1
|
Xiao J, Wang L, Zhang B, Hou A. Cell death in acute lung injury: caspase-regulated apoptosis, pyroptosis, necroptosis, and PANoptosis. Front Pharmacol 2025; 16:1559659. [PMID: 40191423 PMCID: PMC11968751 DOI: 10.3389/fphar.2025.1559659] [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: 01/13/2025] [Accepted: 03/03/2025] [Indexed: 04/09/2025] Open
Abstract
There has been abundant research on the variety of programmed cell death pathways. Apoptosis, pyroptosis, and necroptosis under the action of the caspase family are essential for the innate immune response. Caspases are classified into inflammatory caspase-1/4/5/11, apoptotic caspase-3/6/7, and caspase-2/8/9/10. Although necroptosis is not caspase-dependent to transmit cell death signals, it can cross-link with pyroptosis and apoptosis signals under the regulation of caspase-8. An increasing number of studies have reiterated the involvement of the caspase family in acute lung injuries caused by bacterial and viral infections, blood transfusion, and ventilation, which is influenced by noxious stimuli that activate or inhibit caspase engagement pathways, leading to subsequent lung injury. This article reviews the role of caspases implicated in diverse programmed cell death mechanisms in acute lung injury and the status of research on relevant inhibitors against essential target proteins of the described cell death mechanisms. The findings of this review may help in delineating novel therapeutic targets for acute lung injury.
Collapse
Affiliation(s)
| | | | | | - Ana Hou
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang, China
| |
Collapse
|
2
|
Huang D, Sun H, Su L, Yang F, Huang D, Gao H, Cao M. Inhibition of SIK1 Alleviates the Pathologies of Psoriasis by Disrupting IL-17 Signaling. Mediators Inflamm 2025; 2025:3540219. [PMID: 39959414 PMCID: PMC11828648 DOI: 10.1155/mi/3540219] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2024] [Accepted: 01/20/2025] [Indexed: 02/18/2025] Open
Abstract
Psoriasis is an inflammatory skin disease mediated by multiple immune cells, including T cells, macrophages, and dendritic cells, which exhibit complex pathologies and limited clinical treatment. Here, we found that salt-inducible kinase 1 (SIK1) was upregulated in the imiquimod (IMQ)-induced psoriasis mouse model. This increment may be due to a higher level of interleukin-17, which promoted the expression of SIK1 in keratinocytes. Inhibition of SIK1 kinase activity using a small molecular inhibitor (HG-9-91-01 or YKL-06-062) dramatically alleviated IMQ-induced psoriasis, showing reduced epidermal thickness, inflammation, and hyperproliferative epidermal keratinocytes. Our data demonstrated that SIK1 inhibitors HG-9-91-01 or YKL-06-062 blocked the expression of IL-17-induced proinflammatory cytokines and chemokines, including Il6, Kc, and Ccl20. Mechanistically, we found that SIK1 inhibitor HG-9-91-01 or YKL-06-062 suppressed the phosphorylation of Iκbα and P38. Consistently, SIK1 overexpression in keratinocytes promoted the activation of Iκbα and P38. Collectively, our results reveal that SIK1 participates to promote IL17-induced signaling through enhancing activation of NF-κB and MAPKs and exacerbates psoriasis-like skin inflammation. Thus, inhibition of SIK1 presents a potential new therapeutic target for psoriasis.
