1
|
Wu Q, Qi S, Kang Z, Bai X, Li Z, Cheng J, Dong X. PANoptosis in Sepsis: A Central Role and Emerging Therapeutic Target. J Inflamm Res 2025; 18:6245-6261. [PMID: 40386177 PMCID: PMC12085136 DOI: 10.2147/jir.s513367] [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: 12/20/2024] [Accepted: 05/04/2025] [Indexed: 05/20/2025] Open
Abstract
The pathogenesis of sepsis is intricately linked to regulated cell death. As a novel form of regulated cell death, PANoptosis plays a critical role in driving the inflammatory response, impairing immune cell function, and contributing to multi-organ dysfunction in sepsis. This review explores the molecular mechanisms underlying PANoptosis and its involvement in sepsis. By activating multiple pathways, PANoptosis promotes the release of inflammatory cytokines, triggering a cytokine storm that disrupts immune cell homeostasis and exacerbates organ damage. Emerging therapeutic strategies targeting PANoptosis, including chemotherapeutic agents and herbal remedies, are showing potential for clinical application. The concept of targeting PANoptosis offers a promising avenue for developing innovative treatments for sepsis.
Collapse
Affiliation(s)
- Qiqi Wu
- Trauma Center/Department of Emergency and Traumatic Surgery, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
| | - Siyuan Qi
- Trauma Center/Department of Emergency and Traumatic Surgery, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
| | - Zhaofeng Kang
- Trauma Center/Department of Emergency and Traumatic Surgery, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
| | - Xiangjun Bai
- Trauma Center/Department of Emergency and Traumatic Surgery, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
| | - Zhanfei Li
- Trauma Center/Department of Emergency and Traumatic Surgery, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
| | - Jing Cheng
- Trauma Center/Department of Emergency and Traumatic Surgery, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
| | - Xijie Dong
- Trauma Center/Department of Emergency and Traumatic Surgery, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
| |
Collapse
|
2
|
Zheng Y, Cao Y, Wang W, Tong Y, Wang S, Li C, Zhao M, Song Y, Wang YGS, Qi J, Wu C, Yang J, Zheng J, Gao J, Wang J, Yang Q, Liu G, Zhao J, Zhang Y, Xiao H, Zhang YY, Tang YD. Dusp14-Mediated Dephosphorylation of MLKL Protects Against Cardiomyocyte Necroptosis in Hypothyroidism-Induced Heart Failure. Circulation 2025. [PMID: 40357546 DOI: 10.1161/circulationaha.125.074353] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/20/2025] [Accepted: 04/03/2025] [Indexed: 05/15/2025]
Abstract
BACKGROUND Hypothyroidism leads to multiple organ dysfunction, with the heart the most affected. However, the pathologic mechanism of hypothyroidism-induced heart failure remains to be completely elucidated. Thyroid hormone replacement therapy enhances myocardium systolic function but increases the occurrence of arrythmias. There is an urgent need to explore these mechanisms in detail and to discover and develop drugs that can target and manage heart failure in patients with hypothyroidism. METHODS In this study, a mouse model of hypothyroidism-induced heart failure was established through the administration of propylthiouracil. Dusp14 knockout mice were generated, and adeno-associated virus-mediated cardiomyocyte-specific overexpression of Dusp14 (dual specificity phosphatase 14) was used in combination with related cellular experiments to investigate the protective effects of Dusp14 on hypothyroidism-induced heart failure. Further analyses confirmed the crucial involvement of necroptosis in the pathogenesis of hypothyroidism-induced heart failure, and demonstrated the protective role of Dusp14 in modulating necroptosis. In addition, a novel small molecule compound that effectively regulates Dusp14 activity in vitro was identified through molecular docking, providing a potential therapeutic avenue. RESULTS Dusp14 regulates necroptosis and mitigates hypothyroidism-induced heart failure. Myocardial tissue sections from mice in the hypothyroidism group showed positive Evans blue dye staining, and the serum levels of the myocardial injury marker lactate dehydrogenase were significantly higher compared with the euthyroid group (n=8). In addition, phosphorylation levels of the necroptosis marker MLKL (mixed lineage kinase domain-like protein) were significantly elevated, indicating the activation of necroptosis (n=8). These findings suggest that myocardial necroptosis is activated during hypothyroidism. Myocardial-specific overexpression of Dusp14 reduced myocardial necroptosis and improved myocardial contractile function in hypothyroid mice (n=8). In contrast, Dusp14 knockout exacerbated myocardial contractile dysfunction and necroptosis in these mice (n=5-7). These results indicate that Dusp14 alleviates hypothyroidism-induced heart failure by inhibiting necroptosis. P077-0472, a small molecule compound, was identified as an activator of Dusp14, which could inhibit cardiomyocyte necroptosis from hypothyroidism (n=6). CONCLUSIONS Dusp14 inhibits cardiomyocyte necroptosis from hypothyroidism and consequently rescues damaged cardiomyocytes. P077-0472, a novel small molecule compound that activates the dephosphorylation function of Dusp14, could inhibit cardiomyocyte necroptosis from hypothyroidism.
Collapse
Affiliation(s)
- Yitian Zheng
- Department of Cardiology and Institute of Vascular Medicine, Peking University Third Hospital, Beijing, China (Y. Zheng, Y.C., W.W., Y.T., C.L., M.Z., Y.S., Y.-g.-s.W., J.Q., C.W., J.Y., J. Zheng, J.G., J.W., Y. Zhang, H.X., Y.-Y.Z., Y.-D.T.)
- Beijing Key Laboratory of Clinical Evaluation of Cardiovascular-Kidney-Metabolic and Immuno-Inflammatory Innovative Drugs and Medical Devices, State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, China (Y. Zheng, Y.C., W.W., Y.T., C.L., M.Z., Y.S., Y.-g.-s.W., J.Q., C.W., J.Y., J. Zheng, J.G., J.W., Y. Zhang, H.X., Y.-Y.Z., Y.-D.T.)
- Research Unit of Medical Science Research Management/Basic and Clinical Research of Metabolic Cardiovascular Diseases, Chinese Academy of Medical Sciences, Beijing, China (Y. Zheng, W.W., H.X., Y.-Y.Z., Y.-D.T.)
- Department of Cardiology, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China (Y. Zheng)
| | - Yueyue Cao
- Department of Cardiology and Institute of Vascular Medicine, Peking University Third Hospital, Beijing, China (Y. Zheng, Y.C., W.W., Y.T., C.L., M.Z., Y.S., Y.-g.-s.W., J.Q., C.W., J.Y., J. Zheng, J.G., J.W., Y. Zhang, H.X., Y.-Y.Z., Y.-D.T.)
- Beijing Key Laboratory of Clinical Evaluation of Cardiovascular-Kidney-Metabolic and Immuno-Inflammatory Innovative Drugs and Medical Devices, State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, China (Y. Zheng, Y.C., W.W., Y.T., C.L., M.Z., Y.S., Y.-g.-s.W., J.Q., C.W., J.Y., J. Zheng, J.G., J.W., Y. Zhang, H.X., Y.-Y.Z., Y.-D.T.)
- The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China (Y.C.)
| | - Wenyao Wang
- Department of Cardiology and Institute of Vascular Medicine, Peking University Third Hospital, Beijing, China (Y. Zheng, Y.C., W.W., Y.T., C.L., M.Z., Y.S., Y.-g.-s.W., J.Q., C.W., J.Y., J. Zheng, J.G., J.W., Y. Zhang, H.X., Y.-Y.Z., Y.-D.T.)
- Beijing Key Laboratory of Clinical Evaluation of Cardiovascular-Kidney-Metabolic and Immuno-Inflammatory Innovative Drugs and Medical Devices, State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, China (Y. Zheng, Y.C., W.W., Y.T., C.L., M.Z., Y.S., Y.-g.-s.W., J.Q., C.W., J.Y., J. Zheng, J.G., J.W., Y. Zhang, H.X., Y.-Y.Z., Y.-D.T.)
- Research Unit of Medical Science Research Management/Basic and Clinical Research of Metabolic Cardiovascular Diseases, Chinese Academy of Medical Sciences, Beijing, China (Y. Zheng, W.W., H.X., Y.-Y.Z., Y.-D.T.)
| | - Yicheng Tong
- Department of Cardiology and Institute of Vascular Medicine, Peking University Third Hospital, Beijing, China (Y. Zheng, Y.C., W.W., Y.T., C.L., M.Z., Y.S., Y.-g.-s.W., J.Q., C.W., J.Y., J. Zheng, J.G., J.W., Y. Zhang, H.X., Y.-Y.Z., Y.-D.T.)
- Beijing Key Laboratory of Clinical Evaluation of Cardiovascular-Kidney-Metabolic and Immuno-Inflammatory Innovative Drugs and Medical Devices, State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, China (Y. Zheng, Y.C., W.W., Y.T., C.L., M.Z., Y.S., Y.-g.-s.W., J.Q., C.W., J.Y., J. Zheng, J.G., J.W., Y. Zhang, H.X., Y.-Y.Z., Y.-D.T.)
| | - Shuaixing Wang
- National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China (S.W.)
| | - Chen Li
- Department of Cardiology and Institute of Vascular Medicine, Peking University Third Hospital, Beijing, China (Y. Zheng, Y.C., W.W., Y.T., C.L., M.Z., Y.S., Y.-g.-s.W., J.Q., C.W., J.Y., J. Zheng, J.G., J.W., Y. Zhang, H.X., Y.-Y.Z., Y.-D.T.)
