1
|
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
|
2
|
Chen KQ, Wang SZ, Lei HB, Liu X. Necrostatin-1: a promising compound for neurological disorders. Front Cell Neurosci 2024; 18:1408364. [PMID: 38994325 PMCID: PMC11236683 DOI: 10.3389/fncel.2024.1408364] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Accepted: 06/17/2024] [Indexed: 07/13/2024] Open
Abstract
Necrostatin-1, a small molecular alkaloid, was identified as an inhibitor of necroptosis in 2005. Investigating the fundamental mechanism of Necrostatin-1 and its role in various diseases is of great significance for scientific and clinical research. Accumulating evidence suggests that Necrostatin-1 plays a crucial role in numerous neurological disorders. This review aims to provide a comprehensive overview of the potential functions of Necrostatin-1 in various neurological disorders, offering valuable insights for future research.
Collapse
Affiliation(s)
- Ke-Qian Chen
- Department of Clinical Pharmacy, Xiangtan Central Hospital, Xiangtan, China
| | - Shu-Zhi Wang
- Institute of Pharmacy and Pharmacology, School of Pharmaceutical Sciences, University of South China, Hengyang, China
| | - Hai-Bo Lei
- Department of Clinical Pharmacy, Xiangtan Central Hospital, Xiangtan, China
| | - Xiang Liu
- Department of Clinical Pharmacy, Xiangtan Central Hospital, Xiangtan, China
| |
Collapse
|
3
|
Chandra P, Patra U, Mukhopadhyay U, Mukherjee A, Halder P, Koley H, Chawla-Sarkar M. Rotavirus non-structural protein 4 usurps host cellular RIPK1-RIPK3 complex to induce MLKL-dependent necroptotic cell death. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2024; 1871:119745. [PMID: 38719029 DOI: 10.1016/j.bbamcr.2024.119745] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Revised: 04/11/2024] [Accepted: 04/16/2024] [Indexed: 05/14/2024]
Abstract
The dynamic interface between invading viral pathogens and programmed cell death (PCD) of the host is a finely regulated process. Host cellular demise at the end of the viral life cycle ensures the release of progeny virions to initiate new infection cycles. Rotavirus (RV), a diarrheagenic virus with double-stranded RNA genome, has been reported to trigger different types of PCD such as apoptosis and pyroptosis in a highly regulated way to successfully disseminate progeny virions. Recently our lab also showed that induction of MLKL-driven programmed necroptosis by RV. However, the host cellular machinery involved in RV-induced necroptosis and the upstream viral trigger responsible for it remained unaddressed. In the present study, the signalling upstream of MLKL-driven necroptosis has been delineated where the involvement of Receptor interacting serine/threonine kinase 3 (RIPK3) and 1 (RIPK1) from the host side and RV non-structural protein 4 (NSP4) as the viral trigger for necroptosis has been shown. Interestingly, RV-NSP4 was found to be an integral component of the necrosome complex by interacting with RIPK1, thereby bypassing the requirement of RIPK1 kinase activity. Subsequently, NSP4-driven elevated cytosolic Ca2+ concentration and Ca2+-binding to NSP4 lead further to RHIM domain-dependent RIPK1-RIPK3 interaction, RIPK3-dependent MLKL phosphorylation, and eventual necroptosis. Overall, this study presents the interplay between RV-NSP4 and the host cellular necrosome complex to induce necroptotic death of host cells.
Collapse
Affiliation(s)
- Pritam Chandra
- Division of Virology, ICMR-National Institute of Cholera and Enteric Diseases, Kolkata, West Bengal, India
| | - Upayan Patra
- Institute of Biochemistry II, Faculty of Medicine, Goethe University, Frankfurt, Germany
| | - Urbi Mukhopadhyay
- European Molecular Biology Laboratory, 71 Av. Des Martyrs, 38000 Grenoble, France
| | - Arpita Mukherjee
- Division of Virology, ICMR-National Institute of Cholera and Enteric Diseases, Kolkata, West Bengal, India
| | - Prolay Halder
- Division of Bacteriology, ICMR-National Institute of Cholera and Enteric Diseases, Kolkata, West Bengal, India
| | - Hemanta Koley
- Division of Bacteriology, ICMR-National Institute of Cholera and Enteric Diseases, Kolkata, West Bengal, India
| | - Mamta Chawla-Sarkar
- Division of Virology, ICMR-National Institute of Cholera and Enteric Diseases, Kolkata, West Bengal, India.
| |
Collapse
|
4
|
Zheng LY, Duan Y, He PY, Wu MY, Wei ST, Du XH, Yao RQ, Yao YM. Dysregulated dendritic cells in sepsis: functional impairment and regulated cell death. Cell Mol Biol Lett 2024; 29:81. [PMID: 38816685 PMCID: PMC11140885 DOI: 10.1186/s11658-024-00602-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Accepted: 05/21/2024] [Indexed: 06/01/2024] Open
Abstract
Sepsis is defined as life-threatening organ dysfunction caused by a dysregulated host response to infection. Studies have indicated that immune dysfunction plays a central role in the pathogenesis of sepsis. Dendritic cells (DCs) play a crucial role in the emergence of immune dysfunction in sepsis. The major manifestations of DCs in the septic state are abnormal functions and depletion in numbers, which are linked to higher mortality and vulnerability to secondary infections in sepsis. Apoptosis is the most widely studied pathway of number reduction in DCs. In the past few years, there has been a surge in studies focusing on regulated cell death (RCD). This emerging field encompasses various forms of cell death, such as necroptosis, pyroptosis, ferroptosis, and autophagy-dependent cell death (ADCD). Regulation of DC's RCD can serve as a possible therapeutic focus for the treatment of sepsis. Throughout time, numerous tactics have been devised and effectively implemented to improve abnormal immune response during sepsis progression, including modifying the functions of DCs and inhibiting DC cell death. In this review, we provide an overview of the functional impairment and RCD of DCs in septic states. Also, we highlight recent advances in targeting DCs to regulate host immune response following septic challenge.
Collapse
Affiliation(s)
- Li-Yu Zheng
- Translational Medicine Research Center, Medical Innovation Research Division of the Chinese PLA General Hospital, 28 Fuxing Road, Haidian District, Beijing, 100853, China
| | - Yu Duan
- Department of Critical Care Medicine, Affiliated Chenzhou Hospital (the First People's Hospital of Chenzhou), Southern Medical University, Chenzhou, 423000, China
| | - Peng-Yi He
- Translational Medicine Research Center, Medical Innovation Research Division of the Chinese PLA General Hospital, 28 Fuxing Road, Haidian District, Beijing, 100853, China
| | - Meng-Yao Wu
- Translational Medicine Research Center, Medical Innovation Research Division of the Chinese PLA General Hospital, 28 Fuxing Road, Haidian District, Beijing, 100853, China
| | - Shu-Ting Wei
- Translational Medicine Research Center, Medical Innovation Research Division of the Chinese PLA General Hospital, 28 Fuxing Road, Haidian District, Beijing, 100853, China
| | - Xiao-Hui Du
- Department of General Surgery, The First Medical Center of Chinese PLA General Hospital, 28 Fuxing Road, Haidian District, Beijing, 100853, China.
| | - Ren-Qi Yao
- Translational Medicine Research Center, Medical Innovation Research Division of the Chinese PLA General Hospital, 28 Fuxing Road, Haidian District, Beijing, 100853, China.
- Department of General Surgery, The First Medical Center of Chinese PLA General Hospital, 28 Fuxing Road, Haidian District, Beijing, 100853, China.
| | - Yong-Ming Yao
- Translational Medicine Research Center, Medical Innovation Research Division of the Chinese PLA General Hospital, 28 Fuxing Road, Haidian District, Beijing, 100853, China.
| |
Collapse
|
5
|
Li C, Yu Y, Zhu S, Hu Y, Ling X, Xu L, Zhang H, Guo K. The emerging role of regulated cell death in ischemia and reperfusion-induced acute kidney injury: current evidence and future perspectives. Cell Death Discov 2024; 10:216. [PMID: 38704372 PMCID: PMC11069531 DOI: 10.1038/s41420-024-01979-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2023] [Revised: 04/14/2024] [Accepted: 04/18/2024] [Indexed: 05/06/2024] Open
Abstract
Renal ischemia‒reperfusion injury (IRI) is one of the main causes of acute kidney injury (AKI), which is a potentially life-threatening condition with a high mortality rate. IRI is a complex process involving multiple underlying mechanisms and pathways of cell injury and dysfunction. Additionally, various types of cell death have been linked to IRI, including necroptosis, apoptosis, pyroptosis, and ferroptosis. These processes operate differently and to varying degrees in different patients, but each plays a role in the various pathological conditions of AKI. Advances in understanding the underlying pathophysiology will lead to the development of new therapeutic approaches that hold promise for improving outcomes for patients with AKI. This review provides an overview of the recent research on the molecular mechanisms and pathways underlying IRI-AKI, with a focus on regulated cell death (RCD) forms such as necroptosis, pyroptosis, and ferroptosis. Overall, targeting RCD shows promise as a potential approach to treating IRI-AKI.
Collapse
Affiliation(s)
- Chenning Li
- Department of Anesthesiology, Zhongshan Hospital, Shanghai, China
- Shanghai Key Laboratory of Perioperative Stress and Protection, Shanghai, China
| | - Ying Yu
- Department of Anesthesiology, Zhongshan Hospital, Shanghai, China
- Shanghai Key Laboratory of Perioperative Stress and Protection, Shanghai, China
| | - Shuainan Zhu
- Department of Anesthesiology, Zhongshan Hospital, Shanghai, China
- Shanghai Key Laboratory of Perioperative Stress and Protection, Shanghai, China
| | - Yan Hu
- Department of Anesthesiology, Zhongshan Hospital, Shanghai, China
- Shanghai Key Laboratory of Perioperative Stress and Protection, Shanghai, China
| | - Xiaomin Ling
- Department of Anesthesiology, Zhongshan Hospital, Shanghai, China
- Shanghai Key Laboratory of Perioperative Stress and Protection, Shanghai, China
| | - Liying Xu
- Department of Anesthesiology, Zhongshan Hospital, Shanghai, China
- Shanghai Key Laboratory of Perioperative Stress and Protection, Shanghai, China
| | - Hao Zhang
- Department of Anesthesiology, Zhongshan Hospital, Shanghai, China.
- Shanghai Key Laboratory of Perioperative Stress and Protection, Shanghai, China.
| | - Kefang Guo
- Department of Anesthesiology, Zhongshan Hospital, Shanghai, China.
- Shanghai Key Laboratory of Perioperative Stress and Protection, Shanghai, China.
| |
Collapse
|
6
|
Zhang C, Chen Y, Li Y, Shi N, Teng Y, Li N, Tang M, Ma Z, Deng D, Chen L. Discovery of 4-amino-1,6-dihydro-7H-pyrrolo[2,3-d]pyridazin-7-one derivatives as potential receptor-interacting serine/threonine-protein kinase 1 (RIPK1) inhibitors. Eur J Med Chem 2024; 265:116076. [PMID: 38171150 DOI: 10.1016/j.ejmech.2023.116076] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Revised: 12/12/2023] [Accepted: 12/18/2023] [Indexed: 01/05/2024]
Abstract
Receptor-interacting serine/threonine-protein kinase 1 (RIPK1) is an important regulatory factor in the necroptosis signaling pathway, and is considered an attractive therapeutic target for treating multiple inflammatory diseases. Herein, we describe the design, synthesis, and structure-activity relationships of 4-amino-1,6-dihydro-7H-pyrrolo [2,3-d]pyridazin-7-one derivatives as RIPK1 inhibitors. Among them, 13c showed favorable RIPK1 kinase inhibition activity with an IC50 value of 59.8 nM, and high RIPK1 binding affinity compared with other regulatory kinases of necroptosis (RIPK1 Kd = 3.5 nM, RIPK3 Kd = 1700 nM, and MLKL Kd > 30,000 nM). 13c efficiently blocked TNFα-induced necroptosis in both human and murine cells (EC50 = 1.06-4.58 nM), and inhibited TSZ-induced phosphorylation of the RIPK1/RIPK3/MLKL pathway. In liver microsomal assay studies, the clearance rate and half-life of 13c were 18.40 mL/min/g and 75.33 min, respectively. 13c displayed acceptable pharmacokinetic characteristics, with oral bioavailability of 59.55%. In TNFα-induced systemic inflammatory response syndrome, pretreatment with 13c could effectively protect mice from loss of body temperature and death. Overall, these compounds are promising candidates for future optimization studies.
Collapse
Affiliation(s)
- Chufeng Zhang
- State Key Laboratory of Biotherapy and Cancer Center and Collaborative Innovation Center of Biotherapy, West China Hospital of Sichuan University, Chengdu, 610041, China
| | - Yulian Chen
- State Key Laboratory of Biotherapy and Cancer Center and Collaborative Innovation Center of Biotherapy, West China Hospital of Sichuan University, Chengdu, 610041, China
| | - Yong Li
- State Key Laboratory of Biotherapy and Cancer Center and Collaborative Innovation Center of Biotherapy, West China Hospital of Sichuan University, Chengdu, 610041, China; Innovation Center of Nursing Research, Nursing Key Laboratory of Sichuan Province, West China Hospital of Sichuan University, Chengdu, 610041, China
| | - Na Shi
- State Key Laboratory of Biotherapy and Cancer Center and Collaborative Innovation Center of Biotherapy, West China Hospital of Sichuan University, Chengdu, 610041, China
| | - Yaxin Teng
- State Key Laboratory of Biotherapy and Cancer Center and Collaborative Innovation Center of Biotherapy, West China Hospital of Sichuan University, Chengdu, 610041, China
| | - Na Li
- State Key Laboratory of Biotherapy and Cancer Center and Collaborative Innovation Center of Biotherapy, West China Hospital of Sichuan University, Chengdu, 610041, China
| | - Minghai Tang
- State Key Laboratory of Biotherapy and Cancer Center and Collaborative Innovation Center of Biotherapy, West China Hospital of Sichuan University, Chengdu, 610041, China
| | - Ziyan Ma
- State Key Laboratory of Biotherapy and Cancer Center and Collaborative Innovation Center of Biotherapy, West China Hospital of Sichuan University, Chengdu, 610041, China
| | - Dexin Deng
- State Key Laboratory of Biotherapy and Cancer Center and Collaborative Innovation Center of Biotherapy, West China Hospital of Sichuan University, Chengdu, 610041, China
| | - Lijuan Chen
- State Key Laboratory of Biotherapy and Cancer Center and Collaborative Innovation Center of Biotherapy, West China Hospital of Sichuan University, Chengdu, 610041, China; Chengdu Zenitar Biomedical Technology Co., Ltd, Chengdu, 610041, China.
| |
Collapse
|
7
|
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: 3] [Impact Index Per Article: 3.0] [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
|
8
|
Guo Y, Zhou J, Wang Y, Wu X, Mou Y, Song X. Cell type-specific molecular mechanisms and implications of necroptosis in inflammatory respiratory diseases. Immunol Rev 2024; 321:52-70. [PMID: 37897080 DOI: 10.1111/imr.13282] [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] [Indexed: 10/29/2023]
Abstract
Necroptosis is generally considered as an inflammatory cell death form. The core regulators of necroptotic signaling are receptor-interacting serine-threonine protein kinases 1 (RIPK1) and RIPK3, and the executioner, mixed lineage kinase domain-like pseudokinase (MLKL). Evidence demonstrates that necroptosis contributes profoundly to inflammatory respiratory diseases that are common public health problem. Necroptosis occurs in nearly all pulmonary cell types in the settings of inflammatory respiratory diseases. The influence of necroptosis on cells varies depending upon the type of cells, tissues, organs, etc., which is an important factor to consider. Thus, in this review, we briefly summarize the current state of knowledge regarding the biology of necroptosis, and focus on the key molecular mechanisms that define the necroptosis status of specific cell types in inflammatory respiratory diseases. We also discuss the clinical potential of small molecular inhibitors of necroptosis in treating inflammatory respiratory diseases, and describe the pathological processes that engage cross talk between necroptosis and other cell death pathways in the context of respiratory inflammation. The rapid advancement of single-cell technologies will help understand the key mechanisms underlying cell type-specific necroptosis that are critical to effectively treat pathogenic lung infections and inflammatory respiratory diseases.