Collapse
Affiliation(s)
- Dongxuan Huang
- Department of Respiratory Medicine, Shenzhen Longhua District Central Hospital, Shenzhen 518110, China
| | - Huimin Sun
- Department of Medical Laboratory, Shenzhen Longhua District Central Hospital, Shenzhen, Guangdong 518110, China
| | - Lianhui Su
- Department of Respiratory Medicine, Shenzhen Longhua District Central Hospital, Shenzhen 518110, China
| | - Fan Yang
- Department of Respiratory Medicine, Shenzhen Longhua District Central Hospital, Shenzhen 518110, China
| | - Dongsheng Huang
- Department of Respiratory Medicine, Shenzhen Longhua District Central Hospital, Shenzhen 518110, China
| | - Hanchao Gao
- Department of Nephrology, Shenzhen Longhua District Central Hospital, Shenzhen Longhua District Key Laboratory for Diagnosis and Treatment of Chronic Kidney Disease, Shenzhen 518110, China
| | - Mengtao Cao
- Department of Respiratory Medicine, Shenzhen Longhua District Central Hospital, Shenzhen 518110, China
- Department of Clinical Laboratory, Shenzhen Second People's Hospital, First Affiliated Hospital of Shenzhen University, Shenzhen, Guangdong 518110, China
| |
Collapse
|
3
|
Tu X, Zou Z, Li J, Zeng S, Luo Z, Li G, Gao Y, Zhang K. Artificial intelligence-enabled discovery of a RIPK3 inhibitor with neuroprotective effects in an acute glaucoma mouse model. Chin Med J (Engl) 2025; 138:172-184. [PMID: 39719694 PMCID: PMC11745860 DOI: 10.1097/cm9.0000000000003387] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2024] [Indexed: 12/26/2024] Open
Abstract
BACKGROUND Retinal ganglion cell (RGC) death caused by acute ocular hypertension is an important characteristic of acute glaucoma. Receptor-interacting protein kinase 3 (RIPK3) that mediates necroptosis is a potential therapeutic target for RGC death. However, the current understanding of the targeting agents and mechanisms of RIPK3 in the treatment of glaucoma remains limited. Notably, artificial intelligence (AI) technologies have significantly advanced drug discovery. This study aimed to discover RIPK3 inhibitor with AI assistance. METHODS An acute ocular hypertension model was used to simulate pathological ocular hypertension in vivo . We employed a series of AI methods, including large language and graph neural network models, to identify the target compounds of RIPK3. Subsequently, these target candidates were validated using molecular simulations (molecular docking, absorption, distribution, metabolism, excretion, and toxicity [ADMET] prediction, and molecular dynamics simulations) and biological experiments (Western blotting and fluorescence staining) in vitro and in vivo . RESULTS AI-driven drug screening techniques have the potential to greatly accelerate drug development. A compound called HG9-91-01, identified using AI methods, exerted neuroprotective effects in acute glaucoma. Our research indicates that all five candidates recommended by AI were able to protect the morphological integrity of RGC cells when exposed to hypoxia and glucose deficiency, and HG9-91-01 showed a higher cell survival rate compared to the other candidates. Furthermore, HG9-91-01 was found to protect the retinal structure and reduce the loss of retinal layers in an acute glaucoma model. It was also observed that the neuroprotective effects of HG9-91-01 were highly correlated with the inhibition of PANoptosis (apoptosis, pyroptosis, and necroptosis). Finally, we found that HG9-91-01 can regulate key proteins related to PANoptosis, indicating that this compound exerts neuroprotective effects in the retina by inhibiting the expression of proteins related to apoptosis, pyroptosis, and necroptosis. CONCLUSION AI-enabled drug discovery revealed that HG9-91-01 could serve as a potential treatment for acute glaucoma.