- Beijing Key Laboratory of Clinical Evaluation of Cardiovascular-Kidney-Metabolic and Immuno-Inflammatory Innovative Drugs and Medical Devices, State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, China (Y. Zheng, Y.C., W.W., Y.T., C.L., M.Z., Y.S., Y.-g.-s.W., J.Q., C.W., J.Y., J. Zheng, J.G., J.W., Y. Zhang, H.X., Y.-Y.Z., Y.-D.T.)
| | - Mingming Zhao
- Department of Cardiology and Institute of Vascular Medicine, Peking University Third Hospital, Beijing, China (Y. Zheng, Y.C., W.W., Y.T., C.L., M.Z., Y.S., Y.-g.-s.W., J.Q., C.W., J.Y., J. Zheng, J.G., J.W., Y. Zhang, H.X., Y.-Y.Z., Y.-D.T.)
- Beijing Key Laboratory of Clinical Evaluation of Cardiovascular-Kidney-Metabolic and Immuno-Inflammatory Innovative Drugs and Medical Devices, State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, China (Y. Zheng, Y.C., W.W., Y.T., C.L., M.Z., Y.S., Y.-g.-s.W., J.Q., C.W., J.Y., J. Zheng, J.G., J.W., Y. Zhang, H.X., Y.-Y.Z., Y.-D.T.)
| | - Yao Song
- Department of Cardiology and Institute of Vascular Medicine, Peking University Third Hospital, Beijing, China (Y. Zheng, Y.C., W.W., Y.T., C.L., M.Z., Y.S., Y.-g.-s.W., J.Q., C.W., J.Y., J. Zheng, J.G., J.W., Y. Zhang, H.X., Y.-Y.Z., Y.-D.T.)
- Beijing Key Laboratory of Clinical Evaluation of Cardiovascular-Kidney-Metabolic and Immuno-Inflammatory Innovative Drugs and Medical Devices, State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, China (Y. Zheng, Y.C., W.W., Y.T., C.L., M.Z., Y.S., Y.-g.-s.W., J.Q., C.W., J.Y., J. Zheng, J.G., J.W., Y. Zhang, H.X., Y.-Y.Z., Y.-D.T.)
| | - Yuan-Geng-Shuo Wang
- Department of Cardiology and Institute of Vascular Medicine, Peking University Third Hospital, Beijing, China (Y. Zheng, Y.C., W.W., Y.T., C.L., M.Z., Y.S., Y.-g.-s.W., J.Q., C.W., J.Y., J. Zheng, J.G., J.W., Y. Zhang, H.X., Y.-Y.Z., Y.-D.T.)
- Beijing Key Laboratory of Clinical Evaluation of Cardiovascular-Kidney-Metabolic and Immuno-Inflammatory Innovative Drugs and Medical Devices, State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, China (Y. Zheng, Y.C., W.W., Y.T., C.L., M.Z., Y.S., Y.-g.-s.W., J.Q., C.W., J.Y., J. Zheng, J.G., J.W., Y. Zhang, H.X., Y.-Y.Z., Y.-D.T.)
| | - Jiating Qi
- Department of Cardiology and Institute of Vascular Medicine, Peking University Third Hospital, Beijing, China (Y. Zheng, Y.C., W.W., Y.T., C.L., M.Z., Y.S., Y.-g.-s.W., J.Q., C.W., J.Y., J. Zheng, J.G., J.W., Y. Zhang, H.X., Y.-Y.Z., Y.-D.T.)
- Beijing Key Laboratory of Clinical Evaluation of Cardiovascular-Kidney-Metabolic and Immuno-Inflammatory Innovative Drugs and Medical Devices, State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, China (Y. Zheng, Y.C., W.W., Y.T., C.L., M.Z., Y.S., Y.-g.-s.W., J.Q., C.W., J.Y., J. Zheng, J.G., J.W., Y. Zhang, H.X., Y.-Y.Z., Y.-D.T.)
| | - Chao Wu
- Department of Cardiology and Institute of Vascular Medicine, Peking University Third Hospital, Beijing, China (Y. Zheng, Y.C., W.W., Y.T., C.L., M.Z., Y.S., Y.-g.-s.W., J.Q., C.W., J.Y., J. Zheng, J.G., J.W., Y. Zhang, H.X., Y.-Y.Z., Y.-D.T.)
- Beijing Key Laboratory of Clinical Evaluation of Cardiovascular-Kidney-Metabolic and Immuno-Inflammatory Innovative Drugs and Medical Devices, State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, China (Y. Zheng, Y.C., W.W., Y.T., C.L., M.Z., Y.S., Y.-g.-s.W., J.Q., C.W., J.Y., J. Zheng, J.G., J.W., Y. Zhang, H.X., Y.-Y.Z., Y.-D.T.)
| | - Jie Yang
- Department of Cardiology and Institute of Vascular Medicine, Peking University Third Hospital, Beijing, China (Y. Zheng, Y.C., W.W., Y.T., C.L., M.Z., Y.S., Y.-g.-s.W., J.Q., C.W., J.Y., J. Zheng, J.G., J.W., Y. Zhang, H.X., Y.-Y.Z., Y.-D.T.)
- Beijing Key Laboratory of Clinical Evaluation of Cardiovascular-Kidney-Metabolic and Immuno-Inflammatory Innovative Drugs and Medical Devices, State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, China (Y. Zheng, Y.C., W.W., Y.T., C.L., M.Z., Y.S., Y.-g.-s.W., J.Q., C.W., J.Y., J. Zheng, J.G., J.W., Y. Zhang, H.X., Y.-Y.Z., Y.-D.T.)
| | - Jilin Zheng
- Department of Cardiology and Institute of Vascular Medicine, Peking University Third Hospital, Beijing, China (Y. Zheng, Y.C., W.W., Y.T., C.L., M.Z., Y.S., Y.-g.-s.W., J.Q., C.W., J.Y., J. Zheng, J.G., J.W., Y. Zhang, H.X., Y.-Y.Z., Y.-D.T.)
- Beijing Key Laboratory of Clinical Evaluation of Cardiovascular-Kidney-Metabolic and Immuno-Inflammatory Innovative Drugs and Medical Devices, State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, China (Y. Zheng, Y.C., W.W., Y.T., C.L., M.Z., Y.S., Y.-g.-s.W., J.Q., C.W., J.Y., J. Zheng, J.G., J.W., Y. Zhang, H.X., Y.-Y.Z., Y.-D.T.)
| | - Jun Gao
- Department of Cardiology and Institute of Vascular Medicine, Peking University Third Hospital, Beijing, China (Y. Zheng, Y.C., W.W., Y.T., C.L., M.Z., Y.S., Y.-g.-s.W., J.Q., C.W., J.Y., J. Zheng, J.G., J.W., Y. Zhang, H.X., Y.-Y.Z., Y.-D.T.)
- Beijing Key Laboratory of Clinical Evaluation of Cardiovascular-Kidney-Metabolic and Immuno-Inflammatory Innovative Drugs and Medical Devices, State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, China (Y. Zheng, Y.C., W.W., Y.T., C.L., M.Z., Y.S., Y.-g.-s.W., J.Q., C.W., J.Y., J. Zheng, J.G., J.W., Y. Zhang, H.X., Y.-Y.Z., Y.-D.T.)
| | - Jingjia Wang
- Department of Cardiology and Institute of Vascular Medicine, Peking University Third Hospital, Beijing, China (Y. Zheng, Y.C., W.W., Y.T., C.L., M.Z., Y.S., Y.-g.-s.W., J.Q., C.W., J.Y., J. Zheng, J.G., J.W., Y. Zhang, H.X., Y.-Y.Z., Y.-D.T.)
- Beijing Key Laboratory of Clinical Evaluation of Cardiovascular-Kidney-Metabolic and Immuno-Inflammatory Innovative Drugs and Medical Devices, State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, China (Y. Zheng, Y.C., W.W., Y.T., C.L., M.Z., Y.S., Y.-g.-s.W., J.Q., C.W., J.Y., J. Zheng, J.G., J.W., Y. Zhang, H.X., Y.-Y.Z., Y.-D.T.)
| | - Qing Yang
- Department of Cardiology, Tianjin Medical University General Hospital, China (Q.Y.)
| | - Gang Liu
- Department of Cardiology, the First Hospital of Hebei Medical University, Shijiazhuang, Hebei, China (G.L.)
| | - Jiajun Zhao
- Department of Endocrinology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China (J. Zhao)
| | - Yan Zhang
- Department of Cardiology and Institute of Vascular Medicine, Peking University Third Hospital, Beijing, China (Y. Zheng, Y.C., W.W., Y.T., C.L., M.Z., Y.S., Y.-g.-s.W., J.Q., C.W., J.Y., J. Zheng, J.G., J.W., Y. Zhang, H.X., Y.-Y.Z., Y.-D.T.)
- Beijing Key Laboratory of Clinical Evaluation of Cardiovascular-Kidney-Metabolic and Immuno-Inflammatory Innovative Drugs and Medical Devices, State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, China (Y. Zheng, Y.C., W.W., Y.T., C.L., M.Z., Y.S., Y.-g.-s.W., J.Q., C.W., J.Y., J. Zheng, J.G., J.W., Y. Zhang, H.X., Y.-Y.Z., Y.-D.T.)
| | - Han Xiao
- Department of Cardiology and Institute of Vascular Medicine, Peking University Third Hospital, Beijing, China (Y. Zheng, Y.C., W.W., Y.T., C.L., M.Z., Y.S., Y.-g.-s.W., J.Q., C.W., J.Y., J. Zheng, J.G., J.W., Y. Zhang, H.X., Y.-Y.Z., Y.-D.T.)
- Beijing Key Laboratory of Clinical Evaluation of Cardiovascular-Kidney-Metabolic and Immuno-Inflammatory Innovative Drugs and Medical Devices, State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, China (Y. Zheng, Y.C., W.W., Y.T., C.L., M.Z., Y.S., Y.-g.-s.W., J.Q., C.W., J.Y., J. Zheng, J.G., J.W., Y. Zhang, H.X., Y.-Y.Z., Y.-D.T.)
