, and Leuven Brain Institute (LBI), Leuven, Belgium.,VIB, Center for Brain & Disease Research, Laboratory of Neurobiology, Leuven, Belgium
| | - Kim A Staats
- Department of Stem Cell Biology and Regenerative Medicine, University of Southern California, Los Angeles, California, USA
| | - Ludo Van Den Bosch
- KU Leuven-University of Leuven, Department of Neurosciences, Experimental Neurology, and Leuven Brain Institute (LBI), Leuven, Belgium.,VIB, Center for Brain & Disease Research, Laboratory of Neurobiology, Leuven, Belgium
| |
Collapse
|
19
|
Vetrivel P, Kim SM, Ha SE, Kim HH, Bhosale PB, Senthil K, Kim GS. Compound Prunetin Induces Cell Death in Gastric Cancer Cell with Potent Anti-Proliferative Properties: In Vitro Assay, Molecular Docking, Dynamics, and ADMET Studies. Biomolecules 2020; 10:biom10071086. [PMID: 32708333 PMCID: PMC7408406 DOI: 10.3390/biom10071086] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Revised: 07/17/2020] [Accepted: 07/17/2020] [Indexed: 12/12/2022] Open
Abstract
Gastric cancer is the common type of malignancy positioned at second in mortality rate causing burden worldwide with increasing treatment options. Prunetin (PRU) is an O-methylated flavonoid that belongs to the group of isoflavone executing beneficial activities. In the present study, we investigated the anti-proliferative and cell death effect of the compound PRU in AGS gastric cancer cell line. The in vitro cytotoxic potential of PRU was evaluated and significant proliferation was observed. We identified that the mechanism of cell death was due to necroptosis through double staining and was confirmed by co-treatment with inhibitor necrostatin (Nec-1). We further elucidated the mechanism of action of necroptosis via receptor interacting protein kinase 3 (RIPK3) protein expression and it has been attributed by ROS generation through JNK activation. Furthermore, through computational analysis by molecular docking and dynamics simulation, the efficiency of compound prunetin against RIPK3 binding was validated. In addition, we also briefed the pharmacokinetic properties of the compound by in silico ADMET analysis.
Collapse
Affiliation(s)
- Preethi Vetrivel
- Research Institute of Life science and College of Veterinary Medicine, Gyeongsang National University, Gazwa, Jinju 52828, Korea; (P.V.); (S.M.K.); (S.E.H.); (H.H.K.); (P.B.B.)
| | - Seong Min Kim
- Research Institute of Life science and College of Veterinary Medicine, Gyeongsang National University, Gazwa, Jinju 52828, Korea; (P.V.); (S.M.K.); (S.E.H.); (H.H.K.); (P.B.B.)
| | - Sang Eun Ha
- Research Institute of Life science and College of Veterinary Medicine, Gyeongsang National University, Gazwa, Jinju 52828, Korea; (P.V.); (S.M.K.); (S.E.H.); (H.H.K.); (P.B.B.)
| | - Hun Hwan Kim
- Research Institute of Life science and College of Veterinary Medicine, Gyeongsang National University, Gazwa, Jinju 52828, Korea; (P.V.); (S.M.K.); (S.E.H.); (H.H.K.); (P.B.B.)
| | - Pritam Bhagwan Bhosale
- Research Institute of Life science and College of Veterinary Medicine, Gyeongsang National University, Gazwa, Jinju 52828, Korea; (P.V.); (S.M.K.); (S.E.H.); (H.H.K.); (P.B.B.)
| | - Kalaiselvi Senthil
- Department of Biochemistry, Biotechnology and Bioinformatics, Avinashilingam Institute for Home Science and Higher Education for Women, Coimbatore 641043, India;
| | - Gon Sup Kim
- Research Institute of Life science and College of Veterinary Medicine, Gyeongsang National University, Gazwa, Jinju 52828, Korea; (P.V.); (S.M.K.); (S.E.H.); (H.H.K.); (P.B.B.)