Collapse
Affiliation(s)
- Xing Tu
- Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, Guangdong 510530, China
- Guangzhou National Laboratory, Guangzhou International Bio Island, Guangzhou, Guangdong 510530, China
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, Guangdong 510623, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Zixing Zou
- Guangzhou National Laboratory, Guangzhou International Bio Island, Guangzhou, Guangdong 510530, China
| | - Jiahui Li
- Guangzhou National Laboratory, Guangzhou International Bio Island, Guangzhou, Guangdong 510530, China
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, Guangdong 510623, China
- Guangzhou Women and Children’s Medical Center, Guangzhou Medical University, Guangzhou, Guangdong 510623, China
| | - Simiao Zeng
- Guangzhou National Laboratory, Guangzhou International Bio Island, Guangzhou, Guangdong 510530, China
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, Guangdong 510623, China
- Guangzhou Women and Children’s Medical Center, Guangzhou Medical University, Guangzhou, Guangdong 510623, China
| | - Zhengchao Luo
- Department of Big Data and Biomedical AI, College of Future Technology, Peking University, Beijing 100871, China
| | - Gen Li
- Guangzhou Women and Children’s Medical Center, Guangzhou Medical University, Guangzhou, Guangdong 510623, China
| | - Yuanxu Gao
- Guangzhou National Laboratory, Guangzhou International Bio Island, Guangzhou, Guangdong 510530, China
- Institute for Artificial Intelligence in Medicine and Faculty of Medicine, Macau University of Science and Technology, Macao Special Administrative Region 999078, China
| | - Kang Zhang
- Guangzhou National Laboratory, Guangzhou International Bio Island, Guangzhou, Guangdong 510530, China
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, Guangdong 510623, China
- Institute for Artificial Intelligence in Medicine and Faculty of Medicine, Macau University of Science and Technology, Macao Special Administrative Region 999078, China
| |
Collapse
|
4
|
He C, Chen P, Ning L, Huang X, Sun H, Wang Y, Zhao Y, Zeng C, Huang D, Gao H, Cao M. Inhibition of Mitochondrial Succinate Dehydrogenase with Dimethyl Malonate Promotes M2 Macrophage Polarization by Enhancing STAT6 Activation. Inflammation 2025:10.1007/s10753-024-02207-y. [PMID: 39806091 DOI: 10.1007/s10753-024-02207-y] [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: 09/14/2024] [Revised: 11/13/2024] [Accepted: 12/03/2024] [Indexed: 01/16/2025]
Abstract
Macrophages exhibit diverse phenotypes depending on environment status, which contribute to physiological and pathological processes of immunological diseases, including sepsis, asthma, multiple sclerosis and colitis. The alternative activation of macrophages is tightly regulated to avoid excessive activation and damage of tissues and organs. Certain works characterized that succinate dehydrogenase (SDH) altered function of macrophages and promoted inflammatory response in M1 macrophages via mitochondrial reactive oxygen species (ROS). However, the effect of succinate dehydrogenase on M2 macrophage polarization remains incompletely understood. We employed dimethyl malonate (DMM) to inhibit succinate dehydrogenase activity and took use of RNA-seq to analyze the changes of inflammatory response of LPS-activated M1 macrophages or IL 4-activated M2 macrophages. Our data revealed that inhibition of SDH with DMM increased expression of M2 macrophages-associated signature genes, including Arg1, Ym1 and Mrc1. Consistent with previous work, we also observed that inhibition of SDH decreased the expression of IL-1β and enhanced the levels of IL-10 in M1 macrophages. Additionally, inhibition of SDH with DMM inhibited the production of chemokines, such as Cxcl3, Cxcl12, Ccl20 and Ccl9. DMM also amplified the M2 macrophages-related signature genes in IL-13-activated M2 macrophages. Mechanistic studies revealed that DMM promoted M2 macrophages polarization through mitochondrial ROS dependent STAT6 activation. Blocking ROS with mitoTEMPO or inhibiting STAT6 activation with ruxolitinib abrogated the promotion effect of DMM on M2 macrophages. Finally, dimethyl malonate treatment promoted peritoneal M2 macrophages differentiation and exacerbated OVA-induced allergy asthma in vivo. Collectively, we identified SDH as a braker to suppress M2 macrophage polarization via mitochondrial ROS, suggesting a novel strategy to treatment of M2 macrophages-mediated inflammatory diseases.
Collapse
Affiliation(s)
- Chaowen He
- Department of Respiratory Medicine, Shenzhen Longhua District Central Hospital, Shenzhen, 518110, China
| | - Pengfei Chen
- Department of Respiratory Medicine, Shenzhen Longhua District Central Hospital, Shenzhen, 518110, China
| | - Luwen Ning
- Health Science Center, Biobank, Shenzhen Second People's Hospital, First Affiliated Hospital of Shenzhen University, Shenzhen University, Shenzhen, China
| | - Xiuping Huang
- Department of Respiratory Medicine, Shenzhen Longhua District Central Hospital, Shenzhen, 518110, China
| | - Huimin Sun
- Department of Respiratory Medicine, Shenzhen Longhua District Central Hospital, Shenzhen, 518110, China
| | - Yuanyuan Wang
- Department of Respiratory Medicine, Shenzhen Longhua District Central Hospital, Shenzhen, 518110, China
| | - Yanli Zhao
- Department of Respiratory Medicine, Shenzhen Longhua District Central Hospital, Shenzhen, 518110, China
| | - Changchun Zeng
- Department of Respiratory Medicine, Shenzhen Longhua District Central Hospital, Shenzhen, 518110, China
| | - Dongsheng Huang
- Department of Respiratory Medicine, Shenzhen Longhua District Central Hospital, Shenzhen, 518110, China.