Collapse
Affiliation(s)
- Ying Guo
- Department of Otorhinolaryngology, Head and Neck Surgery, Yantai Yuhuangding Hospital of Qingdao University, Yantai, Shandong, China
- Shandong Provincial Clinical Research Center for Otorhinolaryngologic Diseases, Yantai, Shandong, China
| | - Jin Zhou
- Key Laboratory of Spatiotemporal Single-Cell Technologies and Translational Medicine, Yantai, Shandong, China
- Department of Endocrinology, Yantai Yuhuangding Hospital of Qingdao University, Yantai, Shandong, China
| | - Yaqi Wang
- Department of Otorhinolaryngology, Head and Neck Surgery, Yantai Yuhuangding Hospital of Qingdao University, Yantai, Shandong, China
| | - Xueliang Wu
- Department of General Surgery, The First Affiliated Hospital of Hebei North University, Zhangjiakou, Hebei, China
- Tumor Research Institute, The First Affiliated Hospital of Hebei North University, Zhangjiakou, Hebei, China
| | - Yakui Mou
- Department of Otorhinolaryngology, Head and Neck Surgery, Yantai Yuhuangding Hospital of Qingdao University, Yantai, Shandong, China
- Shandong Provincial Clinical Research Center for Otorhinolaryngologic Diseases, Yantai, Shandong, China
- Yantai Key Laboratory of Otorhinolaryngologic Diseases, Yantai, Shandong, China
| | - Xicheng Song
- Department of Otorhinolaryngology, Head and Neck Surgery, Yantai Yuhuangding Hospital of Qingdao University, Yantai, Shandong, China
- Key Laboratory of Spatiotemporal Single-Cell Technologies and Translational Medicine, Yantai, Shandong, China
| |
Collapse
|
9
|
Prasad Panda S, Kesharwani A, Prasanna Mallick S, Prasanth D, Kumar Pasala P, Bharadwaj Tatipamula V. Viral-induced neuronal necroptosis: Detrimental to brain function and regulation by necroptosis inhibitors. Biochem Pharmacol 2023; 213:115591. [PMID: 37196683 DOI: 10.1016/j.bcp.2023.115591] [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: 03/17/2023] [Revised: 05/09/2023] [Accepted: 05/10/2023] [Indexed: 05/19/2023]
Abstract
Neuronal necroptosis (programmed necrosis) in the CNS naturally occurs through a caspase-independent way and, especially in neurodegenerative diseases (NDDs) such as Alzheimer's disease (AD), Parknson's disease (PD), Amyotrophic Lateral Sclerosis (ALS) and viral infections. Understanding necroptosis pathways (death receptor-dependent and independent), and its connections with other cell death pathways could lead to new insights into treatment. Receptor-interacting protein kinase (RIPK) mediates necroptosis via mixed-lineage kinase-like (MLKL) proteins. RIPK/MLKL necrosome contains FADD, procaspase-8-cellular FLICE-inhibitory proteins (cFLIPs), RIPK1/RIPK3, and MLKL. The necrotic stimuli cause phosphorylation of MLKL and translocate to the plasma membrane, causing an influx of Ca2+ and Na+ ions and, the immediate opening of mitochondrial permeability transition pore (mPTP) with the release of inflammatory cell damage-associated molecular patterns (DAMPs) like mitochondrial DNA (mtDNA), high-mobility group box1 (HMGB1), and interleukin1 (IL-1). The MLKL translocates to the nucleus to induce transcription of the NLRP3 inflammasome complex elements. MLKL-induced NLRP3 activity causes caspase-1 cleavage and, IL-1 activation which promotes neuroinflammation. RIPK1-dependent transcription increases illness-associated microglial and lysosomal abnormalities to facilitate amyloid plaque (Aβ) aggregation in AD. Recent research has linked neuroinflammation and mitochondrial fission with necroptosis. MicroRNAs (miRs) such as miR512-3p, miR874, miR499, miR155, and miR128a regulate neuronal necroptosis by targeting key components of necroptotic pathways. Necroptosis inhibitors act by inhibiting the membrane translocation of MLKL and RIPK1 activity. This review insights into the RIPK/MLKL necrosome-NLRP3 inflammasome interactions during death receptor-dependent and independent neuronal necroptosis, and clinical intervention by miRs to protect the brain from NDDs.
Collapse
Affiliation(s)
- Siva Prasad Panda
- Institute of Pharmaceutical Research, GLA University, Mathura, Uttar Pradesh, India.
| | - Adarsh Kesharwani
- Institute of Pharmaceutical Research, GLA University, Mathura, Uttar Pradesh, India
| | - Sarada Prasanna Mallick
- Department of Biotechnology, Koneru Lakshmaiah Education Foundation, Guntur, Andhrapradesh, India
| | - Dsnbk Prasanth
- Department of Pharmacognosy, KVSR Siddhartha College of Pharmaceutical Sciences, Vijayawada, AP, India
| | | | - Vinay Bharadwaj Tatipamula
- Center for Molecular Biology, College of Medicine and Pharmacy, Duy Tan University, Danang 550000, Viet Nam
| |
Collapse
|
10
|
Patankar JV, Bubeck M, Acera MG, Becker C. Breaking bad: necroptosis in the pathogenesis of gastrointestinal diseases. Front Immunol 2023; 14:1203903. [PMID: 37409125 PMCID: PMC10318896 DOI: 10.3389/fimmu.2023.1203903] [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: 04/11/2023] [Accepted: 06/07/2023] [Indexed: 07/07/2023] Open
Abstract
A delicate balance between programmed cell death and proliferation of intestinal epithelial cells (IEC) exists in the gut to maintain homeostasis. Homeostatic cell death programs such as anoikis and apoptosis ensure the replacement of dead epithelia without overt immune activation. In infectious and chronic inflammatory diseases of the gut, this balance is invariably disturbed by increased levels of pathologic cell death. Pathological forms of cell death such as necroptosis trigger immune activation barrier dysfunction, and perpetuation of inflammation. A leaky and inflamed gut can thus become a cause of persistent low-grade inflammation and cell death in other organs of the gastrointestinal (GI) tract, such as the liver and the pancreas. In this review, we focus on the advances in the molecular and cellular understanding of programmed necrosis (necroptosis) in tissues of the GI tract. In this review, we will first introduce the reader to the basic molecular aspects of the necroptosis machinery and discuss the pathways leading to necroptosis in the GI system. We then highlight the clinical significance of the preclinical findings and finally evaluate the different therapeutic approaches that attempt to target necroptosis against various GI diseases. Finally, we review the recent advances in understanding the biological functions of the molecules involved in necroptosis and the potential side effects that may occur due to their systemic inhibition. This review is intended to introduce the reader to the core concepts of pathological necroptotic cell death, the signaling pathways involved, its immuno-pathological implications, and its relevance to GI diseases. Further advances in our ability to control the extent of pathological necroptosis will provide better therapeutic opportunities against currently intractable GI and other diseases.
Collapse
Affiliation(s)
- Jay V. Patankar
- Department of Medicine 1, University of Erlangen-Nuremberg, Erlangen, Germany
- Deutsches Zentrum Immuntherapie (DZI), Erlangen, Germany
| | - Marvin Bubeck
- Department of Medicine 1, University of Erlangen-Nuremberg, Erlangen, Germany
- Deutsches Zentrum Immuntherapie (DZI), Erlangen, Germany
| | - Miguel Gonzalez Acera
- Department of Medicine 1, University of Erlangen-Nuremberg, Erlangen, Germany
- Deutsches Zentrum Immuntherapie (DZI), Erlangen, Germany
| | - Christoph Becker
- Department of Medicine 1, University of Erlangen-Nuremberg, Erlangen, Germany
- Deutsches Zentrum Immuntherapie (DZI), Erlangen, Germany
| |
Collapse
|
11
|
Feng M, Zhang R, Zhang M, Chen M, Ji L, Duan D, Qiang H. Administration of necrostatin-1 ameliorates glucocorticoid-induced osteonecrosis of the femoral head in rats. J Mol Histol 2023:10.1007/s10735-023-10124-x. [PMID: 37156987 DOI: 10.1007/s10735-023-10124-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2022] [Accepted: 04/30/2023] [Indexed: 05/10/2023]
Abstract
Glucocorticoid (GC)-induced osteonecrosis of the femoral head (ONFH) is a serious complication of glucocorticoid treatment and is characterized by dysfunctional bone reconstruction at necrotic sites. Our previous study confirmed the protective potential of necrostatin-1, a selective blocker of necroptosis, in glucocorticoid-induced osteoporosis. In this study, rat models of GC-induced ONFH were established to evaluate the effects of necrostatin-1 on osteonecrotic changes and repair processes. Osteonecrosis was verified by histopathological staining. An analysis of trabecular bone architecture was performed to evaluate osteogenesis in the osteonecrotic zone. Then, necroptotic signaling molecules such as RIP1 and RIP3 were examined by immunohistochemistry. Histopathological observations indicated that necrostatin-1 administration reduced the incidence of osteonecrosis and the osteogenic response in subchondral areas. Additionally, bone histomorphometry demonstrated that necrostatin-1 intervention could restore bone reconstruction in the necrotic zone. The protective mechanism of necrostatin-1 was related to the inhibition of RIP1 and RIP3. Necrostatin-1 administration alleviated GC-induced ONFH in rats by attenuating the formation of necrotic lesions, recovering the function of osteogenesis, and suppressing glucocorticoid-induced osteocytic necroptosis by inhibiting the expression of RIP1 and RIP3.
Collapse
Affiliation(s)
- Min Feng
- Department of Orthopaedics, Shaanxi Provincial People's Hospital, Xi'an, China
| | - Ruirui Zhang
- Department of ICU, 521 Healthy Institute of North Industries, Xi'an, China
| | - Mingming Zhang
- Department of Anesthesia, 521 Healthy Institute of North Industries, Xi'an, China
| | - Ming Chen
- Department of Orthopaedics, Shaanxi Provincial People's Hospital, Xi'an, China
| | - Le Ji
- Department of Orthopaedics, Shaanxi Provincial People's Hospital, Xi'an, China
| | - Dapeng Duan
- Department of Orthopaedics, Shaanxi Provincial People's Hospital, Xi'an, China
| | - Hui Qiang
- Department of Orthopaedics, Shaanxi Provincial People's Hospital, Xi'an, China.
| |
Collapse
|
12
|
Sanz AB, Sanchez-Niño MD, Ramos AM, Ortiz A. Regulated cell death pathways in kidney disease. Nat Rev Nephrol 2023; 19:281-299. [PMID: 36959481 PMCID: PMC10035496 DOI: 10.1038/s41581-023-00694-0] [Citation(s) in RCA: 68] [Impact Index Per Article: 68.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/16/2023] [Indexed: 03/25/2023]
Abstract
Disorders of cell number that result from an imbalance between the death of parenchymal cells and the proliferation or recruitment of maladaptive cells contributes to the pathogenesis of kidney disease. Acute kidney injury can result from an acute loss of kidney epithelial cells. In chronic kidney disease, loss of kidney epithelial cells leads to glomerulosclerosis and tubular atrophy, whereas interstitial inflammation and fibrosis result from an excess of leukocytes and myofibroblasts. Other conditions, such as acquired cystic disease and kidney cancer, are characterized by excess numbers of cyst wall and malignant cells, respectively. Cell death modalities act to clear unwanted cells, but disproportionate responses can contribute to the detrimental loss of kidney cells. Indeed, pathways of regulated cell death - including apoptosis and necrosis - have emerged as central events in the pathogenesis of various kidney diseases that may be amenable to therapeutic intervention. Modes of regulated necrosis, such as ferroptosis, necroptosis and pyroptosis may cause kidney injury directly or through the recruitment of immune cells and stimulation of inflammatory responses. Importantly, multiple layers of interconnections exist between different modalities of regulated cell death, including shared triggers, molecular components and protective mechanisms.
Collapse
Affiliation(s)
- Ana B Sanz
- Department of Nephrology and Hypertension, IIS-Fundacion Jimenez Diaz UAM, Madrid, Spain
- RICORS2040, Madrid, Spain
| | - Maria Dolores Sanchez-Niño
- Department of Nephrology and Hypertension, IIS-Fundacion Jimenez Diaz UAM, Madrid, Spain
- RICORS2040, Madrid, Spain
- Departamento de Medicina, Facultad de Medicina, Universidad Autónoma de Madrid, Madrid, Spain
| | - Adrian M Ramos
- Department of Nephrology and Hypertension, IIS-Fundacion Jimenez Diaz UAM, Madrid, Spain
- RICORS2040, Madrid, Spain
| | - Alberto Ortiz
- Department of Nephrology and Hypertension, IIS-Fundacion Jimenez Diaz UAM, Madrid, Spain.
- RICORS2040, Madrid, Spain.
- Departamento de Farmacología, Universidad Autonoma de Madrid, Madrid, Spain.
| |
Collapse
|
13
|
Aleem AM, Kang W, Lin S, Milad M, Kingsley PJ, Crews BC, Uddin MJ, Rouzer CA, Marnett LJ. Ferroptosis Inhibitors Suppress Prostaglandin Synthesis in Lipopolysaccharide-Stimulated Macrophages. ACS Chem Biol 2023; 18:404-418. [PMID: 36638351 DOI: 10.1021/acschembio.2c00869] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Necrostatin-1 blocks ferroptosis via an unknown mechanism and necroptosis through inhibition of receptor-interacting protein kinase-1 (RIP1). We report that necrostatin-1 suppresses cyclooxygenase-2-dependent prostaglandin biosynthesis in lipopolysaccharide-treated RAW264.7 macrophages (IC50 ∼ 100 μM). This activity is shared by necrostatin-1i (IC50 ∼ 50 μM), which lacks RIP1 inhibitory activity, but not the RIP1 inhibitors necrostatin-1s or deschloronecrostatin-1s. Furthermore, we show that the potent ferroptosis inhibitors and related compounds ferrostatin-1, phenoxazine, phenothiazine, and 10-methylphenothiazine strongly inhibit cellular prostaglandin biosynthesis with IC50's in the range of 30 nM to 3.5 μM. None of the compounds inhibit lipopolysaccharide-mediated cyclooxygenase-2 protein induction. In the presence of activating hydroperoxides, the necrostatins and ferroptosis inhibitors range from low potency inhibition to stimulation of in vitro cyclooxygenase-2 activity; however, inhibitory potency is increased under conditions of low peroxide tone. The ferroptosis inhibitors are highly effective reducing substrates for cyclooxygenase-2's peroxidase activity, suggesting that they act by suppressing hydroperoxide-mediated activation of the cyclooxygenase active site. In contrast, for the necrostatins, cellular prostaglandin synthesis inhibition does not correlate with peroxidase-reducing activity but rather with the presence of a thiohydantoin substituent, which conveys the ability to reduce the endoperoxide intermediate prostaglandin H2 to prostaglandin F2α in vitro. This finding suggests that necrostatin-1 blocks cellular prostaglandin synthesis and ferroptosis via a redox mechanism distinct from action as a one-electron donor. The results indicate that a wide range of compounds derived from redox-active chemical scaffolds can block cellular prostaglandin biosynthesis.