- Research Unit of Medical Science Research Management/Basic and Clinical Research of Metabolic Cardiovascular Diseases, Chinese Academy of Medical Sciences, Beijing, China (Y. Zheng, W.W., H.X., Y.-Y.Z., Y.-D.T.)
| | - You-Yi Zhang
- Department of Cardiology and Institute of Vascular Medicine, Peking University Third Hospital, Beijing, China (Y. Zheng, Y.C., W.W., Y.T., C.L., M.Z., Y.S., Y.-g.-s.W., J.Q., C.W., J.Y., J. Zheng, J.G., J.W., Y. Zhang, H.X., Y.-Y.Z., Y.-D.T.)
- Beijing Key Laboratory of Clinical Evaluation of Cardiovascular-Kidney-Metabolic and Immuno-Inflammatory Innovative Drugs and Medical Devices, State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, China (Y. Zheng, Y.C., W.W., Y.T., C.L., M.Z., Y.S., Y.-g.-s.W., J.Q., C.W., J.Y., J. Zheng, J.G., J.W., Y. Zhang, H.X., Y.-Y.Z., Y.-D.T.)
- Research Unit of Medical Science Research Management/Basic and Clinical Research of Metabolic Cardiovascular Diseases, Chinese Academy of Medical Sciences, Beijing, China (Y. Zheng, W.W., H.X., Y.-Y.Z., Y.-D.T.)
| | - Yi-Da Tang
- Department of Cardiology and Institute of Vascular Medicine, Peking University Third Hospital, Beijing, China (Y. Zheng, Y.C., W.W., Y.T., C.L., M.Z., Y.S., Y.-g.-s.W., J.Q., C.W., J.Y., J. Zheng, J.G., J.W., Y. Zhang, H.X., Y.-Y.Z., Y.-D.T.)
- Beijing Key Laboratory of Clinical Evaluation of Cardiovascular-Kidney-Metabolic and Immuno-Inflammatory Innovative Drugs and Medical Devices, State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, China (Y. Zheng, Y.C., W.W., Y.T., C.L., M.Z., Y.S., Y.-g.-s.W., J.Q., C.W., J.Y., J. Zheng, J.G., J.W., Y. Zhang, H.X., Y.-Y.Z., Y.-D.T.)
- Research Unit of Medical Science Research Management/Basic and Clinical Research of Metabolic Cardiovascular Diseases, Chinese Academy of Medical Sciences, Beijing, China (Y. Zheng, W.W., H.X., Y.-Y.Z., Y.-D.T.)
- University of Health and Rehabilitation Sciences, Qingdao, China (Y.-D.T.)
| |
Collapse
|
3
|
Shi SN, Xu Q, Jiao X, Wen Y, Wu Y, Liu J, Ma D, Zhao B, Gao Q, Fang Y. ATR-mediated phosphorylation of RIPK1 inhibits DNA damage-induced necroptosis. Biochem Pharmacol 2025; 237:116949. [PMID: 40228635 DOI: 10.1016/j.bcp.2025.116949] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2025] [Revised: 03/30/2025] [Accepted: 04/11/2025] [Indexed: 04/16/2025]
Abstract
Necroptosis induced by DNA damage during chemotherapy is a significant and effective treatment strategy for malignant tumors. Ataxia telangiectasia and rad3-related protein (ATR), a key kinase in DNA damage checkpoints, initiates repair by transmitting damage signals to effectors. However, persistent DNA damage may result in cell death. The mechanisms by which ATR regulates necroptosis remain incompletely understood. In this study, we demonstrated that ATR binds to receptor-interacting serine/threonine-protein kinase 1 (RIPK1) and inhibits its activation, thereby suppressing RIPK1-dependent necroptosis triggered by DNA damage. Mechanistically, ATR directly inhibited RIPK1 and downstream necrosome formation through Ser335 phosphorylation following DNA damage, thereby attenuating RIPK1-dependent necroptosis. In the case of the S335A mutation, RIPK1 repression was relieved, leading to enhanced downstream necroptosis. Furthermore, RIPK1 knockout with complementation of wild-type or S335A mutation in ovarian cancer cell lines revealed that ATR phosphorylation of RIPK1 at S335 promoted chemoresistance, while the S335A mutation significantly increased chemosensitivity. This was characterized by heightened necroptosis activation, reduced cell viability, and increased cell death. These findings expand our understanding of the interaction between DNA damage and cell death regulation and may aid in developing therapeutic drugs to enhance DNA damage-induced tumor necroptosis and improve chemosensitivity.
Collapse
Affiliation(s)
- Shen-Nan Shi
- Department of Gynecological Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; National Clinical Research Center for Obstetrics and Gynecology, Cancer Biology Research Center (Key Laboratory of the Ministry of Education), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Qiuyang Xu
- Department of Gynecological Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; National Clinical Research Center for Obstetrics and Gynecology, Cancer Biology Research Center (Key Laboratory of the Ministry of Education), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaofei Jiao
- Department of Gynecological Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; National Clinical Research Center for Obstetrics and Gynecology, Cancer Biology Research Center (Key Laboratory of the Ministry of Education), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yuanjia Wen
- Department of Gynecological Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; National Clinical Research Center for Obstetrics and Gynecology, Cancer Biology Research Center (Key Laboratory of the Ministry of Education), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yijie Wu
- Department of Gynecological Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; National Clinical Research Center for Obstetrics and Gynecology, Cancer Biology Research Center (Key Laboratory of the Ministry of Education), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jiahao Liu
- Department of Gynecological Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; National Clinical Research Center for Obstetrics and Gynecology, Cancer Biology Research Center (Key Laboratory of the Ministry of Education), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ding Ma
- Department of Gynecological Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; National Clinical Research Center for Obstetrics and Gynecology, Cancer Biology Research Center (Key Laboratory of the Ministry of Education), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Bingbing Zhao
- Department of Obstetrics and Gynecology, Guangxi Medical University of Cancer Hospital, Nanning, Guangxi 530021, China; State Key Laboratory of Targeting Oncology, Guangxi Medical University, Nanning, Guangxi 530021, China.
| | - Qinglei Gao
- Department of Gynecological Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; National Clinical Research Center for Obstetrics and Gynecology, Cancer Biology Research Center (Key Laboratory of the Ministry of Education), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Yong Fang
- Department of Gynecological Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; National Clinical Research Center for Obstetrics and Gynecology, Cancer Biology Research Center (Key Laboratory of the Ministry of Education), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| |
Collapse
|
4
|
Chen J, Zhang WJ, Liu XY, Hu TP, Gao N, Li ZH, Wang Y, Zhang GQ. KW-2449 Ameliorates Cardiac Dysfunction in a Rat Model of Sepsis-Induced Cardiomyopathy. Inflammation 2025:10.1007/s10753-024-02223-y. [PMID: 39843680 DOI: 10.1007/s10753-024-02223-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2024] [Revised: 12/14/2024] [Accepted: 12/17/2024] [Indexed: 01/24/2025]
Abstract
KW-2449 is a novel multitargeted kinase inhibitor that has been reported to alleviate chronic inflammation and altered immunity during the treatment of autoimmune diseases. The aim of the study was to investigate the effect of KW-2449 on sepsis-induced cardiomyopathy (SIC). A rat model of moderate SIC was induced using the cecal ligation and puncture (CLP) method. KW-2449 was administered to rats at 10 mg/kg for 3 consecutive days by intraperitoneal injection. At 24 hours after CLP, echocardiography, electrocardiogram, and hemodynamic analyses were performed. Blood and cardiac tissues were collected for further analysis. RNA sequencing (RNA-seq) analyses were used to identify the key genes affected by KW-2449 treatment during SIC. KW-2449 improved the liver dysfunction in septic rats. KW-2449 significantly improved left ventricular (LV) systolic function and hemodynamics compared to the CLP group. KW-2449 suppressed the systemic inflammatory response, decreased myocardial inflammation and cell apoptosis in the CLP rats. RNA-seq analyses indicated that there were a total of 2256 differentially expressed genes in the CLP group compared to the Control group, among which 63 genes were down-regulated and 59 genes were up-regulated by KW-2449. Specifically, Pparα was identified as a key target gene of KW-2449 in the treatment of SIC by RNA-seq analysis.KW-2449 also significantly upregulated the protein expression of Pparα in the LV tissue of septic rats. KW-2449 reduced systemic inflammation, cardiac inflammation, and improved cardiac dysfunction in the CLP-induced SIC rat model. The underlying mechanism of the cardio-protective role of KW-2449 in the CLP-induced SIC might be related to Pparα.