| | - Hanchao Gao
- Department of Nephrology, Shenzhen Longhua District Key Laboratory for Diagnosis and Treatment of Chronic Kidney Disease, Shenzhen Longhua District Central Hospital, Shenzhen, 518110, China.
| | - Mengtao Cao
- Department of Respiratory Medicine, Shenzhen Longhua District Central Hospital, Shenzhen, 518110, China.
| |
Collapse
|
5
|
Luo M, Wang YM, Zhao FK, Luo Y. Recent Advances in Nanomaterial-Mediated Cell Death for Cancer Therapy. Adv Healthc Mater 2025; 14:e2402697. [PMID: 39498722 DOI: 10.1002/adhm.202402697] [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: 07/22/2024] [Revised: 10/01/2024] [Indexed: 11/07/2024]
Abstract
Nanomedicine has shown great anticancer potential by disrupting redox homeostasis and increasing the levels of oxidative stress, but the therapeutic effect is limited by factors including the intrinsic self-protection mechanism of tumors. Cancer cell death can be induced by the exploration of different cell death mechanisms, such as apoptosis, pyroptosis, necroptosis, cuproptosis, and ferroptosis. The merging of nanotechnology with biomedicine has provided tremendous opportunities to construct cell death-based nanomedicine for innovative cancer therapy. Nanocarriers are not only used for the targeted delivery of cell death inducers, but also as therapeutic components to induce cell death to achieve efficient tumor treatment. This review focuses on seven cell death modalities mediated by nanomaterials, such as apoptosis, pyroptosis, necroptosis, ferroptosis, cuprotosis, immunogenic cell death, and autophagy. The mechanisms of these seven cell death modalities are described in detail, as well as the preparation of nanomaterials that induce them and the mechanisms, they used to exert their effects. Finally, this work describes the potential future development based on the current knowledge related to cell death induced by nanomaterials.
Collapse
Affiliation(s)
- Min Luo
- Department of Clinical Medicine, The Third Affiliated Hospital of Zunyi Medical University, The First People's Hospital of Zunyi, Zunyi, Guizhou, 563000, China
| | - Yuan-Min Wang
- Department of Clinical Medicine, The Third Affiliated Hospital of Zunyi Medical University, The First People's Hospital of Zunyi, Zunyi, Guizhou, 563000, China
| | - Fu-Kun Zhao
- Department of Clinical Medicine, The Third Affiliated Hospital of Zunyi Medical University, The First People's Hospital of Zunyi, Zunyi, Guizhou, 563000, China
| | - Yong Luo
- Department of Neurology, The Third Affiliated Hospital of Zunyi Medical University, The First People's Hospital of Zunyi, Zunyi, Guizhou, 563000, China
| |
Collapse
|
6
|
Cao X, Tan J, Zheng R, Wang F, Zhou L, Yi J, Yuan R, Dai Q, Song L, Dai A. Targeting necroptosis: a promising avenue for respiratory disease treatment. Cell Commun Signal 2024; 22:418. [PMID: 39192326 DOI: 10.1186/s12964-024-01804-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Accepted: 08/22/2024] [Indexed: 08/29/2024] Open
Abstract
Respiratory diseases are a growing concern in public health because of their potential to endanger the global community. Cell death contributes critically to the pathophysiology of respiratory diseases. Recent evidence indicates that necroptosis, a unique form of programmed cell death (PCD), plays a vital role in the molecular mechanisms underlying respiratory diseases, distinguishing it from apoptosis and conventional necrosis. Necroptosis is a type of inflammatory cell death governed by receptor-interacting serine/threonine protein kinase 1 (RIPK1), RIPK3, and mixed-lineage kinase domain-like protein (MLKL), resulting in the release of intracellular contents and inflammatory factors capable of initiating an inflammatory response in adjacent tissues. These necroinflammatory conditions can result in significant organ dysfunction and long-lasting tissue damage within the lungs. Despite evidence linking necroptosis to various respiratory diseases, there are currently no specific alternative treatments that target this mechanism. This review provides a comprehensive overview of the most recent advancements in understanding the significance and mechanisms of necroptosis. Specifically, this review emphasizes the intricate association between necroptosis and respiratory diseases, highlighting the potential use of necroptosis as an innovative therapeutic approach for treating these conditions.