- Correspondence: ; Tel.: +82-010-3834-5823
| |
Collapse
|
20
|
Lin DQ, Cai XY, Wang CH, Yang B, Liang RS. Optimal concentration of necrostatin-1 for protecting against hippocampal neuronal damage in mice with status epilepticus. Neural Regen Res 2020; 15:936-943. [PMID: 31719260 PMCID: PMC6990772 DOI: 10.4103/1673-5374.268903] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2019] [Revised: 03/10/2019] [Accepted: 08/13/2019] [Indexed: 02/05/2023] Open
Abstract
Hippocampal neurons undergo various forms of cell death after status epilepticus. Necrostatin-1 specifically inhibits necroptosis mediated by receptor interacting protein kinase 1 (RIP1) and RIP3 receptors. However, there are no reports of necroptosis in mouse models of status epilepticus. Therefore, in this study, we investigated the effects of necrostatin-1 on hippocampal neurons in mice with status epilepticus, and, furthermore, we tested different amounts of the compound to identify the optimal concentration for inhibiting necroptosis and apoptosis. A mouse model of status epilepticus was produced by intraperitoneal injection of kainic acid, 12 mg/kg. Different concentrations of necrostatin-1 (10, 20, 40, and 80 μM) were administered into the lateral ventricle 15 minutes before kainic acid injection. Hippocampal damage was assessed by hematoxylin-eosin staining 24 hours after the model was successfully produced. Terminal deoxynucleotidyl transferase-mediated dUTP nick end labeling staining, western blot assay and immunohistochemistry were used to evaluate the expression of apoptosis-related and necroptosis-related proteins. Necrostatin-1 alleviated damage to hippocampal tissue in the mouse model of epilepsy. The 40 μM concentration of necrostatin-1 significantly decreased the number of apoptotic cells in the hippocampal CA1 region. Furthermore, necrostatin-1 significantly downregulated necroptosis-related proteins (MLKL, RIP1, and RIP3) and apoptosis-related proteins (cleaved-Caspase-3, Bax), and it upregulated the expression of anti-apoptotic protein Bcl-2. Taken together, our findings show that necrostatin-1 effectively inhibits necroptosis and apoptosis in mice with status epilepticus, with the 40 μM concentration of the compound having an optimal effect. The experiments were approved by the Animal Ethics Committee of Fujian Medical University, China (approval No. 2016-032) on November 9, 2016.
Collapse
Affiliation(s)
- Dong-Qi Lin
- Department of Cardiothoracic Surgery, the Second Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong Province, China
- Department of Neurosurgery, Union Hospital, Fujian Medical University, Fuzhou, Fujian Province, China
| | - Xin-Ying Cai
- Clinical Research Center, Shantou Central Hospital, Affiliated Shantou Hospital of Sun Yat-sen University, Shantou, Guangdong Province, China
| | - Chun-Hua Wang
- Department of Neurosurgery, Union Hospital, Fujian Medical University, Fuzhou, Fujian Province, China
| | - Bin Yang
- Department of Neurosurgery, Union Hospital, Fujian Medical University, Fuzhou, Fujian Province, China
| | - Ri-Sheng Liang
- Department of Neurosurgery, Union Hospital, Fujian Medical University, Fuzhou, Fujian Province, China
- Correspondence to: Ri-Sheng Liang, .