Collapse
Affiliation(s)
- Ansari M Aleem
- A. B. Hancock, Jr., Memorial Laboratory for Cancer Research, Departments of Biochemistry, Chemistry, and Pharmacology, Vanderbilt Institute of Chemical Biology, and Vanderbilt-Ingram Cancer Center, Vanderbilt University School of Medicine, Nashville, Tennessee 37232, United States
| | - Weixi Kang
- A. B. Hancock, Jr., Memorial Laboratory for Cancer Research, Departments of Biochemistry, Chemistry, and Pharmacology, Vanderbilt Institute of Chemical Biology, and Vanderbilt-Ingram Cancer Center, Vanderbilt University School of Medicine, Nashville, Tennessee 37232, United States
| | - Shuyang Lin
- A. B. Hancock, Jr., Memorial Laboratory for Cancer Research, Departments of Biochemistry, Chemistry, and Pharmacology, Vanderbilt Institute of Chemical Biology, and Vanderbilt-Ingram Cancer Center, Vanderbilt University School of Medicine, Nashville, Tennessee 37232, United States
| | - Matthew Milad
- A. B. Hancock, Jr., Memorial Laboratory for Cancer Research, Departments of Biochemistry, Chemistry, and Pharmacology, Vanderbilt Institute of Chemical Biology, and Vanderbilt-Ingram Cancer Center, Vanderbilt University School of Medicine, Nashville, Tennessee 37232, United States
| | - Philip J Kingsley
- A. B. Hancock, Jr., Memorial Laboratory for Cancer Research, Departments of Biochemistry, Chemistry, and Pharmacology, Vanderbilt Institute of Chemical Biology, and Vanderbilt-Ingram Cancer Center, Vanderbilt University School of Medicine, Nashville, Tennessee 37232, United States
| | - Brenda C Crews
- A. B. Hancock, Jr., Memorial Laboratory for Cancer Research, Departments of Biochemistry, Chemistry, and Pharmacology, Vanderbilt Institute of Chemical Biology, and Vanderbilt-Ingram Cancer Center, Vanderbilt University School of Medicine, Nashville, Tennessee 37232, United States
| | - Md Jashim Uddin
- A. B. Hancock, Jr., Memorial Laboratory for Cancer Research, Departments of Biochemistry, Chemistry, and Pharmacology, Vanderbilt Institute of Chemical Biology, and Vanderbilt-Ingram Cancer Center, Vanderbilt University School of Medicine, Nashville, Tennessee 37232, United States
| | - Carol A Rouzer
- A. B. Hancock, Jr., Memorial Laboratory for Cancer Research, Departments of Biochemistry, Chemistry, and Pharmacology, Vanderbilt Institute of Chemical Biology, and Vanderbilt-Ingram Cancer Center, Vanderbilt University School of Medicine, Nashville, Tennessee 37232, United States
| | - Lawrence J Marnett
- A. B. Hancock, Jr., Memorial Laboratory for Cancer Research, Departments of Biochemistry, Chemistry, and Pharmacology, Vanderbilt Institute of Chemical Biology, and Vanderbilt-Ingram Cancer Center, Vanderbilt University School of Medicine, Nashville, Tennessee 37232, United States
| |
Collapse
|
14
|
Horne CR, Samson AL, Murphy JM. The web of death: the expanding complexity of necroptotic signaling. Trends Cell Biol 2023; 33:162-174. [PMID: 35750616 DOI: 10.1016/j.tcb.2022.05.008] [Citation(s) in RCA: 16] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Revised: 05/28/2022] [Accepted: 05/30/2022] [Indexed: 01/25/2023]
Abstract
The past decade has seen the emergence of the necroptosis programmed cell death pathway as an important contributor to the pathophysiology of myriad diseases. The receptor interacting protein kinase (RIPK)1 and RIPK3, and the pseudokinase executioner protein, mixed lineage kinase domain-like (MLKL), have grown to prominence as the core pathway components. Depending on cellular context, these proteins also serve as integrators of signals, such as post-translational modifications and protein or metabolite interactions, adding layers of complexity to pathway regulation. Here, we describe the emerging picture of the web of proteins that tune necroptotic signal transduction and how these events have diverged across species, presumably owing to selective pressures of pathogens upon the RIPK3-MLKL protein pair.
Collapse
Affiliation(s)
- Christopher R Horne
- Walter and Eliza Hall Institute of Medical Research, 1G Royal Parade, Parkville, VIC 3052, Australia; Department of Medical Biology, University of Melbourne, Parkville, VIC 3052, Australia
| | - André L Samson
- Walter and Eliza Hall Institute of Medical Research, 1G Royal Parade, Parkville, VIC 3052, Australia; Department of Medical Biology, University of Melbourne, Parkville, VIC 3052, Australia.
| | - James M Murphy
- Walter and Eliza Hall Institute of Medical Research, 1G Royal Parade, Parkville, VIC 3052, Australia; Department of Medical Biology, University of Melbourne, Parkville, VIC 3052, Australia.
| |
Collapse
|
15
|
Takiishi T, Xiao P, Franchimont M, Gilglioni EH, Arroba EN, Gurzov EN, Bertrand MJM, Cardozo AK. Inhibition of RIPK1 kinase does not affect diabetes development: β-Cells survive RIPK1 activation. Mol Metab 2023; 69:101681. [PMID: 36707047 PMCID: PMC9932129 DOI: 10.1016/j.molmet.2023.101681] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Revised: 12/30/2022] [Accepted: 01/19/2023] [Indexed: 01/26/2023] Open
Abstract
OBJECTIVES Type 1 diabetes (T1D) is caused by progressive immune-mediated loss of insulin-producing β-cells. Inflammation is detrimental to β-cell function and survival, moreover, both apoptosis and necrosis have been implicated as mechanisms of β-cell loss in T1D. The receptor interacting serine/threonine protein kinase 1 (RIPK1) promotes inflammation by serving as a scaffold for NF-κB and MAPK activation, or by acting as a kinase that triggers apoptosis or necroptosis. It is unclear whether RIPK1 kinase activity is involved in T1D pathology. In the present study, we investigated if absence of RIPK1 activation would affect the susceptibility to immune-mediated diabetes or diet induced obesity (DIO). METHODS The RIPK1 knockin mouse line carrying a mutation mimicking serine 25 phosphorylation (Ripk1S25D/S25D), which abrogates RIPK1 kinase activity, was utilized to assess the in vivo role of RIPK1 in immune-mediated diabetes or diet induced obesity (DIO). In vitro, β-cell death and RIPK1 kinase activity was analysed in conditions known to induce RIPK1-dependent apoptosis/necroptosis. RESULTS We demonstrate that Ripk1S25D/S25D mice presented normal glucose metabolism and β-cell function. Furthermore, immune-mediated diabetes and DIO were not different between Ripk1S25D/S25D and Ripk1+/+ mice. Despite strong activation of RIPK1 kinase and other necroptosis effectors (RIPK3 and MLKL) by TNF+BV6+zVAD, no cell death was observed in mouse islets nor human β-cells. CONCLUSION Our results contrast recent literature showing that most cell types undergo necroptosis following RIPK1 kinase activation. This peculiarity may reflect an adaptation to the inability of β-cells to proliferate and self-renewal.
Collapse
Affiliation(s)
- Tatiana Takiishi
- Inflammation and Cell Death Signalling Group, Laboratoire de Gastroentérologie Expérimental et Endotools, Université libre de Bruxelles (ULB), Brussels, Belgium.
| | - Peng Xiao
- Inflammation and Cell Death Signalling Group, Laboratoire de Gastroentérologie Expérimental et Endotools, Université libre de Bruxelles (ULB), Brussels, Belgium.
| | - Marie Franchimont
- Inflammation and Cell Death Signalling Group, Laboratoire de Gastroentérologie Expérimental et Endotools, Université libre de Bruxelles (ULB), Brussels, Belgium.
| | - Eduardo H. Gilglioni
- Signal Transduction and Metabolism Laboratory, Laboratoire de Gastroentérologie Expérimental et Endotools, Université libre de Bruxelles (ULB), Brussels, Belgium
| | - Erick N. Arroba
- Inflammation and Cell Death Signalling Group, Laboratoire de Gastroentérologie Expérimental et Endotools, Université libre de Bruxelles (ULB), Brussels, Belgium,Signal Transduction and Metabolism Laboratory, Laboratoire de Gastroentérologie Expérimental et Endotools, Université libre de Bruxelles (ULB), Brussels, Belgium
| | - Esteban N. Gurzov
- Signal Transduction and Metabolism Laboratory, Laboratoire de Gastroentérologie Expérimental et Endotools, Université libre de Bruxelles (ULB), Brussels, Belgium,WELBIO, WEL Research Institute, Avenue Pasteur 6, Wavre, 1300, Belgium
| | - Mathieu JM. Bertrand
- UGent Center for inflammation Research, Ghent, Belgium,Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Alessandra K. Cardozo
- Inflammation and Cell Death Signalling Group, Laboratoire de Gastroentérologie Expérimental et Endotools, Université libre de Bruxelles (ULB), Brussels, Belgium,Corresponding author. Inflammation and Cell Death Signalling Group, Laboratoire de Gastroentérologie Expérimental et Endotools, Université libre de Bruxelles, Route de Lennik, 808, CP 697/02, 1070, Brussels, Belgium.
| |
Collapse
|
16
|
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
|
17
|
Yuan G, Cao C, Cao D, Li B, Li X, Li H, Shen H, Wang Z, Chen G. Receptor-interacting protein 3-phosphorylated Ca 2+ /calmodulin-dependent protein kinase II and mixed lineage kinase domain-like protein mediate intracerebral hemorrhage-induced neuronal necroptosis. J Neurochem 2023; 164:94-114. [PMID: 36424866 DOI: 10.1111/jnc.15731] [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: 09/13/2021] [Revised: 10/15/2022] [Accepted: 11/13/2022] [Indexed: 11/26/2022]
Abstract
Necroptosis-mediated cell death is an important mechanism in intracerebral hemorrhage (ICH)-induced secondary brain injury (SBI). Our previous study has demonstrated that receptor-interacting protein 1 (RIP1) mediated necroptosis in SBI after ICH. However, further mechanisms, such as the roles of receptor-interacting protein 3 (RIP3), mixed lineage kinase domain-like protein (MLKL), and Ca2+ /calmodulin-dependent protein kinase II (CaMK II), remain unclear. We hypothesized that RIP3, MLKL, and CaMK II might participate in necroptosis after ICH, including their phosphorylation. The ICH model was induced by autologous blood injection. First, we found the activation of necroptosis after ICH in brain tissues surrounding the hematoma (propidium iodide staining). Meanwhile, the phosphorylation and expression of RIP3, MLKL, and CaMK II were differently up-regulated (western blotting and immunofluorescent staining). The specific inhibitors could suppress RIP3, MLKL, and CaMK II (GSK'872 for RIP3, necrosulfonamide for MLKL, and KN-93 for CaMK II). We found the necroptosis surrounding the hematoma and the concrete interactions in RIP3-MLKL/RIP3-CaMK II also both decreased after the specific intervention (co-immunoprecipitation). Then we conducted the short-/long-term neurobehavioral tests, and the rats with specific inhibition mostly had better performance. We also found less blood-brain barrier (BBB) injury, and less neuron loss (Nissl staining) in intervention groups, which supported the neurobehavioral tests. Besides, oxidative stress and inflammation were also alleviated with intervention, which had significant less reactive oxygen species (ROS), tumor necrosis factor (TNF)-α, lactate dehydrogenase (LDH), Iba1, and GFAP surrounding the hematoma. These results confirmed that RIP3-phosphorylated MLKL and CaMK II participate in ICH-induced necroptosis and could provide potential targets for the treatment of ICH patients.
Collapse
Affiliation(s)
- Guiqiang Yuan
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Cheng Cao
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Demao Cao
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Bing Li
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Xiang Li
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Haiying Li
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Haitao Shen
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Zhong Wang
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Gang Chen
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Suzhou, China
| |
Collapse
|
18
|
Zhang R, Song Y, Su X. Necroptosis and Alzheimer's Disease: Pathogenic Mechanisms and Therapeutic Opportunities. J Alzheimers Dis 2023; 94:S367-S386. [PMID: 36463451 PMCID: PMC10473100 DOI: 10.3233/jad-220809] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/07/2022] [Indexed: 11/30/2022]
Abstract
Alzheimer's disease (AD) is considered to be the most common neurodegenerative disease, with clinical symptoms encompassing progressive memory loss and cognitive impairment. Necroptosis is a form of programmed necrosis that promotes cell death and neuroinflammation, which further mediates the pathogenesis of several neurodegenerative diseases, especially AD. Current evidence has strongly suggested that necroptosis is activated in AD brains, resulting in neuronal death and cognitive impairment. We searched the PubMed database, screening all articles published before September 28, 2022 related to necroptosis in the context of AD pathology. The keywords in the search included: "necroptosis", "Alzheimer's disease", "signaling pathways", "Aβ", Aβo", "Tau", "p-Tau", "neuronal death", "BBB damage", "neuroinflammation", "microglia", "mitochondrial dysfunction", "granulovacuolar degeneration", "synaptic loss", "axonal degeneration", "Nec-1", "Nec-1s", "GSK872", "NSA", "OGA", "RIPK1", "RIPK3", and "MLKL". Results show that necroptosis has been involved in multiple pathological processes of AD, including amyloid-β aggregation, Tau accumulation, neuronal death, and blood-brain barrier damage, etc. More importantly, existing research on AD necroptosis interventions, including drug intervention and potential gene targets, as well as its current clinical development status, was discussed. Finally, the issues pertaining to necroptosis in AD were presented. Accordingly, this review may provide further insight into clinical perspectives and challenges for the future treatment of AD by targeting the necroptosis pathway.
Collapse
Affiliation(s)
- Ruxin Zhang
- Linfen People’s Hospital, Linfen, Shanxi, China
| | | | - Xuefeng Su
- Linfen People’s Hospital, Linfen, Shanxi, China
| |
Collapse
|
19
|
Wang PF, Yang LQ, Shi ZH, Li XM, Qiu HY. An updated patent review of IDO1 inhibitors for cancer (2018-2022). Expert Opin Ther Pat 2022; 32:1145-1159. [PMID: 36420761 DOI: 10.1080/13543776.2022.2151894] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
INTRODUCTION Indoleamine 2,3-dioxygenase 1 (IDO1) is highly related to the immune evasion of a wide range of malignancies due to its role in the immune suppression caused by the depletion of tryptophan (Trp) and the accumulation of kynurenine (Kyn). The combination of IDO1 inhibitors with other treatments represents a promising strategy in immunotherapy, although considerable challenges lie ahead. AREAS COVERED This review focuses on patent publications searched from Espacenet and Google Scholar, and related to IDO1 inhibitors with potential anti-cancer utilization during the period 2018-2022. EXPERT OPINION Despite the clinical trial failure of the first-in-class IDO1 inhibitor epacadostat in combination with pembrolizumab, numerous studies have been carried on to pursue more efficient IDO1-based immune-modulating therapeutic solutions. A large number of IDO1 inhibitors with new structures and design concepts have been produced with the impetus of crystallographic studies, and have shown great research potential. The elaboration on the combination of IDO1 inhibitors with other targeting agents, the more precise selection of patients, the identification of more reliable biomarkers for evaluating the IDO1 treatment, and the investigation of possible toxicity, are critical factors to promote IDO1-based immunotherapies from bench to bedside.