Collapse
Affiliation(s)
- Xianya Cao
- School of Integrated Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, 410208, People's Republic of China
- Hunan Provincial Key Laboratory of Vascular Biology and Translational Medicine, Changsha, Hunan, 410208, People's Republic of China
| | - Junlan Tan
- Hunan Provincial Key Laboratory of Vascular Biology and Translational Medicine, Changsha, Hunan, 410208, People's Republic of China
- Department of Respiratory Medicine, School of Medicine, Changsha, Hunan, 410021, People's Republic of China
- The First Affiliated Hospital of Hunan University of Chinese Medicine, Changsha, Hunan, 410021, People's Republic of China
| | - Runxiu Zheng
- School of Integrated Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, 410208, People's Republic of China
- Hunan Provincial Key Laboratory of Vascular Biology and Translational Medicine, Changsha, Hunan, 410208, People's Republic of China
| | - Feiying Wang
- Hunan Provincial Key Laboratory of Vascular Biology and Translational Medicine, Changsha, Hunan, 410208, People's Republic of China
- Department of Respiratory Medicine, School of Medicine, Changsha, Hunan, 410021, People's Republic of China
| | - Lingling Zhou
- Hunan Provincial Key Laboratory of Vascular Biology and Translational Medicine, Changsha, Hunan, 410208, People's Republic of China
- Department of Respiratory Medicine, School of Medicine, Changsha, Hunan, 410021, People's Republic of China
| | - Jian Yi
- Hunan Provincial Key Laboratory of Vascular Biology and Translational Medicine, Changsha, Hunan, 410208, People's Republic of China
- The First Affiliated Hospital of Hunan University of Chinese Medicine, Changsha, Hunan, 410021, People's Republic of China
| | - Rong Yuan
- Hunan Provincial Key Laboratory of Vascular Biology and Translational Medicine, Changsha, Hunan, 410208, People's Republic of China
- Department of Respiratory Medicine, School of Medicine, Changsha, Hunan, 410021, People's Republic of China
| | - Qin Dai
- Hunan Provincial Key Laboratory of Vascular Biology and Translational Medicine, Changsha, Hunan, 410208, People's Republic of China
- Department of Respiratory Medicine, School of Medicine, Changsha, Hunan, 410021, People's Republic of China
| | - Lan Song
- Hunan Provincial Key Laboratory of Vascular Biology and Translational Medicine, Changsha, Hunan, 410208, People's Republic of China
- Department of Respiratory Medicine, School of Medicine, Changsha, Hunan, 410021, People's Republic of China
| | - Aiguo Dai
- Hunan Provincial Key Laboratory of Vascular Biology and Translational Medicine, Changsha, Hunan, 410208, People's Republic of China.
- Department of Respiratory Medicine, School of Medicine, Changsha, Hunan, 410021, People's Republic of China.