Collapse
Affiliation(s)
- Jie Chen
- Department of Emergency, China-Japan Friendship Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100029, China
| | - Wei-Jian Zhang
- Department of Emergency, China-Japan Friendship Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100029, China
| | - Xiao-Yu Liu
- Department of Emergency, China-Japan Friendship Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100029, China
| | - Tian-Peng Hu
- Department of Emergency, China-Japan Friendship Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100029, China
| | - Nan Gao
- Department of Emergency, China-Japan Friendship Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100029, China
| | - Zhong-Hao Li
- Department of Emergency, China-Japan Friendship Hospital, Beijing, 100029, China
| | - Yu Wang
- Department of Emergency, China-Japan Friendship Hospital, Beijing, 100029, China
| | - Guo-Qiang Zhang
- Department of Emergency, China-Japan Friendship Hospital, Beijing, 100029, China.
| |
Collapse
|
5
|
Zhao Y, Wang Q, Zhu J, Cai J, Feng X, Song Q, Jiang H, Ren W, He Y, Wang P, Feng D, Yu J, Liu Y, Wu Q, Jitkaew S, Cai Z. Identification of KW-2449 as a dual inhibitor of ferroptosis and necroptosis reveals that autophagy is a targetable pathway for necroptosis inhibitors to prevent ferroptosis. Cell Death Dis 2024; 15:764. [PMID: 39433736 PMCID: PMC11493980 DOI: 10.1038/s41419-024-07157-9] [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: 03/19/2024] [Revised: 10/10/2024] [Accepted: 10/14/2024] [Indexed: 10/23/2024]
Abstract
Necroptosis and ferroptosis are two distinct forms of necrotic-like cell death in terms of their morphological features and regulatory mechanisms. These two types of cell death can coexist in disease and contribute to pathological processes. Inhibition of both necroptosis and ferroptosis has been shown to enhance therapeutic effects in treating complex necrosis-related diseases. However, targeting both necroptosis and ferroptosis by a single compound can be challenging, as these two forms of cell death involve distinct molecular pathways. In this study, we discovered that KW-2449, a previously described necroptosis inhibitor, also prevented ferroptosis both in vitro and in vivo. Mechanistically, KW-2449 inhibited ferroptosis by targeting the autophagy pathway. We further identified that KW-2449 functioned as a ULK1 (Unc-51-like kinase 1) inhibitor to block ULK1 kinase activity in autophagy. Remarkably, we found that Necrostatin-1, a classic necroptosis inhibitor that has been shown to prevent ferroptosis, also targets the autophagy pathway to suppress ferroptosis. This study provides the first understanding of how necroptosis inhibitors can prevent ferroptosis and suggests that autophagy is a targetable pathway for necroptosis inhibitors to prevent ferroptosis. Therefore, the identification and design of pharmaceutical molecules that target the autophagy pathway from necroptosis inhibitors is a promising strategy to develop dual inhibitors of necroptosis and ferroptosis in clinical application.
Collapse
Affiliation(s)
- Yaxing Zhao
- Tongji University Cancer Center, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Qingsong Wang
- College of Pharmacy, Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region, Yinchuan, China
| | - Jing Zhu
- Tongji University Cancer Center, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, China
- Department of Biochemistry and Molecular Biology, School of Medicine, Tongji University, Shanghai, China
| | - Jin Cai
- Tongji University Cancer Center, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, China
- Department of Biochemistry and Molecular Biology, School of Medicine, Tongji University, Shanghai, China
| | - Xiaona Feng
- College of Pharmacy, Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region, Yinchuan, China
| | - Qianqian Song
- Tongji University Cancer Center, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, China
- Department of Biochemistry and Molecular Biology, School of Medicine, Tongji University, Shanghai, China
| | - Hui Jiang
- Tongji University Cancer Center, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, China
- Department of Biochemistry and Molecular Biology, School of Medicine, Tongji University, Shanghai, China
| | - Wenqing Ren
- Tongji University Cancer Center, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Yuan He
- Tongji University Cancer Center, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Ping Wang
- Tongji University Cancer Center, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Du Feng
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Degradation, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, China
| | - Jianqiang Yu
- College of Pharmacy, Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region, Yinchuan, China
| | - Yue Liu
- College of Pharmacy, Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region, Yinchuan, China
| | - Qihui Wu
- Shanghai Fourth People's Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Siriporn Jitkaew
- Center of Excellence for Cancer and Inflammation, Department of Clinical Chemistry, Faculty of Allied Health Sciences, Chulalongkorn University, Bangkok, Thailand
| | - Zhenyu Cai
- Tongji University Cancer Center, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, China.
- College of Pharmacy, Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region, Yinchuan, China.
- State Key Laboratory of Cardiology and Medical Innovation Center, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China.
| |
Collapse
|
6
|
Liu XM, Zhu ZZ, He XR, Zou YH, Chen Q, Wang XY, Liu HM, Qiao X, Wang X, Xu JY. NIR Light and GSH Dual-Responsive Upconversion Nanoparticles Loaded with Multifunctional Platinum(IV) Prodrug and RGD Peptide for Precise Cancer Therapy. ACS APPLIED MATERIALS & INTERFACES 2024; 16:40753-40766. [PMID: 39046129 DOI: 10.1021/acsami.4c08899] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/25/2024]
Abstract
Platinum(II) drugs as a first-line anticancer reagent are limited by side effects and drug resistance. Stimuli-responsive nanosystems hold promise for precise spatiotemporal manipulation of drug delivery, with the aim to promote bioavailability and minimize side effects. Herein, a multitargeting octahedral platinum(IV) prodrug with octadecyl aliphatic chain and histone deacetylase inhibitor (phenylbutyric acid, PHB) at axial positions to improve the therapeutic effect of cisplatin was loaded on the upconversion nanoparticles (UCNPs) through hydrophobic interaction. Followed attachment of DSPE-PEG2000 and arginine-glycine-aspartic (RGD) peptide endowed the nanovehicles with high biocompatibility and tumor specificity. The fabricated nanoparticles (UCNP/Pt(IV)-RGD) can be triggered by upconversion luminescence (UCL) irradiation and glutathione (GSH) reduction to controllably release Pt(II) species and PHB, inducing profound cytotoxicity. Both in vitro and in vivo experiments demonstrated that UCNP/Pt(IV)-RGD exhibited remarkable antitumor efficiency, high tumor-targeting specificity, and real-time UCL imaging capacity, presenting an intelligent platinum(IV) prodrug-loaded nanovehicle for UCL-guided dual-stimuli-responsive combination therapy.
Collapse
Affiliation(s)
- Xiao-Meng Liu
- Department of Chemical Biology and Tianjin Key Laboratory of Technologies Enabling Development of Clinical Therapeutics and Diagnostics, School of Pharmacy, Tianjin Medical University, Tianjin 300070, China
| | - Zhen-Zhen Zhu
- Department of Chemical Biology and Tianjin Key Laboratory of Technologies Enabling Development of Clinical Therapeutics and Diagnostics, School of Pharmacy, Tianjin Medical University, Tianjin 300070, China
| | - Xin-Rui He
- Department of Chemical Biology and Tianjin Key Laboratory of Technologies Enabling Development of Clinical Therapeutics and Diagnostics, School of Pharmacy, Tianjin Medical University, Tianjin 300070, China
| | - Yun-Hong Zou
- Department of Chemical Biology and Tianjin Key Laboratory of Technologies Enabling Development of Clinical Therapeutics and Diagnostics, School of Pharmacy, Tianjin Medical University, Tianjin 300070, China
| | - Qian Chen
- Department of Chemical Biology and Tianjin Key Laboratory of Technologies Enabling Development of Clinical Therapeutics and Diagnostics, School of Pharmacy, Tianjin Medical University, Tianjin 300070, China
| | - Xiao-Ya Wang
- Department of Chemical Biology and Tianjin Key Laboratory of Technologies Enabling Development of Clinical Therapeutics and Diagnostics, School of Pharmacy, Tianjin Medical University, Tianjin 300070, China
| | - Hui-Mei Liu
- Department of Chemical Biology and Tianjin Key Laboratory of Technologies Enabling Development of Clinical Therapeutics and Diagnostics, School of Pharmacy, Tianjin Medical University, Tianjin 300070, China
| | - Xin Qiao
- Department of Chemical Biology and Tianjin Key Laboratory of Technologies Enabling Development of Clinical Therapeutics and Diagnostics, School of Pharmacy, Tianjin Medical University, Tianjin 300070, China
| | - Xu Wang
- Department of Chemical Biology and Tianjin Key Laboratory of Technologies Enabling Development of Clinical Therapeutics and Diagnostics, School of Pharmacy, Tianjin Medical University, Tianjin 300070, China
| | - Jing-Yuan Xu
- Department of Chemical Biology and Tianjin Key Laboratory of Technologies Enabling Development of Clinical Therapeutics and Diagnostics, School of Pharmacy, Tianjin Medical University, Tianjin 300070, China
- Key Laboratory of Immune Microenvironment and Disease of the Ministry of Education, Tianjin Medical University, Tianjin 300070, China
| |
Collapse
|
7
|
Lee B, Kim YY, Jeong S, Lee SW, Lee SJ, Rho MC, Kim SH, Lee S. Oleanolic Acid Acetate Alleviates Cisplatin-Induced Nephrotoxicity via Inhibition of Apoptosis and Necroptosis In Vitro and In Vivo. TOXICS 2024; 12:301. [PMID: 38668524 PMCID: PMC11054587 DOI: 10.3390/toxics12040301] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Revised: 04/12/2024] [Accepted: 04/16/2024] [Indexed: 04/29/2024]
Abstract
Cisplatin is a widely used anti-cancer drug for treating solid tumors, but it is associated with severe side effects, including nephrotoxicity. Various studies have suggested that the nephrotoxicity of cisplatin could be overcome; nonetheless, an effective adjuvant drug has not yet been established. Oleanolic acid acetate (OAA), a triterpenoid isolated from Vigna angularis, is commonly used to treat inflammatory and allergic diseases. This study aimed to investigate the protective effects of OAA against cisplatin-induced apoptosis and necroptosis using TCMK-1 cells and a mouse model. In cisplatin-treated TCMK-1 cells, OAA treatment significantly reduced Bax and cleaved-caspase3 expression, whereas it increased Bcl-2 expression. Moreover, in a cisplatin-induced kidney injury mouse model, OAA treatment alleviated weight loss in the body and major organs and also relieved cisplatin-induced nephrotoxicity symptoms. RNA sequencing analysis of kidney tissues identified lipocalin-2 as the most upregulated gene by cisplatin. Additionally, necroptosis-related genes such as receptor-interacting protein kinase (RIPK) and mixed lineage kinase domain-like (MLKL) were identified. In an in vitro study, the phosphorylation of RIPKs and MLKL was reduced by OAA pretreatment in both cisplatin-treated cells and cells boosted via co-treatment with z-VAD-FMK. In conclusion, OAA could protect the kidney from cisplatin-induced nephrotoxicity and may serve as an anti-cancer adjuvant.