Collapse
Affiliation(s)
- Peng-Fei Wang
- Chongqing Key Laboratory of Natural Product Synthesis and Drug Research, School of Pharmaceutical Sciences, Chongqing University, Chongqing, People's Republic of China
| | - Li-Qiang Yang
- Chongqing Key Laboratory of Natural Product Synthesis and Drug Research, School of Pharmaceutical Sciences, Chongqing University, Chongqing, People's Republic of China
| | - Zhao-Hang Shi
- Chongqing Key Laboratory of Natural Product Synthesis and Drug Research, School of Pharmaceutical Sciences, Chongqing University, Chongqing, People's Republic of China
| | - Xue-Min Li
- Chongqing Key Laboratory of Natural Product Synthesis and Drug Research, School of Pharmaceutical Sciences, Chongqing University, Chongqing, People's Republic of China
| | - Han-Yue Qiu
- Chongqing Key Laboratory of Natural Product Synthesis and Drug Research, School of Pharmaceutical Sciences, Chongqing University, Chongqing, People's Republic of China
| |
Collapse
|
20
|
Zhou B, Xu Q, Guo J, Chen Q, Lv Q, Xiao K, Zhu H, Zhao J, Liu Y. Necroptosis Contributes to LPS-Induced Activation of the Hypothalamic-Pituitary-Adrenal Axis in a Piglet Model. Int J Mol Sci 2022; 23:ijms231911218. [PMID: 36232518 PMCID: PMC9569845 DOI: 10.3390/ijms231911218] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Revised: 09/08/2022] [Accepted: 09/14/2022] [Indexed: 11/16/2022] Open
Abstract
Stressors cause activation of the hypothalamic-pituitary-adrenal (HPA) axis and a systemic inflammatory response. As a newly proposed cell death manner in recent years, necroptosis occurs in a variety of tissue damage and inflammation. However, the role of necroptosis in HPA axis activation remains to be elucidated. The aim of this study was to investigate the occurrence of necroptosis and its role in HPA activation in a porcine stress model induced by Escherichia coli lipopolysaccharide (LPS). Several typical stress behaviors like fever, anorexia, shivering and vomiting were observed in piglets after LPS injection. HPA axis was activated as shown by increased plasma cortisol concentration and mRNA expression of pituitary corticotropin-releasing hormone receptor 1 (CRHR1) and adrenal steroidogenic acute regulatory protein (StAR). The mRNA expression of tumor necrosis factor α (TNF-α), interleukin-1β (IL-1β) and IL-6 in the hypothalamus, pituitary gland and adrenal gland was elevated by LPS, accompanied by the activation of necroptosis indicated by higher mRNA expression of necroptosis signals including receptor-interacting protein kinase (RIP) 1, RIP3, and phosphorylated mixed-lineage kinase domain-like protein (MLKL). Furthermore, necrostatin-1 (Nec-1), an inhibitor of necroptosis, inhibited necroptosis indicated by decreased mRNA levels of RIP1, RIP3, MLKL, and phosphoglycerate mutase family member 5 (PGAM5) in the hypothalamus, pituitary gland and adrenal gland. Nec-1 also decreased the mRNA expression of TNF-α and IL-β and inhibited the activation of the HPA axis indicated by lower plasma cortisol concentration and mRNA expression of adrenal type 2 melanocortin receptor (MC2R) and StAR. These findings suggest that necroptosis is present and contributes to HPA axis activation induced by LPS. These findings provide a potential possibility for necroptosis as an intervention target for alleviating HPA axis activation and stress responses.
Collapse
Affiliation(s)
- Bei Zhou
- Hubei Key Laboratory of Animal Nutrition and Feed Science, Hubei Collaborative Innovation Center for Animal Nutrition and Feed Safety, Wuhan Polytechnic University, Wuhan 430023, China
| | - Qilong Xu
- Hubei Key Laboratory of Animal Nutrition and Feed Science, Hubei Collaborative Innovation Center for Animal Nutrition and Feed Safety, Wuhan Polytechnic University, Wuhan 430023, China
| | - Junjie Guo
- Hubei Key Laboratory of Animal Nutrition and Feed Science, Hubei Collaborative Innovation Center for Animal Nutrition and Feed Safety, Wuhan Polytechnic University, Wuhan 430023, China
| | - Qinliang Chen
- Hubei Key Laboratory of Animal Nutrition and Feed Science, Hubei Collaborative Innovation Center for Animal Nutrition and Feed Safety, Wuhan Polytechnic University, Wuhan 430023, China
| | - Qingqing Lv
- Hubei Key Laboratory of Animal Nutrition and Feed Science, Hubei Collaborative Innovation Center for Animal Nutrition and Feed Safety, Wuhan Polytechnic University, Wuhan 430023, China
| | - Kan Xiao
- Hubei Key Laboratory of Animal Nutrition and Feed Science, Hubei Collaborative Innovation Center for Animal Nutrition and Feed Safety, Wuhan Polytechnic University, Wuhan 430023, China
| | - Huiling Zhu
- Hubei Key Laboratory of Animal Nutrition and Feed Science, Hubei Collaborative Innovation Center for Animal Nutrition and Feed Safety, Wuhan Polytechnic University, Wuhan 430023, China
| | - Jiangchao Zhao
- Department of Animal Science, Division of Agriculture, University of Arkansas, Fayetteville, AR 72701, USA
| | - Yulan Liu
- Hubei Key Laboratory of Animal Nutrition and Feed Science, Hubei Collaborative Innovation Center for Animal Nutrition and Feed Safety, Wuhan Polytechnic University, Wuhan 430023, China
- Correspondence: ; Tel./Fax: +86-027-8395-6175
| |
Collapse
|
21
|
Mark C, Callander NS, Chng K, Miyamoto S, Warrick J. Timelapse viability assay to detect division and death of primary multiple myeloma cells in response to drug treatments with single cell resolution. Integr Biol (Camb) 2022; 14:49-61. [PMID: 35653717 PMCID: PMC9175638 DOI: 10.1093/intbio/zyac006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Revised: 03/15/2022] [Accepted: 04/12/2022] [Indexed: 11/12/2022]
Abstract
Heterogeneity among cancer cells and in the tumor microenvironment (TME) is thought to be a significant contributor to the heterogeneity of clinical therapy response observed between patients and can evolve over time. A primary example of this is multiple myeloma (MM), a generally incurable cancer where such heterogeneity contributes to the persistent evolution of drug resistance. However, there is a paucity of functional assays for studying this heterogeneity in patient samples or for assessing the influence of the patient TME on therapy response. Indeed, the population-averaged data provided by traditional drug response assays and the large number of cells required for screening remain significant hurdles to advancement. To address these hurdles, we developed a suite of accessible technologies for quantifying functional drug response to a panel of therapies in ex vivo three-dimensional culture using small quantities of a patient's own cancer and TME components. This suite includes tools for label-free single-cell identification and quantification of both cell division and death events with a standard brightfield microscope, an open-source software package for objective image analysis and feasible data management of multi-day timelapse experiments, and a new approach to fluorescent detection of cell death that is compatible with long-term imaging of primary cells. These new tools and capabilities are used to enable sensitive, objective, functional characterization of primary MM cell therapy response in the presence of TME components, laying the foundation for future studies and efforts to enable predictive assessment drug efficacy for individual patients.
Collapse
Affiliation(s)
- Christina Mark
- Cancer Biology Graduate Program, University of Wisconsin, Madison, WI 53705, USA
| | - Natalie S Callander
- University of Wisconsin Carbone Cancer Center, University of Wisconsin, Madison, WI 53705, USA
- Department of Medicine, University of Wisconsin, Madison, WI 53705, USA
| | - Kenny Chng
- McArdle Laboratory of Cancer Research, University of Wisconsin, Madison, WI 53705, USA
| | - Shigeki Miyamoto
- Cancer Biology Graduate Program, University of Wisconsin, Madison, WI 53705, USA
- University of Wisconsin Carbone Cancer Center, University of Wisconsin, Madison, WI 53705, USA
- McArdle Laboratory of Cancer Research, University of Wisconsin, Madison, WI 53705, USA
- Department of Oncology, University of Wisconsin, Madison, WI 53705, USA
| | - Jay Warrick
- Department of Biomedical Engineering, University of Wisconsin, Madison, WI 53705, USA
- Salus Discovery, LLC, 110 E. Main St. Suite 815, Madison, WI 53703, USA
| |
Collapse
|
22
|
Zhang J, Huang J, Lin X, Fei K, Xie Y, Peng Q, Li X, Xie L, Dai L, Zhang W. Phosphoglycerate mutase 5 promotes necroptosis in trophoblast cells through activation of dynamin-related protein 1 in early-onset preeclampsia. Am J Reprod Immunol 2022; 87:e13539. [PMID: 35304783 DOI: 10.1111/aji.13539] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2021] [Revised: 02/01/2022] [Accepted: 03/12/2022] [Indexed: 12/12/2022] Open
Abstract
OBJECTIVES Placentae from patients with preeclampsia have increased susceptibility to necroptosis and phosphoglycerate mutase 5 (PGAM5) plays a role in many necrosis pathways. We determined whether PGAM5 promotes necroptosis of trophoblast cells and the underlying mechanisms in this study. METHODS The injury model was established by treating JEG3 cells with hypoxia for 24 h. The functional measurements were assessed by the cell counting kit-8, propidium iodide (PI)/Annexin V staining, JC-1 staining and firefly luciferase ATP assay. The expression of proteins in human placentae and JEG3 cells was measured Western blot. PGAM5 was knocked down to study its role in hypoxia-induced necroptosis. RESULTS The placentae from patients with preeclampsia showed up-regulation of PGAM5 and decreased levels of p-Drp1-S637, accompanied by increased necroptosis-relevant proteins expression. The expression of PGAM5 in JEG3 cells was up-regulated under hypoxia, which promoted dephosphorylation of Drp1 at Serine 637 residue, mitochondrial dysfunction (elevated ROS level and reduced mitochondrial membrane potential and ATP content) and cellular necroptosis (increased PI+ /Annexin V+ cells and decreased cell viability), accompanied by increased expression of necroptosis-relevant proteins; knockdown of PGAM5 attenuated these phenomena. CONCLUSIONS Our results indicate that PGAM5 can promote necroptosis in trophoblast cells through, at least in part, activation of Drp1. It may be used as a new therapeutic target to prevent trophoblast dysfunction in preeclampsia.
Collapse
Affiliation(s)
- Jiejie Zhang
- Department of Obstetrics, Xiangya Hospital, Central South University, Changsha, China.,Hunan Engineering Research Center of Early Life Development and Disease Prevention, Changsha, China
| | - Jingrui Huang
- Department of Obstetrics, Xiangya Hospital, Central South University, Changsha, China
| | - Xinxiu Lin
- Department of Obstetrics, Xiangya Hospital, Central South University, Changsha, China
| | - Kuilin Fei
- Department of Obstetrics, Xiangya Hospital, Central South University, Changsha, China
| | - Yingming Xie
- Department of Obstetrics, Xiangya Hospital, Central South University, Changsha, China
| | - Qiaozhen Peng
- Department of Obstetrics, Xiangya Hospital, Central South University, Changsha, China
| | - Xun Li
- Department of Obstetrics, Xiangya Hospital, Central South University, Changsha, China
| | - Liangqun Xie
- Department of Obstetrics, Xiangya Hospital, Central South University, Changsha, China
| | - Lei Dai
- Department of Obstetrics, Xiangya Hospital, Central South University, Changsha, China
| | - Weishe Zhang
- Department of Obstetrics, Xiangya Hospital, Central South University, Changsha, China.,Hunan Engineering Research Center of Early Life Development and Disease Prevention, Changsha, China
| |
Collapse
|
23
|
Adameova A, Horvath C, Abdul-Ghani S, Varga ZV, Suleiman MS, Dhalla NS. Interplay of Oxidative Stress and Necrosis-like Cell Death in Cardiac Ischemia/Reperfusion Injury: A Focus on Necroptosis. Biomedicines 2022; 10:biomedicines10010127. [PMID: 35052807 PMCID: PMC8773068 DOI: 10.3390/biomedicines10010127] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Revised: 12/31/2021] [Accepted: 01/05/2022] [Indexed: 02/06/2023] Open
Abstract
Extensive research work has been carried out to define the exact significance and contribution of regulated necrosis-like cell death program, such as necroptosis to cardiac ischemic injury. This cell damaging process plays a critical role in the pathomechanisms of myocardial infarction (MI) and post-infarction heart failure (HF). Accordingly, it has been documented that the modulation of key molecules of the canonical signaling pathway of necroptosis, involving receptor-interacting protein kinases (RIP1 and RIP3) as well as mixed lineage kinase domain-like pseudokinase (MLKL), elicit cardioprotective effects. This is evidenced by the reduction of the MI-induced infarct size, alleviation of myocardial dysfunction, and adverse cardiac remodeling. In addition to this molecular signaling of necroptosis, the non-canonical pathway, involving Ca2+/calmodulin-dependent protein kinase II (CaMKII)-mediated regulation of mitochondrial permeability transition pore (mPTP) opening, and phosphoglycerate mutase 5 (PGAM5)–dynamin-related protein 1 (Drp-1)-induced mitochondrial fission, has recently been linked to ischemic heart injury. Since MI and HF are characterized by an imbalance between reactive oxygen species production and degradation as well as the occurrence of necroptosis in the heart, it is likely that oxidative stress (OS) may be involved in the mechanisms of this cell death program for inducing cardiac damage. In this review, therefore, several observations from different studies are presented to support this paradigm linking cardiac OS, the canonical and non-canonical pathways of necroptosis, and ischemia-induced injury. It is concluded that a multiple therapeutic approach targeting some specific changes in OS and necroptosis may be beneficial in improving the treatment of ischemic heart disease.