- Department of Respiratory Medicine, The First Affiliated Hospital of Hunan University of Chinese Medicine, Changsha, Hunan, 410021, People's Republic of China.
| |
Collapse
|
7
|
Yu X, Yuan J, Shi L, Dai S, Yue L, Yan M. Necroptosis in bacterial infections. Front Immunol 2024; 15:1394857. [PMID: 38933265 PMCID: PMC11199740 DOI: 10.3389/fimmu.2024.1394857] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2024] [Accepted: 05/29/2024] [Indexed: 06/28/2024] Open
Abstract
Necroptosis, a recently discovered form of cell-programmed death that is distinct from apoptosis, has been confirmed to play a significant role in the pathogenesis of bacterial infections in various animal models. Necroptosis is advantageous to the host, but in some cases, it can be detrimental. To understand the impact of necroptosis on the pathogenesis of bacterial infections, we described the roles and molecular mechanisms of necroptosis caused by different bacterial infections in this review.
Collapse
Affiliation(s)
- Xing Yu
- Department of Pathogen Biology and Immunology, Faculty of Basic Medical Science, Kunming Medical University, Kunming, China
| | - Jin Yuan
- Clinical Laboratory, Puer Hospital of Traditional Chinese Medicine, Puer, China
| | - Linxi Shi
- Department of Pathogen Biology and Immunology, Faculty of Basic Medical Science, Kunming Medical University, Kunming, China
| | - Shuying Dai
- Department of Pathogen Biology and Immunology, Faculty of Basic Medical Science, Kunming Medical University, Kunming, China
| | - Lei Yue
- Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Kunming, China
| | - Min Yan
- Department of Pathogen Biology and Immunology, Faculty of Basic Medical Science, Kunming Medical University, Kunming, China
| |
Collapse
|
8
|
Shi Y, Wu C, Shi J, Gao T, Ma H, Li L, Zhao Y. Protein phosphorylation and kinases: Potential therapeutic targets in necroptosis. Eur J Pharmacol 2024; 970:176508. [PMID: 38493913 DOI: 10.1016/j.ejphar.2024.176508] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2023] [Revised: 03/05/2024] [Accepted: 03/14/2024] [Indexed: 03/19/2024]
Abstract
Necroptosis is a pivotal contributor to the pathogenesis of various human diseases, including those affecting the nervous system, cardiovascular system, pulmonary system, and kidneys. Extensive investigations have elucidated the mechanisms and physiological ramifications of necroptosis. Among these, protein phosphorylation emerges as a paramount regulatory process, facilitating the activation or inhibition of specific proteins through the addition of phosphate groups to their corresponding amino acid residues. Currently, the targeting of kinases has gained recognition as a firmly established and efficacious therapeutic approach for diverse diseases, notably cancer. In this comprehensive review, we elucidate the intricate role of phosphorylation in governing key molecular players in the necroptotic pathway. Moreover, we provide an in-depth analysis of recent advancements in the development of kinase inhibitors aimed at modulating necroptosis. Lastly, we deliberate on the prospects and challenges associated with the utilization of kinase inhibitors to modulate necroptotic processes.
Collapse
Affiliation(s)
- Yihui Shi
- Institute of Drug Discovery Technology, Ningbo University, Ningbo, Zhejiang, 315211, China
| | - Chengkun Wu
- School of Medicine, Nankai University, Tianjin, 300071, China
| | - Jiayi Shi
- Institute of Drug Discovery Technology, Ningbo University, Ningbo, Zhejiang, 315211, China
| | - Taotao Gao
- Institute of Drug Discovery Technology, Ningbo University, Ningbo, Zhejiang, 315211, China
| | - Huabin Ma
- Central Laboratory, The First Affiliated Hospital, Fujian Medical University, Fuzhou, 350005, China.
| | - Long Li
- Institute of Drug Discovery Technology, Ningbo University, Ningbo, Zhejiang, 315211, China.