Collapse
Affiliation(s)
- Bori Lee
- Functional Biomaterials Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Jeongeup 56212, Republic of Korea; (B.L.); (Y.-Y.K.); (S.J.); (S.W.L.); (S.-J.L.); (M.-C.R.)
| | - Yeon-Yong Kim
- Functional Biomaterials Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Jeongeup 56212, Republic of Korea; (B.L.); (Y.-Y.K.); (S.J.); (S.W.L.); (S.-J.L.); (M.-C.R.)
| | - Seungwon Jeong
- Functional Biomaterials Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Jeongeup 56212, Republic of Korea; (B.L.); (Y.-Y.K.); (S.J.); (S.W.L.); (S.-J.L.); (M.-C.R.)
| | - Seung Woong Lee
- Functional Biomaterials Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Jeongeup 56212, Republic of Korea; (B.L.); (Y.-Y.K.); (S.J.); (S.W.L.); (S.-J.L.); (M.-C.R.)
| | - Seung-Jae Lee
- Functional Biomaterials Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Jeongeup 56212, Republic of Korea; (B.L.); (Y.-Y.K.); (S.J.); (S.W.L.); (S.-J.L.); (M.-C.R.)
| | - Mun-Chual Rho
- Functional Biomaterials Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Jeongeup 56212, Republic of Korea; (B.L.); (Y.-Y.K.); (S.J.); (S.W.L.); (S.-J.L.); (M.-C.R.)
| | - Sang-Hyun Kim
- Cell and Matrix Research Institute, Department of Pharmacology, School of Medicine, Kyungpook National University, Daegu 41944, Republic of Korea
| | - Soyoung Lee
- Functional Biomaterials Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Jeongeup 56212, Republic of Korea; (B.L.); (Y.-Y.K.); (S.J.); (S.W.L.); (S.-J.L.); (M.-C.R.)
| |
Collapse
|
8
|
Bai Y, Qiao Y, Li M, Yang W, Chen H, Wu Y, Zhang H. RIPK1 inhibitors: A key to unlocking the potential of necroptosis in drug development. Eur J Med Chem 2024; 265:116123. [PMID: 38199165 DOI: 10.1016/j.ejmech.2024.116123] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2023] [Revised: 01/02/2024] [Accepted: 01/02/2024] [Indexed: 01/12/2024]
Abstract
Within the field of medical science, there is a great deal of interest in investigating cell death pathways in the hopes of discovering new drugs. Over the past two decades, pharmacological research has focused on necroptosis, a cell death process that has just been discovered. Receptor-interacting protein kinase 1 (RIPK1), an essential regulator in the cell death receptor signalling pathway, has been shown to be involved in the regulation of important events, including necrosis, inflammation, and apoptosis. Therefore, researching necroptosis inhibitors offers novel ways to treat a variety of disorders that are not well-treated by the therapeutic medications now on the market. The research and medicinal potential of RIPK1 inhibitors, a promising class of drugs, are thoroughly examined in this study. The journey from the discovery of Necrostatin-1 (Nec-1) to the recent advancements in RIPK1 inhibitors is marked by significant progress, highlighting the integration of traditional medicinal chemistry approaches with modern technologies like high-throughput screening and DNA-encoded library technology. This review presents a thorough exploration of the development and therapeutic potential of RIPK1 inhibitors, a promising class of compounds. Simultaneously, this review highlights the complex roles of RIPK1 in various pathological conditions and discusses potential inhibitors discovered through diverse pathways, emphasizing their efficacy against multiple disease models, providing significant guidance for the expansion of knowledge about RIPK1 and its inhibitors to develop more selective, potent, and safe therapeutic agents.
Collapse
Affiliation(s)
- Yinliang Bai
- Department of Pharmacy, Lanzhou University Second Hospital, Lanzhou, 730030, China; School of Pharmacy, Lanzhou University, Lanzhou, 730000, China
| | - Yujun Qiao
- Department of Pharmacy, Lanzhou University Second Hospital, Lanzhou, 730030, China
| | - Mingming Li
- Department of Neurology, Lanzhou University Second Hospital, Lanzhou, 730030, China
| | - Wenzhen Yang
- Department of Neurosurgery, Lanzhou University Second Hospital, Lanzhou, 730030, China
| | - Haile Chen
- Department of Pharmacy, Lanzhou University Second Hospital, Lanzhou, 730030, China
| | - Yanqing Wu
- Department of Pharmacy, Lanzhou University Second Hospital, Lanzhou, 730030, China
| | - Honghua Zhang
- Department of Pharmacy, National University of Singapore, Singapore, 117544, Singapore.
| |
Collapse
|
9
|
Zhou Y, Cai Z, Zhai Y, Yu J, He Q, He Y, Jitkaew S, Cai Z. Necroptosis inhibitors: mechanisms of action and therapeutic potential. Apoptosis 2024; 29:22-44. [PMID: 38001341 DOI: 10.1007/s10495-023-01905-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/15/2023] [Indexed: 11/26/2023]
Abstract
Necroptosis is a type of programmed cell death that is morphologically similar to necrosis. This type of cell death is involved in various pathophysiological disorders, including inflammatory, neurodegenerative, infectious, and malignant diseases. Receptor-interacting protein kinase 1 (RIPK1), RIPK3, and mixed lineage kinase domain-like protein (MLKL) pseudokinase constitute the core components of the necroptosis signaling pathway and are considered the most promising targets for therapeutic intervention. The discovery and characterization of necroptosis inhibitors not only accelerate our understanding of the necroptosis signaling pathway but also provide important drug candidates for the treatment of necroptosis-related diseases. Here, we will review recent research progress on necroptosis inhibitors, mechanisms of action and their potential applications for disease treatment.
Collapse
Affiliation(s)
- Yingbo Zhou
- School of Medicine, Tongji University, Shanghai, 200092, China
| | - Zhangtao Cai
- School of Medicine, Tongji University, Shanghai, 200092, China
| | - Yijia Zhai
- School of Medicine, Tongji University, Shanghai, 200092, China
| | - Jintao Yu
- School of Medicine, Tongji University, Shanghai, 200092, China
| | - Qiujing He
- School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China
| | - Yuan He
- Tongji University Cancer Center, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, 200092, China
| | - Siriporn Jitkaew
- Center of Excellence for Cancer and Inflammation, Department of Clinical Chemistry, Faculty of Allied Health Sciences, Chulalongkorn University, Bangkok, 10330, Thailand
| | - Zhenyu Cai
- School of Medicine, Tongji University, Shanghai, 200092, China.
- Tongji University Cancer Center, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, 200092, China.
| |
Collapse
|
10
|
Hao M, Han X, Yao Z, Zhang H, Zhao M, Peng M, Wang K, Shan Q, Sang X, Wu X, Wang L, Lv Q, Yang Q, Bao Y, Kuang H, Zhang H, Cao G. The pathogenesis of organ fibrosis: Focus on necroptosis. Br J Pharmacol 2023; 180:2862-2879. [PMID: 36111431 DOI: 10.1111/bph.15952] [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: 04/14/2022] [Revised: 07/20/2022] [Accepted: 07/28/2022] [Indexed: 11/29/2022] Open
Abstract
Fibrosis is a common process of tissue repair response to multiple injuries in all chronic progressive diseases, which features with excessive deposition of extracellular matrix. Fibrosis can occur in all organs and tends to be nonreversible with the progress of the disease. Different cells types in different organs are involved in the occurrence and development of fibrosis, that is, hepatic stellate cells, pancreatic stellate cells, fibroblasts and myofibroblasts. Various types of programmed cell death, including apoptosis, autophagy, ferroptosis and necroptosis, are closely related to organ fibrosis. Among these programmed cell death types, necroptosis, an emerging regulated cell death type, is regarded as a huge potential target to ameliorate organ fibrosis. In this review, we summarize the role of necroptosis signalling in organ fibrosis and collate the small molecule compounds targeting necroptosis. In addition, we discuss the potential challenges, opportunities and open questions in using necroptosis signalling as a potential target for antifibrotic therapies. LINKED ARTICLES: This article is part of a themed issue on Translational Advances in Fibrosis as a Therapeutic Target. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v180.22/issuetoc.