Collapse
Affiliation(s)
- Adriana Adameova
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Comenius University in Bratislava, 83232 Bratislava, Slovakia;
- Centre of Experimental Medicine, Institute for Heart Research, Slovak Academy of Sciences, 81438 Bratislava, Slovakia
- Correspondence:
| | - Csaba Horvath
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Comenius University in Bratislava, 83232 Bratislava, Slovakia;
| | - Safa Abdul-Ghani
- Department of Physiology, Faculty of Medicine, Al-Quds University, Abu Dis P.O. Box 89, Palestine;
| | - Zoltan V. Varga
- HCEMM-SU Cardiometabolic Immunology Research Group, Department of Pharmacology and Pharmacotherapy, Semmelweis University, 1089 Budapest, Hungary;
| | - M. Saadeh Suleiman
- Faculty of Health Sciences, Bristol Heart Institute, The Bristol Medical School, University of Bristol, Bristol BS8 1TH, UK;
| | - Naranjan S. Dhalla
- Institute of Cardiovascular Sciences, St. Boniface Hospital Albrechtsen Research Center, 351 Tache Avenue, Winnipeg, MB R2H 2A6, Canada;
- Department of Physiology and Pathophysiology, Max Rady College of Medicine, Faculty of Health Sciences, University of Manitoba, Winnipeg, MB R3E 0J9, Canada
| |
Collapse
|
24
|
Hameed H, Farooq M, Piquet-Pellorce C, Hamon A, Samson M, Le Seyec J. Questioning the RIPK1 kinase activity involvement in acetaminophen-induced hepatotoxicity in mouse. Free Radic Biol Med 2022; 178:243-245. [PMID: 34879229 DOI: 10.1016/j.freeradbiomed.2021.11.042] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Revised: 11/18/2021] [Accepted: 11/30/2021] [Indexed: 12/14/2022]
Affiliation(s)
- Huma Hameed
- Univ Rennes, Inserm, EHESP, IRSET (Institut de recherche en santé, environnement et travail) - UMR_S, 1085, Rennes, France
| | - Muhammad Farooq
- Univ Rennes, Inserm, EHESP, IRSET (Institut de Recherche en santé, environnement et travail) - UMR_S, 1085, Rennes, France; Department of Clinical Sciences, College of Veterinary and Animal Sciences, Jhang, Pakistan
| | - Claire Piquet-Pellorce
- Univ Rennes, Inserm, EHESP, IRSET (Institut de Recherche en santé, environnement et travail) - UMR_S, 1085, Rennes, France
| | - Annaïg Hamon
- Univ Rennes, Inserm, EHESP, IRSET (Institut de Recherche en santé, environnement et travail) - UMR_S, 1085, Rennes, France
| | - Michel Samson
- Univ Rennes, Inserm, EHESP, IRSET (Institut de Recherche en santé, environnement et travail) - UMR_S, 1085, Rennes, France.
| | - Jacques Le Seyec
- Univ Rennes, Inserm, EHESP, IRSET (Institut de Recherche en santé, environnement et travail) - UMR_S, 1085, Rennes, France
| |
Collapse
|
25
|
Jantas D, Lasoń W. Preclinical Evidence for the Interplay between Oxidative Stress and RIP1-Dependent Cell Death in Neurodegeneration: State of the Art and Possible Therapeutic Implications. Antioxidants (Basel) 2021; 10:antiox10101518. [PMID: 34679652 PMCID: PMC8532910 DOI: 10.3390/antiox10101518] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 09/17/2021] [Accepted: 09/21/2021] [Indexed: 12/15/2022] Open
Abstract
Neurodegenerative diseases are the most frequent chronic, age-associated neurological pathologies having a major impact on the patient’s quality of life. Despite a heavy medical, social and economic burden they pose, no causative treatment is available for these diseases. Among the important pathogenic factors contributing to neuronal loss during neurodegeneration is elevated oxidative stress resulting from a disturbed balance between endogenous prooxidant and antioxidant systems. For many years, it was thought that increased oxidative stress was a cause of neuronal cell death executed via an apoptotic mechanism. However, in recent years it has been postulated that rather programmed necrosis (necroptosis) is the key form of neuronal death in the course of neurodegenerative diseases. Such assumption was supported by biochemical and morphological features of the dying cells as well as by the fact that various necroptosis inhibitors were neuroprotective in cellular and animal models of neurodegenerative diseases. In this review, we discuss the relationship between oxidative stress and RIP1-dependent necroptosis and apoptosis in the context of the pathomechanism of neurodegenerative disorders. Based on the published data mainly from cellular models of neurodegeneration linking oxidative stress and necroptosis, we postulate that administration of multipotential neuroprotectants with antioxidant and antinecroptotic properties may constitute an efficient pharmacotherapeutic strategy for the treatment of neurodegenerative diseases.
Collapse
|
26
|
Yu J, Zhong B, Zhao L, Hou Y, Wang X, Chen X. Receptor-interacting serine/threonine-protein kinase 1 (RIPK1) inhibitors Necrostatin-1 (Nec-1) and 7-Cl-O-Nec-1 (Nec-1s) are potent inhibitors of NAD(P)H: Quinone oxidoreductase 1 (NQO1). Free Radic Biol Med 2021; 173:64-69. [PMID: 34252539 DOI: 10.1016/j.freeradbiomed.2021.07.017] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Revised: 07/01/2021] [Accepted: 07/07/2021] [Indexed: 11/20/2022]
Abstract
Receptor-interacting serine/threonine-protein kinase 1 (RIPK1) has been identified as a critical mediator of cell death (necroptosis and apoptosis) and inflammation. Necrostatin-1 (Nec-1) and 7-Cl-O-Nec-1 (Nec-1s) are widely used as selective small-molecule inhibitors of RIPK1 in various culture cells and disease models. NAD(P)H: quinone oxidoreductase 1 (NQO1) is a ubiquitous flavoenzyme that catalyzes the reduction and detoxification of quinones and other organic compounds. Here, we showed that Nec-1 and Nec-1s could bind and inhibit NQO1 activity. Similar to dicoumarol, the specific inhibitor of NQO1, both Nec-1 and Nec-1s significantly suppress NQO1-dependent cell death. However, dicoumarol failed to reverse necroptosis induced by TNFα/BV6/Z-VAD-FMK (TBZ) in HT29 cells. These findings suggest that besides RIPK1, NQO1 might be another target for Nec-1 and Nec-1s and provide new insights for the interpretation of Nec-1-based experimental results.
Collapse
Affiliation(s)
- Jie Yu
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau, China; College of Life Science and Technology, Wuhan Polytechnic University, Wuhan, China
| | - Bingling Zhong
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau, China
| | - Lin Zhao
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau, China
| | - Ying Hou
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau, China
| | - Xianzhe Wang
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau, China
| | - Xiuping Chen
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau, China; Department of Pharmaceutical Sciences, Faculty of Health Sciences, University of Macau, Macau, China.
| |
Collapse
|
27
|
Necrostatin-1 Prevents Ferroptosis in a RIPK1- and IDO-Independent Manner in Hepatocellular Carcinoma. Antioxidants (Basel) 2021; 10:antiox10091347. [PMID: 34572979 PMCID: PMC8469492 DOI: 10.3390/antiox10091347] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2021] [Revised: 08/15/2021] [Accepted: 08/17/2021] [Indexed: 01/31/2023] Open
Abstract
Ferroptosis is caused by the iron-mediated accumulation of lipid peroxidation, which is distinct from apoptosis and necroptosis. Necrostatin-1 inhibits receptor-interacting serine/threonine-protein kinase 1 (RIPK1) to initiate necroptosis; it also inhibits indoleamine 2,3-dioxygenase (IDO) to regulate tumor immunity. However, few studies have examined the off-target effect of necrostatin-1 on the ferroptosis pathway. The present study examined whether necrostatin-1 could interrupt ferroptosis induced by system xc- inhibitors (sulfasalazine and erastin) and a glutathione peroxidase 4 inhibitor (RSL3) in Huh7 and SK-HEP-1 cells. Necrostatin-1 completely prevented decreases in cell viability induced by sulfasalazine and erastin; it partially blunted decreases in cell viability induced by RSL3. Necrostatin-1, ferrostatin-1, and deferoxamine repressed sulfasalazine-provoked membrane permeabilization, as detected by 7-aminoactinomycin D staining and lipid peroxidation measured using a C11-BODIPY probe. However, other RIPK1 inhibitors (necrostatin-1s and GSK2982772) and an IDO inhibitor (1-methyl-D-tryptophan) did not recover the decrease in cell viability induced by sulfasalazine. Necrostatin-1 potentiated sulfasalazine-induced expression of xCT, a catalytic subunit of system xc- in these cells. These results demonstrated that necrostatin-1 blocked ferroptosis through a mechanism independent from RIPK1 and IDO inhibition in Huh7 and SK-HEP-1 cells, indicating that its antioxidant activity should be considered when using necrostatin-1 as a RIPK1 inhibitor.
Collapse
|
28
|
Lima IS, Pêgo AC, Barros JT, Prada AR, Gozzelino R. Cell Death-Osis of Dopaminergic Neurons and the Role of Iron in Parkinson's Disease. Antioxid Redox Signal 2021; 35:453-473. [PMID: 33233941 DOI: 10.1089/ars.2020.8229] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Significance: There is still no cure for neurodegenerative diseases, such as Parkinson's disease (PD). Current treatments are based on the attempt to reduce dopaminergic neuronal loss, and multidisciplinary approaches have been used to provide only a temporary symptoms' relief. In addition to the difficulties of drugs developed against PD to access the brain, the specificity of those inhibitory compounds could be a concern. This because neurons might degenerate by activating distinct signaling pathways, which are often initiated by the same stimulus. Recent Advances: Apoptosis, necroptosis, and ferroptosis were shown to significantly contribute to PD progression and, so far, are the main death programs described as capable to alter brain homeostasis. Their activation is characterized by different biochemical and morphological features, some of which might even share the same molecular players. Critical Issues: If there is a pathological need to engage, in PD, multiple death programs, sequentially or simultaneously, is not clear yet. Possibly the activation of apoptosis, necroptosis, and/or ferroptosis correlates to different PD stages and symptom severities. This would imply that the efficacy of therapeutic approaches against neuronal death might depend on the death program they target and the relevance of this death pathway on a specific PD phase. Future Directions: In this review, we describe the molecular mechanisms underlying the activation of apoptosis, necroptosis, and ferroptosis in PD. Understanding the interrelationship between different death pathways' activation in PD is of utmost importance for the development of therapeutic approaches against disease progression. Antioxid. Redox Signal. 35, 453-473.
Collapse
Affiliation(s)
- Illyane Sofia Lima
- Inflammation and Neurodegeneration Laboratory, Centro de Estudos de Doenças Crónicas (CEDOC)/NOVA Medical School, Universidade NOVA de Lisboa, Lisbon, Portugal
| | - Ana Catarina Pêgo
- Inflammation and Neurodegeneration Laboratory, Centro de Estudos de Doenças Crónicas (CEDOC)/NOVA Medical School, Universidade NOVA de Lisboa, Lisbon, Portugal
| | - João Tomas Barros
- Inflammation and Neurodegeneration Laboratory, Centro de Estudos de Doenças Crónicas (CEDOC)/NOVA Medical School, Universidade NOVA de Lisboa, Lisbon, Portugal
| | - Ana Rita Prada
- Inflammation and Neurodegeneration Laboratory, Centro de Estudos de Doenças Crónicas (CEDOC)/NOVA Medical School, Universidade NOVA de Lisboa, Lisbon, Portugal
| | - Raffaella Gozzelino
- Inflammation and Neurodegeneration Laboratory, Centro de Estudos de Doenças Crónicas (CEDOC)/NOVA Medical School, Universidade NOVA de Lisboa, Lisbon, Portugal.,Universidade Técnica do Atlântico (UTA), São Vicente, Cabo Verde
| |
Collapse
|
29
|
Oliveira SR, Dionísio PA, Gaspar MM, Ferreira MBT, Rodrigues CAB, Pereira RG, Estevão MS, Perry MJ, Moreira R, Afonso CAM, Amaral JD, Rodrigues CMP. Discovery of a Necroptosis Inhibitor Improving Dopaminergic Neuronal Loss after MPTP Exposure in Mice. Int J Mol Sci 2021; 22:ijms22105289. [PMID: 34069782 PMCID: PMC8157267 DOI: 10.3390/ijms22105289] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2021] [Revised: 05/10/2021] [Accepted: 05/14/2021] [Indexed: 11/29/2022] Open
Abstract
Parkinson’s disease (PD) is the second most common neurodegenerative disorder, mainly characterized by motor deficits correlated with progressive dopaminergic neuronal loss in the substantia nigra pars compacta (SN). Necroptosis is a caspase-independent form of regulated cell death mediated by the concerted action of receptor-interacting protein 3 (RIP3) and the pseudokinase mixed lineage domain-like protein (MLKL). It is also usually dependent on RIP1 kinase activity, influenced by further cellular clues. Importantly, necroptosis appears to be strongly linked to several neurodegenerative diseases, including PD. Here, we aimed at identifying novel chemical inhibitors of necroptosis in a PD-mimicking model, by conducting a two-step screening. Firstly, we phenotypically screened a library of 31 small molecules using a cellular model of necroptosis and, thereafter, the hit compound effect was validated in vivo in a sub-acute 1-methyl-1-4-phenyl-1,2,3,6-tetrahydropyridine hydrochloride (MPTP) PD-related mouse model. From the initial compounds, we identified one hit—Oxa12—that strongly inhibited necroptosis induced by the pan-caspase inhibitor zVAD-fmk in the BV2 murine microglia cell line. More importantly, mice exposed to MPTP and further treated with Oxa12 showed protection against MPTP-induced dopaminergic neuronal loss in the SN and striatum. In conclusion, we identified Oxa12 as a hit compound that represents a new chemotype to tackle necroptosis. Oxa12 displays in vivo effects, making this compound a drug candidate for further optimization to attenuate PD pathogenesis.
Collapse
|
30
|
Moin I, Biswas L, Zafaryab M, Kumari N, Leekha A, Mittal D, Verma AK. In vitro Toxico-genomics of Etoposide Loaded Gelatin Nanoparticles and Its in-vivo Therapeutic Potential: Pharmacokinetics, Biodistribution and Tumor Regression in Ehrlich Ascites Carcinoma (EAC) Mice Model. FRONTIERS IN NANOTECHNOLOGY 2021. [DOI: 10.3389/fnano.2021.624083] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Globally, breast cancer is the foremost cause of mortality among women detected with cancer, with 21% diagnosed in India alone. Etoposide loaded gelatin nanoparticles (EGNP) were prepared and its physical characterization (size:150nm±0.241; zeta potential −29.32 mV) was done along with in-vitro studies to assess biotoxicity, intracellular ROS, cell cycle arrest and death caused by EGNPs. We report the molecular pathways induced by EGNP in-vitro, pharmacokinetics, biodistribution and tumor regression in-vivo in Balb/c mice.Gene expression profiling of Bax, Bcl2, p53, Caspase-3, RIPK1, RIPK3 and ß-actin as internal control were done by RT-PCR wherein Etoposide and EGNP treated MCF-7 cells showed higher expressions of apoptotic genes-Bax, p53, caspase-3, lower expression of anti-apoptotic gene-Bcl2 when compared to control. Enhanced expression of necroptosis-RIPK1 were observed, while RIPK3 was insignificant. Since, RIPK1 regulates necroptosis and apoptosis, expression of apoptotic markers confirmed apoptotic molecular mechanisms. Negligible hemolysis of Gelatin nanoparticles (GNP), and EGNP at selected dosages confirmed biocompatibility. In vivo pharmacokinetics and biodistribution were done by 99Tc-labelled nanoparticles indicating increased circulation of EGNPs, allowing accumulation at the tumor site by Enhanced permeability and retention (EPR) phenomena. Tumor regression indicates the efficacy of EGNP by reducing the tumor burden when compared to void GNP and Etop per se, resulting in increased life span. High biocompatibility and bio-efficacy of EGNPs prove their therapeutic potential in cancer treatment.