| |
Collapse
|
21
|
Hu S, Chang X, Zhu H, Wang D, Chen G. PI3K mediates tumor necrosis factor induced-necroptosis through initiating RIP1-RIP3-MLKL signaling pathway activation. Cytokine 2020; 129:155046. [PMID: 32114297 DOI: 10.1016/j.cyto.2020.155046] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2019] [Revised: 02/07/2020] [Accepted: 02/19/2020] [Indexed: 01/15/2023]
Abstract
Necroptosis is a recently identified programmed cell death, which is initiated by receptor-interacting serine/threonine-protein kinase 1 (RIP1), RIP3 and mixed-lineage kinase domain-like protein (MLKL). It has been reported that necroptosis induced by tumor necrosis factor (TNF) was inhibited by the inhibitor of phosphatidylinositol-3-kinase (PI3K) and its substrate protein AKT, indicating that PI3K-AKT signaling pathway was involved in mediating TNF-induced necroptosis, whereas it is unclear how PI3K initiates necroptosis. In this study, we found that TNF-induced necroptosis was inhibited by chemical inhibition or genetic deletion of PI3K. Moreover, knockdown of p110α, the catalytic subunit of PI3K, significantly suppressed the phosphorylation of PI3K substrate protein AKT, and TNF-induced necroptosis was blocked by AKT inhibitors. Furthermore, we found that p110α knockdown also suppressed the phosphorylation and oligomerization of RIP1, RIP3 and MLKL in response to TNF stimulation. In addition to the critical role in mediating TNF-induced necrosome formation, p110α was also essential for the spontaneous phosphorylation of RIP1 and RIP3. Finally, we found that p110α bound to RIP3, but not RIP1, to form protein complex in the process of TNF-induced necroptosis, and mediated TNF-induced necroptosis in the absence of RIP1. Our results demonstrate that PI3K is essential for TNF-induced necroptosis, which may act as the partner of RIP3 to initiate the activation of RIP1-RIP3-MLKL signal pathway and the subsequent necroptosis.
Collapse
Affiliation(s)
- Shiping Hu
- Department of Gastroenterology, The 983rd Hospital of Chinese PLA Joint Logistics Support Force, Tianjin 300142, China
| | - Xixi Chang
- Institute of Military Cognition and Brain Sciences, Academy of Military Medical Sciences, Beijing 100850, China
| | - Hongbin Zhu
- Department of Gastroenterology, The 983rd Hospital of Chinese PLA Joint Logistics Support Force, Tianjin 300142, China
| | - Dongxu Wang
- Department of Gastroenterology, The 983rd Hospital of Chinese PLA Joint Logistics Support Force, Tianjin 300142, China.
| | - Guozhu Chen
- Institute of Military Cognition and Brain Sciences, Academy of Military Medical Sciences, Beijing 100850, China.
| |
Collapse
|
22
|
Wang L, Chang X, Feng J, Yu J, Chen G. TRADD Mediates RIPK1-Independent Necroptosis Induced by Tumor Necrosis Factor. Front Cell Dev Biol 2020; 7:393. [PMID: 32039207 PMCID: PMC6987388 DOI: 10.3389/fcell.2019.00393] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2019] [Accepted: 12/24/2019] [Indexed: 01/10/2023] Open
Abstract
As a programmed necrotic cell death, necroptosis has the intrinsic initiators, including receptor-interacting serine/threonine-protein kinase 1 (RIPK1), RIPK3 and mixed-lineage kinase domain-like protein (MLKL), which combine to form necroptotic signaling pathway and mediate necroptosis induced by various necroptotic stimuli, such as tumor necrosis factor (TNF). Although chemical inhibition of RIPK1 blocks TNF-induced necroptosis, genetic elimination of RIPK1 does not suppress but facilitate necroptosis triggered by TNF. Moreover, RIPK3 has been reported to mediate the RIPK1-independent necroptosis, but the involved mechanism is unclear. In this study, we found that TRADD was essential for TNF-induced necroptosis in RIPK1-knockdown L929 and HT-22 cells. Mechanistic study demonstrated that TRADD bound RIPK3 to form new protein complex, which then promoted RIPK3 phosphorylation via facilitating RIPK3 oligomerization, leading to RIPK3-MLKL signaling pathway activation. Therefore, TRADD acted as a partner of RIPK3 to initiate necroptosis in RIPK1-knockdown L929 and HT-22 cells in response to TNF stimulation. In addition, TRADD was critical for the accumulation of reactive oxygen species (ROS), which contributed to RIPK1-independent necroptosis triggered by TNF. Collectively, our data demonstrate that TRADD acts as the new target protein for TNF-induced RIPK3 activation and the subsequent necroptosis in a RIPK1-independent manner.