Collapse
Affiliation(s)
- Min Hao
- School of Pharmacy, Zhejiang Chinese Medical University, Hangzhou, China
| | - Xin Han
- School of Pharmacy, Zhejiang Chinese Medical University, Hangzhou, China
| | - Zhouhui Yao
- School of Pharmacy, Zhejiang Chinese Medical University, Hangzhou, China
| | - Han Zhang
- The Third Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, China
| | - Mengting Zhao
- School of Pharmacy, Zhejiang Chinese Medical University, Hangzhou, China
| | - Mengyun Peng
- School of Pharmacy, Zhejiang Chinese Medical University, Hangzhou, China
| | - Kuilong Wang
- School of Pharmacy, Zhejiang Chinese Medical University, Hangzhou, China
| | - Qiyuan Shan
- School of Pharmacy, Zhejiang Chinese Medical University, Hangzhou, China
| | - Xianan Sang
- School of Pharmacy, Zhejiang Chinese Medical University, Hangzhou, China
| | - Xin Wu
- School of Pharmacy, Zhejiang Chinese Medical University, Hangzhou, China
| | - Lu Wang
- School of Pharmacy, Zhejiang Chinese Medical University, Hangzhou, China
| | - Qiang Lv
- School of Pharmacy, Zhejiang Chinese Medical University, Hangzhou, China
| | - Qiao Yang
- School of Pharmacy, Zhejiang Chinese Medical University, Hangzhou, China
| | - Yini Bao
- School of Pharmacy, Zhejiang Chinese Medical University, Hangzhou, China
| | - Haodan Kuang
- School of Pharmacy, Zhejiang Chinese Medical University, Hangzhou, China
| | - Hongyan Zhang
- Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Hangzhou, China
| | - Gang Cao
- School of Pharmacy, Zhejiang Chinese Medical University, Hangzhou, China
| |
Collapse
|
11
|
Li M, Wei J, Zhu G, Fu S, He X, Hu X, Yu Y, Mou Y, Wang J, You X, Xiao X, Gu T, Ye Z, Zha Y. Quizartinib inhibits necroptosis by targeting receptor-interacting serine/threonine protein kinase 1. FASEB J 2023; 37:e23178. [PMID: 37698367 DOI: 10.1096/fj.202300600rr] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Revised: 08/14/2023] [Accepted: 08/23/2023] [Indexed: 09/13/2023]
Abstract
Systemic inflammatory response syndrome (SIRS), at least in part driven by necroptosis, is characterized by life-threatening multiple organ failure. Blocking the progression of SIRS and consequent multiple organ dysfunction is challenging. Receptor-interacting serine/threonine protein kinase 1 (RIPK1) is an important cell death and inflammatory mediator, making it a potential treatment target in several diseases. Here, using a drug repurposing approach, we show that inhibiting RIPK1 is also an effective treatment for SIRS. We performed cell-based high-throughput drug screening of an US Food and Drug Administration (FDA)-approved drug library that contains 1953 drugs to identify effective inhibitors of necroptotic cell death by SYTOX green staining. Dose-response validation of the top candidate, quizartinib, was conducted in two cell lines of HT-22 and MEFs. The effect of quizartinib on necroptosis-related proteins was evaluated using western blotting, immunoprecipitation, and an in vitro RIPK1 kinase assay. The in vivo effects of quizartinib were assessed in a murine tumor necrosis factor α (TNFα)-induced SIRS model. High-throughput screening identified quizartinib as the top "hit" in the compound library that rescued cells from necroptosis in vitro. Quizartinib inhibited necroptosis by directly inhibiting RIPK1 kinase activity and blocking downstream complex IIb formation. Furthermore, quizartinib protected mice against TNFα-induced SIRS. Quizartinib, as an FDA-approved drug with proven safety and efficacy, was repurposed for targeted inhibition of RIPK1. This work provides essential preclinical data for transferring quizartinib to the treatment of RIPK1-dependent necroptosis-induced inflammatory diseases, including SIRS.
Collapse
Affiliation(s)
- Min Li
- Institute of Neural Regeneration and Repair, The First College of Clinical Medical Science, China Three Gorges University, Yichang, China
- Department of Neurology, The First College of Clinical Medical Science, China Three Gorges University, Yichang, China
- Yichang Central People's Hospital, Yichang, China
| | - Jun Wei
- Institute of Neural Regeneration and Repair, The First College of Clinical Medical Science, China Three Gorges University, Yichang, China
- Department of Neurology, The First College of Clinical Medical Science, China Three Gorges University, Yichang, China
- Yichang Central People's Hospital, Yichang, China
| | - Guofeng Zhu
- Institute of Neural Regeneration and Repair, The First College of Clinical Medical Science, China Three Gorges University, Yichang, China
- Department of Neurology, The First College of Clinical Medical Science, China Three Gorges University, Yichang, China
- Yichang Central People's Hospital, Yichang, China
| | - Shufang Fu
- Yichang Central People's Hospital, Yichang, China
- Department of Paediatrics, The First College of Clinical Medical Science, China Three Gorges University, Yichang, China
| | - Xiaoyan He
- Institute of Neural Regeneration and Repair, The First College of Clinical Medical Science, China Three Gorges University, Yichang, China
- Department of Neurology, The First College of Clinical Medical Science, China Three Gorges University, Yichang, China
- Yichang Central People's Hospital, Yichang, China
| | - Xinqian Hu
- Institute of Neural Regeneration and Repair, The First College of Clinical Medical Science, China Three Gorges University, Yichang, China
- Department of Neurology, The First College of Clinical Medical Science, China Three Gorges University, Yichang, China
- Yichang Central People's Hospital, Yichang, China
| | - Yajie Yu
- Institute of Neural Regeneration and Repair, The First College of Clinical Medical Science, China Three Gorges University, Yichang, China
- Department of Neurology, The First College of Clinical Medical Science, China Three Gorges University, Yichang, China
- Yichang Central People's Hospital, Yichang, China
| | - Yan Mou
- Institute of Neural Regeneration and Repair, The First College of Clinical Medical Science, China Three Gorges University, Yichang, China
- Department of Neurology, The First College of Clinical Medical Science, China Three Gorges University, Yichang, China
- Yichang Central People's Hospital, Yichang, China
| | - Jia Wang
- Institute of Neural Regeneration and Repair, The First College of Clinical Medical Science, China Three Gorges University, Yichang, China
- Department of Neurology, The First College of Clinical Medical Science, China Three Gorges University, Yichang, China
- Yichang Central People's Hospital, Yichang, China
| | - Xiaoling You
- Institute of Neural Regeneration and Repair, The First College of Clinical Medical Science, China Three Gorges University, Yichang, China
- Department of Neurology, The First College of Clinical Medical Science, China Three Gorges University, Yichang, China
- Yichang Central People's Hospital, Yichang, China
| | - Xin Xiao
- Institute of Neural Regeneration and Repair, The First College of Clinical Medical Science, China Three Gorges University, Yichang, China
- Department of Neurology, The First College of Clinical Medical Science, China Three Gorges University, Yichang, China
- Yichang Central People's Hospital, Yichang, China
| | - Tanrong Gu
- Institute of Neural Regeneration and Repair, The First College of Clinical Medical Science, China Three Gorges University, Yichang, China
- Department of Neurology, The First College of Clinical Medical Science, China Three Gorges University, Yichang, China
- Yichang Central People's Hospital, Yichang, China
| | - Zhi Ye
- Institute of Neural Regeneration and Repair, The First College of Clinical Medical Science, China Three Gorges University, Yichang, China
- Department of Neurology, The First College of Clinical Medical Science, China Three Gorges University, Yichang, China
- Yichang Central People's Hospital, Yichang, China
| | - Yunhong Zha
- Institute of Neural Regeneration and Repair, The First College of Clinical Medical Science, China Three Gorges University, Yichang, China
- Department of Neurology, The First College of Clinical Medical Science, China Three Gorges University, Yichang, China
- Yichang Central People's Hospital, Yichang, China
| |
Collapse
|
12
|
Xu B, Fang J, Wang J, Jin X, Liu S, Song K, Wang P, Liu J, Liu S. Inhibition of autophagy and RIP1/RIP3/MLKL-mediated necroptosis by edaravone attenuates blood spinal cord barrier disruption following spinal cord injury. Biomed Pharmacother 2023; 165:115165. [PMID: 37459660 DOI: 10.1016/j.biopha.2023.115165] [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: 05/03/2023] [Revised: 07/09/2023] [Accepted: 07/11/2023] [Indexed: 08/17/2023] Open
Abstract
The disruption of the blood spinal cord barrier (BSCB) after spinal cord injury (SCI) can trigger secondary tissue damage. Edaravone is likely to protect the BSCB as a free radical scavenger, whereas it has been rarely reported thus far. In this study, the protective effect of edaravone was investigated with the use of compression spinal cord injured rats and human brain microvascular endothelial cells (HBMECs) injury. As indicated by the result of this study, edaravone treatment facilitated functional recovery after rats were subjected to SCI, ameliorated the vascular damage, and up-regulated the expression of BSCB-associated proteins. In vitro results, edaravone improved HBMECs viability, restored intercellular junctions, and promoted cellular angiogenic activities. It is noteworthy that autophagy was activated and RIP1/RIP3/MLKL phosphorylation was notably up-regulated. However, edaravone treatment exhibited the capability of mitigating above-mentioned tendency in vivo and in vitro. Moreover, rapamycin (Rapa) treatment deteriorated the protective effect of edaravone while aggravating the phosphorylation of RIP1/RIP3/MLKL expression. In the model of necrotic activator-induced HBMECs, autophagic expression was increased, whereas edaravone prevented autophagy and phosphorylation of RIP1/RIP3/MLKL. In general, our results suggested that edaravone is capable of reducing the destruction of BSCB and promoting functional recovery after SCI. The possible underlying mechanism is that edaravone is capable of protecting angiogenic activity and improving autophagy and the phosphorylation of RIP1/RIP3/MLKL, as well as their mutual deterioration. Accordingly, edaravone can be a favorable option for the treatment of SCI.
Collapse
Affiliation(s)
- Bo Xu
- Department of Orthopaedics, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Jiaqi Fang
- Department of Orthopaedics, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Jianguang Wang
- Department of Orthopaedics, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Xuehan Jin
- Department of Orthopaedics, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Shengfu Liu
- Department of Orthopaedics, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Kaihang Song
- Department of Orthopaedics, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Ping Wang
- Department of Operating Room, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, China.
| | - Junjian Liu
- Department of Orthopaedics, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, China.
| | - Shuhao Liu
- Department of Orthopaedics, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, China.
| |
Collapse
|
13
|
Piao L, Wu D, Rui C, Yang Y, Liu S, Liu J, Jin Z, Zhang H, Feng X, Bai L. The Bcr-Abl inhibitor DCC-2036 inhibits necroptosis and ameliorates osteoarthritis by targeting RIPK1 and RIPK3 kinases. Biomed Pharmacother 2023; 161:114528. [PMID: 36931029 DOI: 10.1016/j.biopha.2023.114528] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Revised: 03/08/2023] [Accepted: 03/11/2023] [Indexed: 03/18/2023] Open
Abstract
Osteoarthritis (OA) is a chronic progressive degenerative joint disease. Owing to its complex pathogenesis, OA treatment is typically challenging. Necroptosis is a form of programmed cell death mainly mediated by the serine/threonine kinases, RIPK1 and RIPK3, and mixed lineage kinase-like domain (MLKL). In this study, we found that the multi-targeted kinase inhibitor DCC-2036 can inhibit TSZ (TNF-α, Smac mimetic, and z-VAD-FMK)-induced necroptosis of chondrocytes and synovial fibroblast cells (SFs). In addition, we found that oral DCC-2036 inhibited chondrocyte damage in a rat model of OA induced by intra-articular injection of monosodium iodoacetate (MIA). A mechanistic study showed that DCC-2036 directly inhibited the activities of RIPK1 and RIPK3 kinases to block necroptosis, inhibiting the inflammatory response and protecting chondrocytes. In summary, our research suggests that DCC-2036, a new necroptosis inhibitor targeting RIPK1 and RIPK3 kinase activity, may be useful for the clinical treatment of OA and provides a new direction for the research and treatment of OA.