Collapse
|
31
|
An G, Liu J. TfOH/DMEDA‐Mediated Cyclization of 2‐(3‐Arylthioureido)‐amide to Form 2‐Thioimidazoline‐4‐ones. ChemistrySelect 2021. [DOI: 10.1002/slct.202100661] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Affiliation(s)
- Guanghui An
- School of Petroleum and Chemical Engineering Dalian University of Technology, Panjin Campus Panjin Liaoning Province 124221 P. R. China
| | - Jianhui Liu
- School of Petroleum and Chemical Engineering Dalian University of Technology, Panjin Campus Panjin Liaoning Province 124221 P. R. China
- State Key Laboratory of Fine Chemicals Dalian University of Technology Dalian Liaoning Province 116024 P. R. China
| |
Collapse
|
32
|
Pleiotropic, non-cell death-associated effects of inhibitors of receptor-interacting protein kinase 1 in the heart. Mol Cell Biochem 2021; 476:3079-3087. [PMID: 33811579 DOI: 10.1007/s11010-021-04136-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Accepted: 03/11/2021] [Indexed: 12/25/2022]
Abstract
Inhibition of receptor-interacting protein kinase 1 (RIP1) has been recognized as a compelling tool for limiting necroptosis. Recent findings have indicated that RIP1 inhibitor, necrostatin-1 (Nec-1), is also able to modify heart function under non-cell death conditions. In this study, we investigated its underlying molecular mechanisms and compared with those of novel pharmacologically improved agents (Nec-1s and GSK'772) and its inactive analog (Nec-1i). Heart function was examined in Langendorff-perfused rat hearts. Certain proteins regulating myocardial contraction-relaxation cycle and oxidative stress (OS) were evaluated by immunoblotting and as the extent of lipid peroxidation, protein carbonylation and nitration, respectively. In spite of the increase of left ventricular developed pressure (LVDP) due to treatment by both Nec-1 and Nec-1i, only the former agent increased the phosphorylation of Ca2+/calmodulin-dependent protein kinase II delta (CaMKIIδ) at threonine 287 and cardiac myosin-binding protein-C (cMyBPc) at serine 282. In contrast, Nec-1s did not elicit such changes, while it also increased LVDP. GSK'772 activated CaMKIIδ-phospholamban (PLN) axis. Neither protein kinase A (PKA) nor its selected molecular targets, such as serine 16 phosphorylated PLN and sarco/endoplasmic reticulum Ca2+-ATPase 2a (SERCA2a), were affected by either RIP1 inhibitor. Nec-1, like other necrostatins (Nec-1i, Nec-1s), but not GSK'772, elevated protein tyrosine nitration without affecting other markers of OS. In conclusion, this study indicated for the first time that Nec-1 may affect basal heart function by the modulation of OS and activation of some proteins of contraction-relaxation cycle.
Collapse
|
33
|
Guiney SJ, Adlard PA, Lei P, Mawal CH, Bush AI, Finkelstein DI, Ayton S. Fibrillar α-synuclein toxicity depends on functional lysosomes. J Biol Chem 2021; 295:17497-17513. [PMID: 33453994 DOI: 10.1074/jbc.ra120.013428] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2020] [Revised: 10/01/2020] [Indexed: 02/05/2023] Open
Abstract
Neurodegeneration in Parkinson's disease (PD) can be recapitulated in animals by administration of α-synuclein preformed fibrils (PFFs) into the brain. However, the mechanism by which these PFFs induce toxicity is unknown. Iron is implicated in PD pathophysiology, so we investigated whether α-synuclein PFFs induce ferroptosis, an iron-dependent cell death pathway. A range of ferroptosis inhibitors were added to a striatal neuron-derived cell line (STHdhQ7/7 cells), a dopaminergic neuron-derived cell line (SN4741 cells), and WT primary cortical neurons, all of which had been intoxicated with α-synuclein PFFs. Viability was not recovered by these inhibitors except for liproxstatin-1, a best-in-class ferroptosis inhibitor, when used at high doses. High-dose liproxstatin-1 visibly enlarged the area of a cell that contained acidic vesicles and elevated the expression of several proteins associated with the autophagy-lysosomal pathway similarly to the known lysosomal inhibitors, chloroquine and bafilomycin A1. Consistent with high-dose liproxstatin-1 protecting via a lysosomal mechanism, we further de-monstrated that loss of viability induced by α-synuclein PFFs was attenuated by chloroquine and bafilomycin A1 as well as the lysosomal cysteine protease inhibitors, leupeptin, E-64D, and Ca-074-Me, but not other autophagy or lysosomal enzyme inhibitors. We confirmed using immunofluorescence microscopy that heparin prevented uptake of α-synuclein PFFs into cells but that chloroquine did not stop α-synuclein uptake into lysosomes despite impairing lysosomal function and inhibiting α-synuclein toxicity. Together, these data suggested that α-synuclein PFFs are toxic in functional lysosomes in vitro. Therapeutic strategies that prevent α-synuclein fibril uptake into lysosomes may be of benefit in PD.
Collapse
Affiliation(s)
- Stephanie J Guiney
- Melbourne Dementia Research Centre, Parkville, Victoria, Australia; Florey Institute of Neuroscience and Mental Health, Parkville, Victoria, Australia; University of Melbourne, Parkville, Victoria Australia
| | - Paul A Adlard
- Melbourne Dementia Research Centre, Parkville, Victoria, Australia; Florey Institute of Neuroscience and Mental Health, Parkville, Victoria, Australia; University of Melbourne, Parkville, Victoria Australia
| | - Peng Lei
- Melbourne Dementia Research Centre, Parkville, Victoria, Australia; Florey Institute of Neuroscience and Mental Health, Parkville, Victoria, Australia; University of Melbourne, Parkville, Victoria Australia; Department of Neurology and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University/Collaborative Center for Biotherapy, Chengdu, China
| | - Celeste H Mawal
- Melbourne Dementia Research Centre, Parkville, Victoria, Australia; Florey Institute of Neuroscience and Mental Health, Parkville, Victoria, Australia
| | - Ashley I Bush
- Melbourne Dementia Research Centre, Parkville, Victoria, Australia; Florey Institute of Neuroscience and Mental Health, Parkville, Victoria, Australia; University of Melbourne, Parkville, Victoria Australia
| | - David I Finkelstein
- Melbourne Dementia Research Centre, Parkville, Victoria, Australia; Florey Institute of Neuroscience and Mental Health, Parkville, Victoria, Australia; University of Melbourne, Parkville, Victoria Australia
| | - Scott Ayton
- Melbourne Dementia Research Centre, Parkville, Victoria, Australia; Florey Institute of Neuroscience and Mental Health, Parkville, Victoria, Australia; University of Melbourne, Parkville, Victoria Australia.
| |
Collapse
|
34
|
Deng F, Zheng X, Sharma I, Dai Y, Wang Y, Kanwar YS. Regulated cell death in cisplatin-induced AKI: relevance of myo-inositol metabolism. Am J Physiol Renal Physiol 2021; 320:F578-F595. [PMID: 33615890 PMCID: PMC8083971 DOI: 10.1152/ajprenal.00016.2021] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Revised: 02/08/2021] [Accepted: 02/09/2021] [Indexed: 12/14/2022] Open
Abstract
Regulated cell death (RCD), distinct from accidental cell death, refers to a process of well-controlled programmed cell death with well-defined pathological mechanisms. In the past few decades, various terms for RCDs were coined, and some of them have been implicated in the pathogenesis of various types of acute kidney injury (AKI). Cisplatin is widely used as a chemotherapeutic drug for a broad spectrum of cancers, but its usage was hampered because of being highly nephrotoxic. Cisplatin-induced AKI is commonly seen clinically, and it also serves as a well-established prototypic model for laboratory investigations relevant to acute nephropathy affecting especially the tubular compartment. Literature reports over a period of three decades have indicated that there are multiple types of RCDs, including apoptosis, necroptosis, pyroptosis, ferroptosis, and mitochondrial permeability transition-mediated necrosis, and some of them are pertinent to the pathogenesis of cisplatin-induced AKI. Interestingly, myo-inositol metabolism, a vital biological process that is largely restricted to the kidney, seems to be relevant to the pathogenesis of certain forms of RCDs. A comprehensive understanding of RCDs in cisplatin-induced AKI and their relevance to myo-inositol homeostasis may yield novel therapeutic targets for the amelioration of cisplatin-related nephropathy.
Collapse
Affiliation(s)
- Fei Deng
- Department of Urology, The Second Xiangya Hospital, Central South University, Changsha, China
- Department of Pathology, Northwestern University, Chicago, Illinois
- Department of Medicine, Northwestern University, Chicago, Illinois
| | - Xiaoping Zheng
- Department of Pathology, Northwestern University, Chicago, Illinois
- Department of Medicine, Northwestern University, Chicago, Illinois
- Department of Urology, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Isha Sharma
- Department of Pathology, Northwestern University, Chicago, Illinois
- Department of Medicine, Northwestern University, Chicago, Illinois
| | - Yingbo Dai
- Department of Urology, The Third Xiangya Hospital, Central South University, Changsha, China
- Department of Urology, The Fifth Affiliated Hospital of Sun Yet-Sen University, Zhuhai, China
| | - Yinhuai Wang
- Department of Urology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Yashpal S Kanwar
- Department of Pathology, Northwestern University, Chicago, Illinois
- Department of Medicine, Northwestern University, Chicago, Illinois
| |
Collapse
|
35
|
Nah J, Zhai P, Huang CY, Fernández ÁF, Mareedu S, Levine B, Sadoshima J. Upregulation of Rubicon promotes autosis during myocardial ischemia/reperfusion injury. J Clin Invest 2021; 130:2978-2991. [PMID: 32364533 DOI: 10.1172/jci132366] [Citation(s) in RCA: 82] [Impact Index Per Article: 27.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2019] [Accepted: 02/20/2020] [Indexed: 12/29/2022] Open
Abstract
Although autophagy is generally protective, uncontrolled or excessive activation of autophagy can be detrimental. However, it is often difficult to distinguish death by autophagy from death with autophagy, and whether autophagy contributes to death in cardiomyocytes (CMs) is still controversial. Excessive activation of autophagy induces a morphologically and biochemically defined form of cell death termed autosis. Whether autosis is involved in tissue injury induced under pathologically relevant conditions is poorly understood. In the present study, myocardial ischemia/reperfusion (I/R) induced autosis in CMs, as evidenced by cell death with numerous vacuoles and perinuclear spaces, and depleted intracellular membranes. Autosis was observed frequently after 6 hours of reperfusion, accompanied by upregulation of Rubicon, attenuation of autophagic flux, and marked accumulation of autophagosomes. Genetic downregulation of Rubicon inhibited autosis and reduced I/R injury, whereas stimulation of autosis during the late phase of I/R with Tat-Beclin 1 exacerbated injury. Suppression of autosis by ouabain, a cardiac glycoside, in humanized Na+,K+-ATPase-knockin mice reduced I/R injury. Taken together, these results demonstrate that autosis is significantly involved in I/R injury in the heart and triggered by dysregulated accumulation of autophagosomes due to upregulation of Rubicon.
Collapse
Affiliation(s)
- Jihoon Nah
- Cardiovascular Research Institute, Department of Cell Biology and Molecular Medicine, Rutgers New Jersey Medical School, Newark, New Jersey, USA
| | - Peiyong Zhai
- Cardiovascular Research Institute, Department of Cell Biology and Molecular Medicine, Rutgers New Jersey Medical School, Newark, New Jersey, USA
| | - Chun-Yang Huang
- Cardiovascular Research Institute, Department of Cell Biology and Molecular Medicine, Rutgers New Jersey Medical School, Newark, New Jersey, USA
| | | | - Satvik Mareedu
- Cardiovascular Research Institute, Department of Cell Biology and Molecular Medicine, Rutgers New Jersey Medical School, Newark, New Jersey, USA
| | - Beth Levine
- Center for Autophagy Research, Department of Internal Medicine, and.,Howard Hughes Medical Institute, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Junichi Sadoshima
- Cardiovascular Research Institute, Department of Cell Biology and Molecular Medicine, Rutgers New Jersey Medical School, Newark, New Jersey, USA
| |
Collapse
|
36
|
Urban Aerosol Particulate Matter Promotes Necrosis and Autophagy via Reactive Oxygen Species-Mediated Cellular Disorders that are Accompanied by Cell Cycle Arrest in Retinal Pigment Epithelial Cells. Antioxidants (Basel) 2021; 10:antiox10020149. [PMID: 33498524 PMCID: PMC7909535 DOI: 10.3390/antiox10020149] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Revised: 01/15/2021] [Accepted: 01/15/2021] [Indexed: 12/12/2022] Open
Abstract
Urban particulate matter (UPM) is recognized as a grave public health problem worldwide. Although a few studies have linked UPM to ocular surface diseases, few studies have reported on retinal dysfunction. Thus, the aim of the present study was to evaluate the influence of UPM on the retina and identify the main mechanism of UPM toxicity. In this study, we found that UPM significantly induced cytotoxicity with morphological changes in ARPE-19 human retinal pigment epithelial (RPE) cells and increased necrosis and autophagy but not apoptosis. Furthermore, UPM significantly increased G2/M arrest and simultaneously induced alterations in cell cycle regulators. In addition, DNA damage and mitochondrial dysfunction were remarkably enhanced by UPM. However, the pretreatment with the potent reactive oxygen species (ROS) scavenger N-acetyl-L-cysteine (NAC) effectively suppressed UPM-mediated cytotoxicity, necrosis, autophagy, and cell cycle arrest. Moreover, NAC markedly restored UPM-induced DNA damage and mitochondrial dysfunction. Meanwhile, UPM increased the expression of mitophagy-regulated proteins, but NAC had no effect on mitophagy. Taken together, although further studies are needed to identify the role of mitophagy in UPM-induced RPE injury, the present study provides the first evidence that ROS-mediated cellular damage through necrosis and autophagy is one of the mechanisms of UPM-induced retinal disorders.
Collapse
|
37
|
Liu Y, Xu Q, Wang Y, Liang T, Li X, Wang D, Wang X, Zhu H, Xiao K. Necroptosis is active and contributes to intestinal injury in a piglet model with lipopolysaccharide challenge. Cell Death Dis 2021; 12:62. [PMID: 33431831 PMCID: PMC7801412 DOI: 10.1038/s41419-020-03365-1] [Citation(s) in RCA: 46] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Revised: 12/13/2020] [Accepted: 12/17/2020] [Indexed: 12/24/2022]
Abstract
Necroptosis, a newly discovered form of programmed cell death that combines the features of apoptosis and necrosis, is important in various physiological and pathological disorders. However, the role of necroptosis on intestinal injury during sepsis has been rarely evaluated. This study aimed to investigate the presence of necroptosis in intestinal injury, and its contribution to intestinal injury in a piglet model challenged with Escherichia coli lipopolysaccharide (LPS). Firstly, a typical cell necrotic phenomenon was observed in jejunum of LPS-challenged pigs by transmission electron microscope. Protein expression of necroptosis signals including receptor-interacting protein kinase (RIP) 1, RIP3, and phosphorylated mixed-lineage kinase domain-like protein (MLKL), mitochondrial proteins including phosphoglycerate mutase family member 5 (PGAM5) and dynamin-related protein 1 (DRP1), and cytoplasmic high-mobility group box 1 (HMGB1) were time-independently increased in jejunum of LPS-challenged piglets, which was accompanied by the impairment of jejunal morphology, and digestive and barrier function indicated by lower activities of jejunal disaccharidases and protein expression of jejunal tight junction proteins claudin-1 and occludin. Pro-inflammatory cytokines including tumor necrosis factor-α (TNF-α), interleukin (IL)-1β, and IL-6 were also dynamically induced in serum and jejunum of piglets after LPS challenge. Moreover, pretreatment with necrostatin-1 (Nec-1), an specific inhibitor of necroptosis, inhibited necroptosis indicated by decreased necrotic ultrastructural changes and decreased protein expression of RIP1, RIP3, and phosphorylated MLKL as well as PGAM5, DRP1, and cytoplasmic HMGB1. Nec-1 pretreatment reduced jejunal morphological injury, and improved digestive and barrier function. Nec-1 pretreatment also decreased the levels of serum and jejunal pro-inflammatory cytokines and the numbers of jejunal macrophages and monocytes. These findings indicate for the first time that necroptosis is present and contributes to LPS-induced intestinal injury. Nec-1 may have a preventive effect on intestinal injury during sepsis.