| | - Yufen Zhao
- Institute of Drug Discovery Technology, Ningbo University, Ningbo, Zhejiang, 315211, China
| |
Collapse
|
9
|
Zhang JB, Zhang QR, Jin Q, Yang J, Lin SZ, Fan JG. Sestrin2 maintains hepatic immune homeostasis and redox balance partially via inhibiting RIPK3-mediated necroptosis in metabolic dysfunction-associated steatohepatitis. Mol Metab 2024; 80:101865. [PMID: 38163459 PMCID: PMC10825057 DOI: 10.1016/j.molmet.2023.101865] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Revised: 12/22/2023] [Accepted: 12/23/2023] [Indexed: 01/03/2024] Open
Abstract
BACKGROUND & AIMS Necroptosis, a novel type of programmed cell death, is intricately associated with inflammatory response. Currently, most studies focus on the activation of necroptosis, while the mechanisms underlying the negative regulation of necroptosis remain poorly understood. METHODS The effects of sestrin2 (SESN2) overexpression or knockdown on the regulation of necroptosis were assessed in the TNFα/Smac-mimetic/Z-VAD-FMK (T/S/Z)-induced necroptosis model and palmitic acid (PA)-induced lipotoxicity model. Western-blot, co-Immunoprecipitation, Glutathione S-transferase pull-down, and confocal assays were employed to explore the regulatory mechanisms including protein-protein interactions and post-translational modification. Furthermore, we used GSK'872, a specific inhibitor of receptor-interacting serine/threonine-protein kinase (RIPK) 3, to evaluate the relationship between SESN2-related alterations and RIPK3-mediated necroptosis in T/S/Z-induced necroptosis model, PA-induced lipotoxicity model, and high-fat high-cholesterol diet (HFHCD)-induced non-alcoholic steatohepatitis model. RESULTS Our findings revealed that SESN2 was upregulated under conditions that induce necroptosis and functioned as a negative regulator of necroptosis. High levels of SESN2 could equipped hepatocytes with the ability to defend against necroptotic inflammation and oxidative stress. Mechanistically, SESN2 interacted with RIPK3 and tuned down necroptosis by inhibiting the phosphorylation of RIPK3, promoting the ubiquitination of RIPK3, and preventing the formation of the RIPK1/RIPK3 necrosome. The depletion of SESN2 resulted in excessive necroptosis, accompanied by increased fat accumulation, inflammation, and oxidative stress in the experimental steatohepatitis model. Blocking necroptosis by GSK'872 reduced the liberation of pro-inflammatory cytokines and reactive oxygen species generation, but not hepatocyte fat deposition, in both PA-treated SESN2 knockout cells and HFHCD-fed SESN2 knockout mice, suggesting that the activation of RIPK3-mediated necroptosis may partially account for the hyperinflammation and excessive oxidative stress induced by SESN2 deficiency. CONCLUSION Our results suggested that SESN2 inhibited RIPK3-mediated necroptosis; this regulation is an important for the immune homeostasis and the redox balance in the liver.
Collapse
Affiliation(s)
- Jian-Bin Zhang
- Department of Gastroenterology, Xin Hua Hospital affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China
| | - Qian-Ren Zhang
- Department of Gastroenterology, Xin Hua Hospital affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China
| | - Qian Jin
- Department of Gastroenterology, Xin Hua Hospital affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China
| | - Jing Yang
- Department of Gastroenterology, Xin Hua Hospital affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China
| | - Shuang-Zhe Lin
- Department of Gastroenterology, Xin Hua Hospital affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China
| | - Jian-Gao Fan
- Department of Gastroenterology, Xin Hua Hospital affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China.
| |
Collapse
|
10
|
Mei X, Zhang Y, Wang S, Wang H, Chen R, Ma K, Yang Y, Jiang P, Feng Z, Zhang C, Zhang Z. Necroptosis in Pneumonia: Therapeutic Strategies and Future Perspectives. Viruses 2024; 16:94. [PMID: 38257794 PMCID: PMC10818625 DOI: 10.3390/v16010094] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Revised: 01/02/2024] [Accepted: 01/04/2024] [Indexed: 01/24/2024] Open
Abstract
Pneumonia remains a major global health challenge, necessitating the development of effective therapeutic approaches. Recently, necroptosis, a regulated form of cell death, has garnered attention in the fields of pharmacology and immunology for its role in the pathogenesis of pneumonia. Characterized by cell death and inflammatory responses, necroptosis is a key mechanism contributing to tissue damage and immune dysregulation in various diseases, including pneumonia. This review comprehensively analyzes the role of necroptosis in pneumonia and explores potential pharmacological interventions targeting this cell death pathway. Moreover, we highlight the intricate interplay between necroptosis and immune responses in pneumonia, revealing a bidirectional relationship between necrotic cell death and inflammatory signaling. Importantly, we assess current therapeutic strategies modulating necroptosis, encompassing synthetic inhibitors, natural products, and other drugs targeting key components of the programmed necrosis pathway. The article also discusses challenges and future directions in targeting programmed necrosis for pneumonia treatment, proposing novel therapeutic strategies that combine antibiotics with necroptosis inhibitors. This review underscores the importance of understanding necroptosis in pneumonia and highlights the potential of pharmacological interventions to mitigate tissue damage and restore immune homeostasis in this devastating respiratory infection.