Collapse
Affiliation(s)
- Longhuan Piao
- Department of Orthopedic Surgery, Shengjing Hospital of China Medical University, Shenyang, Liaoning 110004, China
| | - Dong Wu
- Department of Orthopedic Surgery, Shengjing Hospital of China Medical University, Shenyang, Liaoning 110004, China
| | - Chunhua Rui
- Cancer Institute, Fudan University Shanghai Cancer Center, Shanghai 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Yue Yang
- Department of Orthopedic Surgery, Shengjing Hospital of China Medical University, Shenyang, Liaoning 110004, China
| | - Shuai Liu
- Department of Orthopedic Surgery, Shengjing Hospital of China Medical University, Shenyang, Liaoning 110004, China
| | - Jiabao Liu
- Department of Orthopedic Surgery, Shengjing Hospital of China Medical University, Shenyang, Liaoning 110004, China
| | - Zhuangzhuang Jin
- Department of Emergency Medicine, Shengjing Hospital of China Medical University, Shenyang, Liaoning 110004, China
| | - He Zhang
- Department of Orthopedic Surgery, Shengjing Hospital of China Medical University, Shenyang, Liaoning 110004, China
| | - Xinyuan Feng
- Department of Orthopedic Surgery, Shengjing Hospital of China Medical University, Shenyang, Liaoning 110004, China
| | - Lunhao Bai
- Department of Orthopedic Surgery, Shengjing Hospital of China Medical University, Shenyang, Liaoning 110004, China.
| |
Collapse
|
14
|
Wang Q, Ye Q, Xi X, Cao X, Wang X, Zhang M, Xu Y, Deng T, Deng X, Zhang G, Xiao C. KW2449 ameliorates collagen-induced arthritis by inhibiting RIPK1-dependent necroptosis. Front Immunol 2023; 14:1135014. [PMID: 36993980 PMCID: PMC10040599 DOI: 10.3389/fimmu.2023.1135014] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2022] [Accepted: 02/28/2023] [Indexed: 03/14/2023] Open
Abstract
ObjectiveNecroptosis has recently been found to be associated with the pathogenesis of many autoimmune diseases, including rheumatoid arthritis (RA). This study was undertaken to explore the role of RIPK1-dependent necroptosis in the pathogenesis of RA and the potential new treatment options.MethodsThe plasma levels of receptor-interacting protein kinase 1 (RIPK1) and mixed lineage kinase domain-like pseudokinase (MLKL) in 23 controls and 42 RA patients were detected by ELISA. Collagen-induced arthritis (CIA) rats were treated with KW2449 by gavage for 28 days. Arthritis index score, H&E staining, and Micro-CT analysis were used to evaluate joint inflammation. The levels of RIPK1-dependent necroptosis related proteins and inflammatory cytokines were detected by qRT-PCR, ELISA and Western blot, and the cell death morphology was detected by flow cytometry analysis and high-content imaging analysis.ResultsThe plasma levels of RIPK1 and MLKL in RA patients were higher than those in healthy people, and were positively correlated with the severity of RA. KW2449 could reduce joint swelling, joint bone destruction, tissue damage, and the plasma levels of inflammatory cytokines in CIA rats. Lipopolysaccharide combined with zVAD (LZ) could induce necroptosis in RAW 264.7 cells, which could be reduced by KW2449. RIPK1-dependent necroptosis related proteins and inflammatory factors increased after LZ induction and decreased after KW2449 treatment or knockdown of RIPK1.ConclusionThese findings suggest that the overexpression of RIPK1 is positively correlated with the severity of RA. KW2449, as a small molecule inhibitor targeting RIPK1, has the potential to be a therapeutic strategy for RA treatment by inhibiting RIPK1-dependent necroptosis.
Collapse
Affiliation(s)
- Qiong Wang
- Beijing University of Chinese Medicine, China-Japan Friendship Hospital Clinical Medicine, Beijing, China
- Institute of Clinical Medicine, China-Japan Friendship Hospital, Beijing, China
| | - Qinbin Ye
- Beijing University of Chinese Medicine, China-Japan Friendship Hospital Clinical Medicine, Beijing, China
- Institute of Clinical Medicine, China-Japan Friendship Hospital, Beijing, China
| | - Xiaoyu Xi
- Beijing University of Chinese Medicine, China-Japan Friendship Hospital Clinical Medicine, Beijing, China
- Institute of Clinical Medicine, China-Japan Friendship Hospital, Beijing, China
| | - Xiaoxue Cao
- Institute of Clinical Medicine, China-Japan Friendship Hospital, Beijing, China
- Graduate School of Peking Union Medical College, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xing Wang
- Beijing University of Chinese Medicine, China-Japan Friendship Hospital Clinical Medicine, Beijing, China
- Institute of Clinical Medicine, China-Japan Friendship Hospital, Beijing, China
| | - Mengxiao Zhang
- Institute of Clinical Medicine, China-Japan Friendship Hospital, Beijing, China
| | - Yuan Xu
- Beijing Key Laboratory of Research of Chinese Medicine on Prevention and Treatment for Major Diseases, Experimental Research Center, China Academy of Chinese Medical Sciences, Beijing, China
| | - Tingting Deng
- Institute of Clinical Medicine, China-Japan Friendship Hospital, Beijing, China
| | - Xiaobing Deng
- Department of Drug Discovery, Double-Crane Run Therapeutics, Beijing, China
| | - Guoqiang Zhang
- Department of Emergency, China-Japan Friendship Hospital, Beijing, China
- *Correspondence: Guoqiang Zhang, ; Cheng Xiao,
| | - Cheng Xiao
- Institute of Clinical Medicine, China-Japan Friendship Hospital, Beijing, China
- Department of Emergency, China-Japan Friendship Hospital, Beijing, China
- *Correspondence: Guoqiang Zhang, ; Cheng Xiao,
| |
Collapse
|
15
|
Abstract
Cell death, particularly that of tubule epithelial cells, contributes critically to the pathophysiology of kidney disease. A body of evidence accumulated over the past 15 years has ascribed a central pathophysiological role to a particular form of regulated necrosis, termed necroptosis, to acute tubular necrosis, nephron loss and maladaptive renal fibrogenesis. Unlike apoptosis, which is a non-immunogenic process, necroptosis results in the release of cellular contents and cytokines, which triggers an inflammatory response in neighbouring tissue. This necroinflammatory environment can lead to severe organ dysfunction and cause lasting tissue injury in the kidney. Despite evidence of a link between necroptosis and various kidney diseases, there are no available therapeutic options to target this process. Greater understanding of the molecular mechanisms, triggers and regulators of necroptosis in acute and chronic kidney diseases may identify shortcomings in current approaches to therapeutically target necroptosis regulators and lead to the development of innovative therapeutic approaches.
Collapse
|
16
|
Shi K, Zhang J, Zhou E, Wang J, Wang Y. Small-Molecule Receptor-Interacting Protein 1 (RIP1) Inhibitors as Therapeutic Agents for Multifaceted Diseases: Current Medicinal Chemistry Insights and Emerging Opportunities. J Med Chem 2022; 65:14971-14999. [DOI: 10.1021/acs.jmedchem.2c01518] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
- Kunyu Shi
- Targeted Tracer Research and Development Laboratory, Institute of Respiratory Health, Frontiers Science Center for Disease-related Molecular Network, Joint Research Institution of Altitude Health, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041 Sichuan, China
| | - Jifa Zhang
- Targeted Tracer Research and Development Laboratory, Institute of Respiratory Health, Frontiers Science Center for Disease-related Molecular Network, Joint Research Institution of Altitude Health, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041 Sichuan, China
- Precision Medicine Key Laboratory of Sichuan Province & Precision Medicine Research Center, West China Hospital, Sichuan University, Chengdu, 610041 Sichuan, China
- Tianfu Jincheng Laboratory, Chengdu, 610041 Sichuan, China
| | - Enda Zhou
- West China School of Pharmacy, Sichuan University, Chengdu, 610041 Sichuan, China
| | - Jiaxing Wang
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, Memphis, Tennessee 38163, United States
| | - Yuxi Wang
- Targeted Tracer Research and Development Laboratory, Institute of Respiratory Health, Frontiers Science Center for Disease-related Molecular Network, Joint Research Institution of Altitude Health, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041 Sichuan, China
- Precision Medicine Key Laboratory of Sichuan Province & Precision Medicine Research Center, West China Hospital, Sichuan University, Chengdu, 610041 Sichuan, China
- Tianfu Jincheng Laboratory, Chengdu, 610041 Sichuan, China
| |
Collapse
|
17
|
Fan Y, Lu J, Yu Z, Qu X, Guan S. 1,3-Dichloro-2-propanol-Induced Renal Tubular Cell Necroptosis through the ROS/RIPK3/MLKL Pathway. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2022; 70:10847-10857. [PMID: 36000575 DOI: 10.1021/acs.jafc.2c02619] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
1,3-Dichloro-2-propanol (1,3-DCP), as a food pollutant, exists in a variety of foods. Studies have shown that it has nephrotoxicity. In the study, we found that 1,3-DCP caused renal injury with necroptosis in C57BL/6J mice. The mechanism of 1,3-DCP-caused nephrotoxicity was further explored in NRK-52E cells in vitro. We found that 1,3-DCP caused cell necroptosis with the increase in lactate dehydrogenase (LDH) levels and the expressions of RIPK3 and MLKL. But pretreatment with a ROS inhibitor N-acetyl-l-cysteine (NAC), a RIPK3 inhibitor GSK'872, or RIPK3 gene silencing alleviated 1,3-DCP-induced cell necroptosis. The data indicated that 1,3-DCP induced necroptosis through the ROS/RIPK3/MLKL pathway in NRK-52E cells. In further mechanistic studies, we explored how 1,3-DCP induced ROS production. We found that 1,3-DCP inhibited the expressions of nuclear and cytoplasmic Nrf2. But pretreatment with an Nrf2 activator dimethyl fumarate (DMF) up-regulated the expressions of nuclear and cytoplasmic Nrf2 and down-regulated ROS levels and RIPK3 and MLKL expressions. We also examined the effects of mitophagy on 1,3-DCP-induced ROS. The data manifested that 1,3-DCP suppressed mitophagy in NRK-52E cells by decreasing LC3-II, Pink1, and Parkin levels, increasing p62 levels, and decreasing colocalization of LC3 and Mito-Tracker Red. Pretreatment with an autophagy activator rapamycin (Rapa) decreased 1,3-DCP-induced ROS. Taken together, our data identified that 1,3-DCP caused renal necroptosis through the ROS/RIPK3/MLKL pathway.