Collapse
Affiliation(s)
- Yulan Liu
- Hubei Key Laboratory of Animal Nutrition and Feed Science, Wuhan Polytechnic University, 430023, Wuhan, China.
| | - Qiao Xu
- Hubei Key Laboratory of Animal Nutrition and Feed Science, Wuhan Polytechnic University, 430023, Wuhan, China
| | - Yang Wang
- Hubei Key Laboratory of Animal Nutrition and Feed Science, Wuhan Polytechnic University, 430023, Wuhan, China
| | - Tianzeng Liang
- Hubei Key Laboratory of Animal Nutrition and Feed Science, Wuhan Polytechnic University, 430023, Wuhan, China
| | - Xiangen Li
- Hubei Key Laboratory of Animal Nutrition and Feed Science, Wuhan Polytechnic University, 430023, Wuhan, China
| | - Dan Wang
- Hubei Key Laboratory of Animal Nutrition and Feed Science, Wuhan Polytechnic University, 430023, Wuhan, China
| | - Xiuying Wang
- Hubei Key Laboratory of Animal Nutrition and Feed Science, Wuhan Polytechnic University, 430023, Wuhan, China
| | - Huiling Zhu
- Hubei Key Laboratory of Animal Nutrition and Feed Science, Wuhan Polytechnic University, 430023, Wuhan, China
| | - Kan Xiao
- Hubei Key Laboratory of Animal Nutrition and Feed Science, Wuhan Polytechnic University, 430023, Wuhan, China
| |
Collapse
|
38
|
Dai Q, Zhang Y, Liao X, Jiang Y, Lv X, Yuan X, Meng J, Xie Y, Peng Z, Yuan Q, Tao L, Huang L. Fluorofenidone Alleviates Renal Fibrosis by Inhibiting Necroptosis Through RIPK3/MLKL Pathway. Front Pharmacol 2021; 11:534775. [PMID: 33390935 PMCID: PMC7772387 DOI: 10.3389/fphar.2020.534775] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2020] [Accepted: 11/09/2020] [Indexed: 12/24/2022] Open
Abstract
Cell death and sterile inflammation are major mechanisms of renal fibrosis, which eventually develop into end-stage renal disease. “Necroptosis” is a type of caspase-independent regulated cell death, and sterile inflammatory response caused by tissue injury is strongly related to necrosis. Fluorofenidone (AKF-PD) is a novel compound shown to ameliorate renal fibrosis and associated inflammation. We investigated whether AKF-PD could alleviate renal fibrosis by inhibiting necroptosis. Unilateral ureteral obstruction (UUO) was used to induce renal tubulointerstitial fibrosis in C57BL/6J mice. AKF-PD (500 mg/kg) or necrostatin-1 (Nec-1; 1.65 mg/kg) was administered simultaneously for 3 and 7 days. Obstructed kidneys and serum were harvested after euthanasia. AKF-PD and Nec-1 ameliorated renal tubular damage, inflammatory-cell infiltration, and collagen deposition, and the expression of proinflammatory factors (interlukin-1β, tumor necrosis factor [TNF]-α) and chemokines (monocyte chemoattractant protein-1) decreased. AKF-PD or Nec-1 treatment protected renal tubular epithelial cells from necrosis and reduced the release of lactate dehydrogenase in serum. Simultaneously, production of receptor-interacting protein kinase (RIPK)3 and mixed lineage kinase domain-like protein (MLKL) was also reduced 3 and 7 days after UUO. AKF-PD and Nec-1 significantly decreased the percentage of cell necrosis, inhibiting the phosphorylation of MLKL and RIPK3 in TNF-α- and Z-VAD–stimulated human proximal tubular epithelial (HK-2) cells. In conclusion, AKF-PD and Nec-1 have effective anti-inflammatory and antifibrotic activity in UUO-induced renal tubulointerstitial fibrosis, potentially mediated by the RIPK3/MLKL pathway.
Collapse
Affiliation(s)
- Qin Dai
- Department of Nephrology, Xiangya Hospital, Central South University, Changsha, China
| | - Yan Zhang
- Department of Nephrology, Xiangya Hospital, Central South University, Changsha, China
| | - Xiaohua Liao
- Department of Nephrology, Xiangya Hospital, Central South University, Changsha, China
| | - Yupeng Jiang
- Department of Nephrology, Xiangya Hospital, Central South University, Changsha, China
| | - Xin Lv
- Department of Nephrology, Xiangya Hospital, Central South University, Changsha, China
| | - Xiangning Yuan
- Department of Nephrology, Xiangya Hospital, Central South University, Changsha, China
| | - Jie Meng
- Department of Respirology, Xiangya Hospital, Central South University, Changsha, China
| | - Yanyun Xie
- Department of Nephrology, Xiangya Hospital, Central South University, Changsha, China
| | - Zhangzhe Peng
- Department of Nephrology, Xiangya Hospital, Central South University, Changsha, China
| | - Qiongjing Yuan
- Department of Nephrology, Xiangya Hospital, Central South University, Changsha, China
| | - LiJian Tao
- Department of Nephrology, Xiangya Hospital, Central South University, Changsha, China
| | - Ling Huang
- Department of Nephrology, Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
39
|
Benchekroun M, Ermolenko L, Tran MQ, Vagneux A, Nedev H, Delehouzé C, Souab M, Baratte B, Josselin B, Iorga BI, Ruchaud S, Bach S, Al-Mourabit A. Discovery of simplified benzazole fragments derived from the marine benzosceptrin B as necroptosis inhibitors involving the receptor interacting protein Kinase-1. Eur J Med Chem 2020; 201:112337. [DOI: 10.1016/j.ejmech.2020.112337] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2020] [Revised: 04/04/2020] [Accepted: 04/13/2020] [Indexed: 12/12/2022]
|
40
|
Baidya R, Crawford DHG, Gautheron J, Wang H, Bridle KR. Necroptosis in Hepatosteatotic Ischaemia-Reperfusion Injury. Int J Mol Sci 2020; 21:ijms21165931. [PMID: 32824744 PMCID: PMC7460692 DOI: 10.3390/ijms21165931] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Revised: 08/12/2020] [Accepted: 08/12/2020] [Indexed: 02/07/2023] Open
Abstract
While liver transplantation remains the sole treatment option for patients with end-stage liver disease, there are numerous limitations to liver transplantation including the scarcity of donor livers and a rise in livers that are unsuitable to transplant such as those with excess steatosis. Fatty livers are susceptible to ischaemia-reperfusion (IR) injury during transplantation and IR injury results in primary graft non-function, graft failure and mortality. Recent studies have described new cell death pathways which differ from the traditional apoptotic pathway. Necroptosis, a regulated form of cell death, has been associated with hepatic IR injury. Receptor-interacting protein kinase 3 (RIPK3) and mixed-lineage kinase domain-like pseudokinase (MLKL) are thought to be instrumental in the execution of necroptosis. The study of hepatic necroptosis and potential therapeutic approaches to attenuate IR injury will be a key factor in improving our knowledge regarding liver transplantation with fatty donor livers. In this review, we focus on the effect of hepatic steatosis during liver transplantation as well as molecular mechanisms of necroptosis and its involvement during liver IR injury. We also discuss the immune responses triggered during necroptosis and examine the utility of necroptosis inhibitors as potential therapeutic approaches to alleviate IR injury.
Collapse
Affiliation(s)
- Raji Baidya
- Faculty of Medicine, The University of Queensland, Brisbane, Queensland QLD 4006, Australia; (R.B.); (D.H.G.C.)
- Gallipoli Medical Research Institute, Brisbane, Queensland QLD 4120, Australia;
| | - Darrell H. G. Crawford
- Faculty of Medicine, The University of Queensland, Brisbane, Queensland QLD 4006, Australia; (R.B.); (D.H.G.C.)
- Gallipoli Medical Research Institute, Brisbane, Queensland QLD 4120, Australia;
| | - Jérémie Gautheron
- Sorbonne University, Inserm, Centre de Recherche Saint-Antoine (CRSA), 75012 Paris, France;
- Institute of Cardiometabolism and Nutrition (ICAN), 75013 Paris, France
| | - Haolu Wang
- Gallipoli Medical Research Institute, Brisbane, Queensland QLD 4120, Australia;
- Diamantina Institute, The University of Queensland, Brisbane, Queensland QLD 4102, Australia
| | - Kim R. Bridle
- Faculty of Medicine, The University of Queensland, Brisbane, Queensland QLD 4006, Australia; (R.B.); (D.H.G.C.)
- Gallipoli Medical Research Institute, Brisbane, Queensland QLD 4120, Australia;
- Correspondence: ; Tel.: +61-7-3346-0698
| |
Collapse
|
41
|
Simon Serrano S, Sime W, Abassi Y, Daams R, Massoumi R, Jemaà M. Inhibition of mitotic kinase Mps1 promotes cell death in neuroblastoma. Sci Rep 2020; 10:11997. [PMID: 32686724 PMCID: PMC7371706 DOI: 10.1038/s41598-020-68829-y] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2020] [Accepted: 06/29/2020] [Indexed: 12/27/2022] Open
Abstract
Neuroblastoma is the most common paediatric cancer type. Patients diagnosed with high-risk neuroblastoma have poor prognosis and occasionally tumours relapse. As a result, novel treatment strategies are needed for relapse and refractory neuroblastoma patients. Here, we found that high expression of Mps1 kinase (mitotic kinase Monopolar Spindle 1) was associated with relapse-free neuroblastoma patient outcomes and poor overall survival. Silencing and inhibition of Mps1 in neuroblastoma or PDX-derived cells promoted cell apoptosis via the caspase-dependent mitochondrial apoptotic pathway. The mechanism of cell death upon Mps1 inhibition was dependent on the polyploidization/aneuploidization of the cells before undergoing mitotic catastrophe. Furthermore, tumour growth retardation was confirmed in a xenograft mouse model after Mps1-inhibitor treatment. Altogether, these results suggest that Mps1 expression and inhibition can be considered as a novel prognostic marker as well as a therapeutic strategy for the treatment of high-risk neuroblastoma patients.
Collapse
Affiliation(s)
- Sonia Simon Serrano
- Department of Laboratory Medicine, Translational Cancer Research, Faculty of Medicine, Lund University, 22381, Lund, Sweden
| | - Wondossen Sime
- Department of Laboratory Medicine, Translational Cancer Research, Faculty of Medicine, Lund University, 22381, Lund, Sweden
| | - Yasmin Abassi
- Department of Laboratory Medicine, Translational Cancer Research, Faculty of Medicine, Lund University, 22381, Lund, Sweden
| | - Renée Daams
- Department of Laboratory Medicine, Translational Cancer Research, Faculty of Medicine, Lund University, 22381, Lund, Sweden
| | - Ramin Massoumi
- Department of Laboratory Medicine, Translational Cancer Research, Faculty of Medicine, Lund University, 22381, Lund, Sweden.
| | - Mohamed Jemaà
- Department of Laboratory Medicine, Translational Cancer Research, Faculty of Medicine, Lund University, 22381, Lund, Sweden.
| |
Collapse
|
42
|
Zinngrebe J, Schlichtig F, Kraus JM, Meyer M, Boldrin E, Kestler HA, Meyer L, Fischer‐Posovszky P, Debatin K. Biomarker profile for prediction of response to SMAC mimetic monotherapy in pediatric precursor B‐cell acute lymphoblastic leukemia. Int J Cancer 2020; 146:3219-3231. [DOI: 10.1002/ijc.32799] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2019] [Accepted: 11/04/2019] [Indexed: 12/23/2022]
Affiliation(s)
- Julia Zinngrebe
- Department of Pediatrics and Adolescent MedicineUlm University Medical Center Ulm Germany
| | - Ferdinand Schlichtig
- Department of Pediatrics and Adolescent MedicineUlm University Medical Center Ulm Germany
| | - Johann M. Kraus
- Institute of Medical Systems Biology, Ulm University Ulm Germany
| | - Malcolm Meyer
- Department of Pediatrics and Adolescent MedicineUlm University Medical Center Ulm Germany
| | - Elena Boldrin
- Department of Pediatrics and Adolescent MedicineUlm University Medical Center Ulm Germany
| | - Hans A. Kestler
- Institute of Medical Systems Biology, Ulm University Ulm Germany
| | - Lüder‐Hinrich Meyer
- Department of Pediatrics and Adolescent MedicineUlm University Medical Center Ulm Germany
| | | | - Klaus‐Michael Debatin
- Department of Pediatrics and Adolescent MedicineUlm University Medical Center Ulm Germany
| |
Collapse
|
43
|
Yu J, Zhong B, Xiao Q, Du L, Hou Y, Sun HS, Lu JJ, Chen X. Induction of programmed necrosis: A novel anti-cancer strategy for natural compounds. Pharmacol Ther 2020; 214:107593. [PMID: 32492512 DOI: 10.1016/j.pharmthera.2020.107593] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/25/2020] [Indexed: 02/08/2023]
Abstract
Cell death plays a critical role in organism development and the pathogenesis of diseases. Necrosis is considered a non-programmed cell death in an extreme environment. Recent advances have provided solid evidence that necrosis could be programmed and quite a few types of programmed necrosis, such as necroptosis, ferroptosis, pyroptosis, paraptosis, mitochondrial permeability transition-driven necrosis, and oncosis, have been identified. The specific biomarkers, detailed signaling, and precise pathophysiological importance of programmed necrosis are yet to be clarified, but these forms of necrosis provide novel strategies for the treatment of various diseases, including cancer. Natural compounds are a unique source of lead compounds for the discovery of anti-cancer drugs. Natural compounds can induce both apoptosis and programmed necrosis. In this review, we summarized the recent progress of programmed necrosis and introduced their natural inducers. Noptosis, which is a novel type of programmed necrosis that is strictly dependent on NAD(P)H: quinone oxidoreductase 1-derived oxidative stress was proposed. Furthermore, the anti-cancer strategies that take advantage of programmed necrosis and the main concerns from the scientific community in this regard were discussed.