Collapse
Affiliation(s)
- Lili Wang
- Institute of Military Cognition and Brain Sciences, Academy of Military Medical Sciences, Beijing, China
| | - Xixi Chang
- Institute of Military Cognition and Brain Sciences, Academy of Military Medical Sciences, Beijing, China
| | - Jinli Feng
- Department of Neurology, The Eighth Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Jiyun Yu
- Institute of Military Cognition and Brain Sciences, Academy of Military Medical Sciences, Beijing, China.,Beijing Zhendandingtai Biotechnology Co., Ltd, Beijing, China
| | - Guozhu Chen
- Institute of Military Cognition and Brain Sciences, Academy of Military Medical Sciences, Beijing, China
| |
Collapse
|
23
|
Molnár T, Mázló A, Tslaf V, Szöllősi AG, Emri G, Koncz G. Current translational potential and underlying molecular mechanisms of necroptosis. Cell Death Dis 2019; 10:860. [PMID: 31719524 PMCID: PMC6851151 DOI: 10.1038/s41419-019-2094-z] [Citation(s) in RCA: 79] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2019] [Revised: 10/22/2019] [Accepted: 10/24/2019] [Indexed: 12/27/2022]
Abstract
Cell death has a fundamental impact on the evolution of degenerative disorders, autoimmune processes, inflammatory diseases, tumor formation and immune surveillance. Over the past couple of decades extensive studies have uncovered novel cell death pathways, which are independent of apoptosis. Among these is necroptosis, a tightly regulated, inflammatory form of cell death. Necroptosis contribute to the pathogenesis of many diseases and in this review, we will focus exclusively on necroptosis in humans. Necroptosis is considered a backup mechanism of apoptosis, but the in vivo appearance of necroptosis indicates that both caspase-mediated and caspase-independent mechanisms control necroptosis. Necroptosis is regulated on multiple levels, from the transcription, to the stability and posttranslational modifications of the necrosome components, to the availability of molecular interaction partners and the localization of receptor-interacting serine/threonine-protein kinase 1 (RIPK1), receptor-interacting serine/threonine-protein kinase 3 (RIPK3) and mixed lineage kinase domain-like protein (MLKL). Accordingly, we classified the role of more than seventy molecules in necroptotic signaling based on consistent in vitro or in vivo evidence to understand the molecular background of necroptosis and to find opportunities where regulating the intensity and the modality of cell death could be exploited in clinical interventions. Necroptosis specific inhibitors are under development, but >20 drugs, already used in the treatment of various diseases, have the potential to regulate necroptosis. By listing necroptosis-modulated human diseases and cataloging the currently available drug-repertoire to modify necroptosis intensity, we hope to kick-start approaches with immediate translational potential. We also indicate where necroptosis regulating capacity should be considered in the current applications of these drugs.
Collapse
Affiliation(s)
- Tamás Molnár
- Department of Immunology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
- Doctoral School of Molecular Cellular and Immune Biology, University of Debrecen, Debrecen, Hungary
| | - Anett Mázló
- Department of Immunology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
- Doctoral School of Molecular Cellular and Immune Biology, University of Debrecen, Debrecen, Hungary
- MTA-DE Cell Biology and Signaling Research Group, Faculty of Medicine, University of Debrecen, Debrecen, 4032, Hungary
| | - Vera Tslaf
- Department of Immunology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Attila Gábor Szöllősi
- Department of Immunology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Gabriella Emri
- Department of Dermatology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Gábor Koncz
- Department of Immunology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary.
| |
Collapse
|