Collapse
Affiliation(s)
- Yong Fan
- College of Food Science and Engineering, Jilin University, Changchun, Jilin 130062, People's Republic of China
| | - Jing Lu
- College of Food Science and Engineering, Jilin University, Changchun, Jilin 130062, People's Republic of China
- Key Laboratory of Zoonosis, Ministry of Education College of Veterinary Medicine, Jilin University, Changchun, Jilin 130062, People's Republic of China
| | - Zelin Yu
- College of Food Science and Engineering, Jilin University, Changchun, Jilin 130062, People's Republic of China
| | - Xiao Qu
- College of Food Science and Engineering, Jilin University, Changchun, Jilin 130062, People's Republic of China
| | - Shuang Guan
- College of Food Science and Engineering, Jilin University, Changchun, Jilin 130062, People's Republic of China
- Key Laboratory of Zoonosis, Ministry of Education College of Veterinary Medicine, Jilin University, Changchun, Jilin 130062, People's Republic of China
| |
Collapse
|
18
|
Fang K, Tang DS, Yan CS, Ma J, Cheng L, Li Y, Wang G. Comprehensive Analysis of Necroptosis in Pancreatic Cancer for Appealing its Implications in Prognosis, Immunotherapy, and Chemotherapy Responses. Front Pharmacol 2022; 13:862502. [PMID: 35662734 PMCID: PMC9157651 DOI: 10.3389/fphar.2022.862502] [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: 01/26/2022] [Accepted: 04/19/2022] [Indexed: 11/13/2022] Open
Abstract
Objective: Necroptosis represents a new target for cancer immunotherapy and is considered a form of cell death that overcomes apoptosis resistance and enhances tumor immunogenicity. Herein, we aimed to determine necroptosis subtypes and investigate the roles of necroptosis in pancreatic cancer therapy. Methods: Based on the expression of prognostic necroptosis genes in pancreatic cancer samples from TCGA and ICGC cohorts, a consensus clustering approach was implemented for robustly identifying necroptosis subtypes. Immunogenic features were evaluated according to immune cell infiltrations, immune checkpoints, HLA molecules, and cancer-immunity cycle. The sensitivity to chemotherapy agents was estimated using the pRRophetic package. A necroptosis-relevant risk model was developed with a multivariate Cox regression analysis. Results: Five necroptosis subtypes were determined for pancreatic cancer (C1∼C5) with diverse prognosis, immunogenic features, and chemosensitivity. In particular, C4 and C5 presented favorable prognosis and weakened immunogenicity; C2 had high immunogenicity; C1 had undesirable prognosis and high genetic mutations. C5 was the most sensitive to known chemotherapy agents (cisplatin, gemcitabine, docetaxel, and paclitaxel), while C4 displayed resistance to aforementioned agents. The necroptosis-relevant risk model could accurately predict prognosis, immunogenicity, and chemosensitivity. Conclusion: Our findings provided a conceptual framework for comprehending necroptosis in pancreatic cancer biology. Future work is required for evaluating its relevance in the design of combined therapeutic regimens and guiding the best choice for immuno- and chemotherapy.
Collapse
Affiliation(s)
- Kun Fang
- Department of Pancreatic and Biliary Surgery, First Affiliated Hospital of Harbin Medical University, Harbin, China.,Key Laboratory of Hepatosplenic Surgery, First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - De-Sheng Tang
- Department of Pancreatic and Biliary Surgery, First Affiliated Hospital of Harbin Medical University, Harbin, China.,Key Laboratory of Hepatosplenic Surgery, First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Chang-Sheng Yan
- Department of Pancreatic and Biliary Surgery, First Affiliated Hospital of Harbin Medical University, Harbin, China.,Key Laboratory of Hepatosplenic Surgery, First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Jiamin Ma
- Department of Pancreatic and Biliary Surgery, First Affiliated Hospital of Harbin Medical University, Harbin, China.,Key Laboratory of Hepatosplenic Surgery, First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Long Cheng
- Department of Pancreatic and Biliary Surgery, First Affiliated Hospital of Harbin Medical University, Harbin, China.,Key Laboratory of Hepatosplenic Surgery, First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Yilong Li
- Department of Pancreatic and Biliary Surgery, First Affiliated Hospital of Harbin Medical University, Harbin, China.,Key Laboratory of Hepatosplenic Surgery, First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Gang Wang
- Department of Pancreatic and Biliary Surgery, First Affiliated Hospital of Harbin Medical University, Harbin, China.,Key Laboratory of Hepatosplenic Surgery, First Affiliated Hospital of Harbin Medical University, Harbin, China
| |
Collapse
|
19
|
Wu Y, Wu J, Lin Z, Wang Q, Li Y, Wang A, Shan X, Liu J. Administration of a Probiotic Mixture Ameliorates Cisplatin-Induced Mucositis and Pica by Regulating 5-HT in Rats. J Immunol Res 2021; 2021:9321196. [PMID: 34568500 PMCID: PMC8461230 DOI: 10.1155/2021/9321196] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Revised: 08/18/2021] [Accepted: 08/21/2021] [Indexed: 12/24/2022] Open
Abstract
Probiotic-based therapies have been shown to be beneficial for chemotherapy-induced mucositis. Previous research has demonstrated that a probiotic mixture (Bifidobacterium brevis, Lactobacillus acidophilus, Lactobacillus casei, and Streptococcus thermophilus) can ameliorate chemotherapy-induced mucositis and dysbiosis in rats, but the underlying mechanism has not been completely elucidated. We aimed to determine the inhibitory effects of the probiotic mixture on cisplatin-induced mucositis and pica and the underlying mechanism, focusing on the levels of 5-hydroxytryptamine (5-HT, serotonin) regulated by the gut microbiota. A rat model of mucositis and pica was established by daily intraperitoneal injection of cisplatin (6 mg/kg) for 3 days. In the probiotic+cisplatin group, predaily intragastric injection of the probiotic mixture (1 × 109 CFU/kg BW) was administrated for 1 week before cisplatin injection. This was then followed by further daily probiotic injections for 6 days. Histopathology, pro-/anti-inflammatory cytokines, oxidative status, and 5-HT levels were assessed on days 3 and 6. The structure of the gut microbiota was analyzed by 16S rRNA gene sequencing and quantitative PCR. Additionally, 5-HT levels in enterochromaffin (EC) cells (RIN-14B cell line) treated with cisplatin and/or various probiotic bacteria were also determined. The probiotic mixture significantly attenuated kaolin consumption, inflammation, oxidative stress, and the increase in 5-HT concentrations in rats with cisplatin-induced intestinal mucositis and pica. Cisplatin markedly increased the relative abundances of Enterobacteriaceae_other, Blautia, Clostridiaceae_other, and members of Clostridium clusters IV and XIVa. These levels were significantly restored by the probiotic mixture. Importantly, most of the genera increased by cisplatin were significantly positively correlated with colonic 5-HT. Furthermore, in vitro, the probiotic mixture had direct inhibitory effects on the 5-HT secretion by EC cells. The probiotic mixture protects against cisplatin-induced intestine injury, exhibiting both anti-inflammatory and antiemetic properties. These results were closely related to the reestablishment of intestinal microbiota ecology and normalization of the dysbiosis-driven 5-HT overproduction.
Collapse
Affiliation(s)
- Yuanhang Wu
- Department of Medical Oncology, The First Affiliated Hospital, Dalian Medical University, Dalian, China
| | - Jianlin Wu
- Affiliated Zhongshan Hospital of Dalian University, Dalian, China
| | - Zhikun Lin
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Qian Wang
- Liaoning CapitalBio Technology Co., Ltd., Dalian, China
| | - Ying Li
- Department of Medical Oncology, The First Affiliated Hospital, Dalian Medical University, Dalian, China
| | - Aman Wang
- Department of Medical Oncology, The First Affiliated Hospital, Dalian Medical University, Dalian, China
| | - Xiu Shan
- Department of Medical Oncology, The First Affiliated Hospital, Dalian Medical University, Dalian, China
| | - Jiwei Liu
- Department of Medical Oncology, The First Affiliated Hospital, Dalian Medical University, Dalian, China
| |
Collapse
|