Collapse
Affiliation(s)
- Jie Yu
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau, China
| | - Bingling Zhong
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau, China
| | - Qingwen Xiao
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau, China
| | - Lida Du
- Department of Surgery, University of Toronto, Ontario, Canada
| | - Ying Hou
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau, China
| | - Hong-Shuo Sun
- Department of Surgery, University of Toronto, Ontario, Canada
| | - Jin-Jian Lu
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau, China
| | - Xiuping Chen
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau, China.
| |
Collapse
|
44
|
Zeng T, Deng G, Zhong W, Gao Z, Ma S, Mo C, Li Y, Huang S, Zhou C, Lai Y, Xie S, Xie Z, Chen Y, He S, Lv Z, Gao L. Indoleamine 2, 3-dioxygenase 1enhanceshepatocytes ferroptosis in acute immune hepatitis associated with excess nitrative stress. Free Radic Biol Med 2020; 152:668-679. [PMID: 31945497 DOI: 10.1016/j.freeradbiomed.2020.01.009] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/06/2020] [Accepted: 01/09/2020] [Indexed: 12/16/2022]
Abstract
Ferroptosis is a recently recognized form of regulated cell death that is characterized by lipid peroxidation. However, the molecular mechanisms of ferroptosis in acute immune hepatitis (AIH) are largely unknown. In this study, we investigated the classical ferroptotic events in the livers of mice with concanavalin A (ConA) to induce AIH. The dramatically upregulated gene indoleamine 2, 3-dioxygenase 1 (IDO1) was identified with AIH, and its role in generation of ferroptosis and reactive nitrogen species (RNS) was assessed both in vitro and in vivo by genetic deletion or pharmacologic inhibition of IDO1. We observed that ferroptosis contributed to the ConA-induced hepatic damage, which was confirmed by the therapeutical effects of ferroptosis inhibitor (ferrostatin-1). Noteworthy, upregulation of hepatic IDO1 and nitrative stress in ConA-induced hepatic damage were also remarkably inhibited by the ferroptosis abolishment. Additionally, IDO1 deficiency contributed to ferroptosis resistance by activating solute carrier family 7 member 11 (SLC7A11; also known as xCT) expression, accompanied with the reductions of murine liver lesions and RNS. Meanwhile, IDO inhibitor 1-methyl tryptophan alleviated murine liver damage with the reduction of inducible nitric oxide synthase and 3-nitrotyrosine expression. Consistent with the results in vivo, hepatocytes-specific knockdown of IDO1 led to ferroptosis resistance upon exposure to ferroptosis-inducing compound (Erastin) in vitro, whereas IDO1 overexpression aggravated the classical ferroptotic events, and the RNS stress. Overall, these results revealed a novel molecular mechanism of ferroptosis with the key feature of nitrative stress in ConA-induced liver injury, and also identified IDO1-dependent ferroptosis as a potential target for the treatment of AIH.
Collapse
Affiliation(s)
- Ting Zeng
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, Guangdong, China
| | - Guanghui Deng
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, Guangdong, China
| | - Weichao Zhong
- Shenzhen Traditional Chinese Medicine Hospital, No.1, Fuhua Road, Futian District, Shenzhen, Guangdong, China
| | - Zhuowei Gao
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, Guangdong, China
| | - Shuoyi Ma
- Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, Guangdong, China
| | - Chan Mo
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, Guangdong, China
| | - Yunjia Li
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, Guangdong, China
| | - Sha Huang
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, Guangdong, China
| | - Chuying Zhou
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, Guangdong, China
| | - Yuqi Lai
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, Guangdong, China
| | - Shuwen Xie
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, Guangdong, China
| | - Zeping Xie
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, Guangdong, China
| | - Yuyao Chen
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, Guangdong, China
| | - Songqi He
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, Guangdong, China.
| | - Zhiping Lv
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, Guangdong, China.
| | - Lei Gao
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, Guangdong, China.
| |
Collapse
|
45
|
Abstract
Immune cells use a variety of membrane-disrupting proteins [complement, perforin, perforin-2, granulysin, gasdermins, mixed lineage kinase domain-like pseudokinase (MLKL)] to induce different kinds of death of microbes and host cells, some of which cause inflammation. After activation by proteolytic cleavage or phosphorylation, these proteins oligomerize, bind to membrane lipids, and disrupt membrane integrity. These membrane disruptors play a critical role in both innate and adaptive immunity. Here we review our current knowledge of the functions, specificity, activation, and regulation of membrane-disrupting immune proteins and what is known about the mechanisms behind membrane damage, the structure of the pores they form, how the cells expressing these lethal proteins are protected, and how cells targeted for destruction can sometimes escape death by repairing membrane damage.
Collapse
Affiliation(s)
- Xing Liu
- Center for Microbes, Development and Health; Key Laboratory of Molecular Virology and Immunology; Institut Pasteur of Shanghai; Chinese Academy of Sciences, Shanghai 200031, China;
| | - Judy Lieberman
- Program in Cellular and Molecular Medicine, Boston Children's Hospital and Department of Pediatrics, Harvard Medical School, Boston, Massachusetts 02115, USA;
| |
Collapse
|
46
|
Han XA, Jie HY, Wang JH, Zhang XM, Wang J, Yu CX, Zhang JL, He J, Chen JQ, Lai KF, Sun EW. Necrostatin-1 Ameliorates Neutrophilic Inflammation in Asthma by Suppressing MLKL Phosphorylation to Inhibiting NETs Release. Front Immunol 2020; 11:666. [PMID: 32391007 PMCID: PMC7194114 DOI: 10.3389/fimmu.2020.00666] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2019] [Accepted: 03/24/2020] [Indexed: 11/26/2022] Open
Abstract
Neutrophilic inflammation occurs during asthma exacerbation, and especially, in patients with steroid-refractory asthma, but the underlying mechanisms are poorly understood. Recently, a significant accumulation of neutrophil extracellular traps (NETs) in the airways of neutrophilic asthma has been documented, suggesting that NETs may play an important role in the pathogenesis. In this study, we firstly demonstrated that NETs could induce human airway epithelial cell damage in vitro. In a mouse asthmatic model of neutrophil-dominated airway inflammation, we found that NETs were markedly increased in bronchoalveolar lavage (BAL), and the formation of NETs exacerbated the airway inflammation. Additionally, a small-molecule drug necrostatin-1 (Nec-1) shown to inhibit NETs formation was found to alleviate the neutrophil-dominated airway inflammation. Nec-1 reduced total protein concentration, myeloperoxidase activity, and the levels of inflammatory cytokines in BAL. Finally, further experiments proved that the inhibition of Nec-1 on NETs formation might be related to its ability to inhibiting mixed lineage kinase domain-like (MLKL) phosphorylation and perforation. Together, these results document that NETs are closely associated with the pathogenesis of neutrophilic asthma and inhibition of the formation of NETs by Nec-1 may be a new therapeutic strategy to ameliorate neutrophil-dominated airway inflammation.
Collapse
Affiliation(s)
- X A Han
- Department of Rheumatology and Immunology, The Third Affiliated Hospital, Southern Medical University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, The Third Affiliated Hospital, Southern Medical University, Guangzhou, China
| | - H Y Jie
- Department of Rheumatology and Immunology, The Third Affiliated Hospital, Southern Medical University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, The Third Affiliated Hospital, Southern Medical University, Guangzhou, China
| | - J H Wang
- Department of Respiratory and Critical Care Medicine, The Third Affiliated Hospital, Southern Medical University, Guangzhou, China
| | - X M Zhang
- Department of Respiratory and Critical Care Medicine, The Third Affiliated Hospital, Southern Medical University, Guangzhou, China
| | - Jun Wang
- Department of Respiration, Nan Fang Hospital, Southern Medical University, Guangzhou, China
| | - C X Yu
- Department of Rheumatology and Immunology, The Third Affiliated Hospital, Southern Medical University, Guangzhou, China
| | - J L Zhang
- Department of Rheumatology and Immunology, The Third Affiliated Hospital, Southern Medical University, Guangzhou, China
| | - J He
- Department of Rheumatology and Immunology, Peking University Shenzhen Hospital, Shenzhen, China
| | - J Q Chen
- Department of Rehabilitation Medicine, The Third Affiliated Hospital, Southern Medical University, Guangzhou, China
| | - K F Lai
- State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Disease, The First Affiliated Hospital, Guangzhou Medical College, Guangzhou, China
| | - E W Sun
- Department of Rheumatology and Immunology, The Third Affiliated Hospital, Southern Medical University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, The Third Affiliated Hospital, Southern Medical University, Guangzhou, China
| |
Collapse
|
47
|
Moros M, Lewinska A, Merola F, Ferraro P, Wnuk M, Tino A, Tortiglione C. Gold Nanorods and Nanoprisms Mediate Different Photothermal Cell Death Mechanisms In Vitro and In Vivo. ACS APPLIED MATERIALS & INTERFACES 2020; 12:13718-13730. [PMID: 32134240 DOI: 10.1021/acsami.0c02022] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Photothermal therapy (PTT) is an efficient method of inducing localized hyperthermia and can be achieved using gold nanoparticles as photothermal agents. However, there are many hurdles to get over before this therapy can safely reach the clinics, including nanoparticles' optimal shape and the accurate prediction of cellular responses. Here, we describe the synthesis of gold nanorods and nanoprisms with similar surface plasmon resonances in the near-infrared (NIR) and comparable photothermal conversion efficiencies and characterize the response to NIR irradiation in two biological systems, melanoma cells and the small invertebrate Hydra vulgaris. By integrating animal, cellular, and molecular biology approaches, we show a diverse outcome of nanorods and nanoprisms on the two systems, sustained by the elicitation of different pathways, from necrosis to programmed cell death mechanisms (apoptosis and necroptosis). The comparative multilevel analysis shows great accuracy of in vivo invertebrate models to predict overall responses to photothermal challenging and superior photothermal performance of nanoprisms. Understanding the molecular pathways of these responses may help develop optimized nanoheaters that, safe by design, may improve PTT efficacy for clinical purposes.
Collapse
Affiliation(s)
- Maria Moros
- Istituto di Scienze Applicate e Sistemi Intelligenti "Eduardo Caianiello", Consiglio Nazionale delle Ricerche, Via Campi Flegrei 34, 80078 Pozzuoli, Italy
| | - Anna Lewinska
- Department of Cell Biochemistry, Faculty of Biotechnology, University of Rzeszow, Pigonia 1, 35-310 Rzeszow, Poland
| | - Francesco Merola
- Istituto di Scienze Applicate e Sistemi Intelligenti "Eduardo Caianiello", Consiglio Nazionale delle Ricerche, Via Campi Flegrei 34, 80078 Pozzuoli, Italy
| | - Pietro Ferraro
- Istituto di Scienze Applicate e Sistemi Intelligenti "Eduardo Caianiello", Consiglio Nazionale delle Ricerche, Via Campi Flegrei 34, 80078 Pozzuoli, Italy
| | - Maciej Wnuk
- Department of Genetics, Faculty of Biotechnology, University of Rzeszow, Pigonia 1, 35-310 Rzeszow, Poland
| | - Angela Tino
- Istituto di Scienze Applicate e Sistemi Intelligenti "Eduardo Caianiello", Consiglio Nazionale delle Ricerche, Via Campi Flegrei 34, 80078 Pozzuoli, Italy
| | - Claudia Tortiglione
- Istituto di Scienze Applicate e Sistemi Intelligenti "Eduardo Caianiello", Consiglio Nazionale delle Ricerche, Via Campi Flegrei 34, 80078 Pozzuoli, Italy
| |
Collapse
|
48
|
Heib M, Rose-John S, Adam D. Necroptosis, ADAM proteases and intestinal (dys)function. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2020; 353:83-152. [PMID: 32381179 DOI: 10.1016/bs.ircmb.2020.02.001] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/09/2023]
Abstract
Recently, an unexpected connection between necroptosis and members of the a disintegrin and metalloproteinase (ADAM) protease family has been reported. Necroptosis represents an important cell death routine which helps to protect from viral, bacterial, fungal and parasitic infections, maintains adult T cell homeostasis and contributes to the elimination of potentially defective organisms before parturition. Equally important for organismal homeostasis, ADAM proteases control cellular processes such as development and differentiation, immune responses or tissue regeneration. Notably, necroptosis as well as ADAM proteases have been implicated in the control of inflammatory responses in the intestine. In this review, we therefore provide an overview of the physiology and pathophysiology of necroptosis, ADAM proteases and intestinal (dys)function, discuss the contribution of necroptosis and ADAMs to intestinal (dys)function, and review the current knowledge on the role of ADAMs in necroptotic signaling.
Collapse
Affiliation(s)
- Michelle Heib
- Institut für Immunologie, Christian-Albrechts-Universität zu Kiel, Kiel, Germany
| | - Stefan Rose-John
- Institut für Biochemie, Christian-Albrechts-Universität zu Kiel, Kiel, Germany
| | - Dieter Adam
- Institut für Immunologie, Christian-Albrechts-Universität zu Kiel, Kiel, Germany.
| |
Collapse
|
49
|
Andreone BJ, Larhammar M, Lewcock JW. Cell Death and Neurodegeneration. Cold Spring Harb Perspect Biol 2020; 12:cshperspect.a036434. [PMID: 31451511 DOI: 10.1101/cshperspect.a036434] [Citation(s) in RCA: 59] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Neurodegenerative disease is characterized by the progressive deterioration of neuronal function caused by the degeneration of synapses, axons, and ultimately the death of nerve cells. An increased understanding of the mechanisms underlying altered cellular homeostasis and neurodegeneration is critical to the development of effective treatments for disease. Here, we review what is known about neuronal cell death and how it relates to our understanding of neurodegenerative disease pathology. First, we discuss prominent molecular signaling pathways that drive neuronal loss, and highlight the upstream cell biology underlying their activation. We then address how neuronal death may occur during disease in response to neuron intrinsic and extrinsic stressors. An improved understanding of the molecular mechanisms underlying neuronal dysfunction and cell death will open up avenues for clinical intervention in a field lacking disease-modifying treatments.
Collapse
|
50
|
Colijn S, Muthukumar V, Xie J, Gao S, Griffin CT. Cell-specific and athero-protective roles for RIPK3 in a murine model of atherosclerosis. Dis Model Mech 2020; 13:dmm041962. [PMID: 31953345 PMCID: PMC6994951 DOI: 10.1242/dmm.041962] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2019] [Accepted: 12/18/2019] [Indexed: 12/15/2022] Open
Abstract
Receptor-interacting protein kinase 3 (RIPK3) was recently implicated in promoting atherosclerosis progression through a proposed role in macrophage necroptosis. However, RIPK3 has been connected to numerous other cellular pathways, which raises questions about its actual role in atherosclerosis. Furthermore, RIPK3 is expressed in a multitude of cell types, suggesting that it may be physiologically relevant to more than just macrophages in atherosclerosis. In this study, Ripk3 was deleted in macrophages, endothelial cells, vascular smooth muscle cells or globally on the Apoe-/- background using Cre-lox technology. To induce atherosclerosis progression, male and female mice were fed a Western diet for three months before tissue collection and analysis. Surprisingly, necroptosis markers were nearly undetectable in atherosclerotic aortas. Furthermore, en face lesion area was increased in macrophage- and endothelial-specific deletions of Ripk3 in the descending and abdominal regions of the aorta. Analysis of bone-marrow-derived macrophages and cultured endothelial cells revealed that Ripk3 deletion promotes expression of monocyte chemoattractant protein 1 (MCP-1) and E-selectin in these cell types, respectively. Western blot analysis showed upregulation of MCP-1 in aortas with Ripk3-deficient macrophages. Altogether, these data suggest that RIPK3 in macrophages and endothelial cells protects against atherosclerosis through a mechanism that likely does not involve necroptosis. This protection may be due to RIPK3-mediated suppression of pro-inflammatory MCP-1 expression in macrophages and E-selectin expression in endothelial cells. These findings suggest a novel and unexpected cell-type specific and athero-protective function for RIPK3.This article has an associated First Person interview with the first author of the paper.
Collapse
Affiliation(s)
- Sarah Colijn
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK 73104, USA
- Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73190, USA
| | - Vijay Muthukumar
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK 73104, USA
| | - Jun Xie
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK 73104, USA
| | - Siqi Gao
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK 73104, USA
- Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73190, USA
| | - Courtney T Griffin
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK 73104, USA
- Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73190, USA
| |
Collapse
|