Collapse
Affiliation(s)
- Xiuzhen Mei
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
- Institute of Veterinary Medicine, Jiangsu Academy of Agricultural Sciences, Nanjing 210014, China
- GuoTai (Taizhou) Center of Technology Innovation for Veterinary Biologicals, Taizhou 225300, China
| | - Yuchen Zhang
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
- Institute of Veterinary Medicine, Jiangsu Academy of Agricultural Sciences, Nanjing 210014, China
- GuoTai (Taizhou) Center of Technology Innovation for Veterinary Biologicals, Taizhou 225300, China
| | - Shu Wang
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
- Institute of Veterinary Medicine, Jiangsu Academy of Agricultural Sciences, Nanjing 210014, China
- GuoTai (Taizhou) Center of Technology Innovation for Veterinary Biologicals, Taizhou 225300, China
| | - Hui Wang
- Institute of Veterinary Medicine, Jiangsu Academy of Agricultural Sciences, Nanjing 210014, China
- GuoTai (Taizhou) Center of Technology Innovation for Veterinary Biologicals, Taizhou 225300, China
- Jiangsu Key Laboratory for Aquatic Crustacean Diseases, College of Marine Science and Engineering, Nanjing Normal University, Nanjing 210023, China
| | - Rong Chen
- Institute of Veterinary Medicine, Jiangsu Academy of Agricultural Sciences, Nanjing 210014, China
- GuoTai (Taizhou) Center of Technology Innovation for Veterinary Biologicals, Taizhou 225300, China
| | - Ke Ma
- Institute of Veterinary Medicine, Jiangsu Academy of Agricultural Sciences, Nanjing 210014, China
- GuoTai (Taizhou) Center of Technology Innovation for Veterinary Biologicals, Taizhou 225300, China
- College of Life Sciences, Nanjing Agricultural University, Nanjing 210095, China
| | - Yue Yang
- Institute of Veterinary Medicine, Jiangsu Academy of Agricultural Sciences, Nanjing 210014, China
- GuoTai (Taizhou) Center of Technology Innovation for Veterinary Biologicals, Taizhou 225300, China
- Jiangsu Key Laboratory for Aquatic Crustacean Diseases, College of Marine Science and Engineering, Nanjing Normal University, Nanjing 210023, China
| | - Ping Jiang
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
| | - Zhixin Feng
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
- Institute of Veterinary Medicine, Jiangsu Academy of Agricultural Sciences, Nanjing 210014, China
- GuoTai (Taizhou) Center of Technology Innovation for Veterinary Biologicals, Taizhou 225300, China
- Jiangsu Key Laboratory for Aquatic Crustacean Diseases, College of Marine Science and Engineering, Nanjing Normal University, Nanjing 210023, China
| | - Chao Zhang
- School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Zhenzhen Zhang
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
- Institute of Veterinary Medicine, Jiangsu Academy of Agricultural Sciences, Nanjing 210014, China
- GuoTai (Taizhou) Center of Technology Innovation for Veterinary Biologicals, Taizhou 225300, China
| |
Collapse
|
11
|
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
|
12
|
Wang X, Wang S, Cui Y. Protective effects of the salt -induced kinase inhibitor HG -9 -91 -01 on sepsis -associated cognitive dysfunction in mice and the underlying mechanisms. ZHONG NAN DA XUE XUE BAO. YI XUE BAN = JOURNAL OF CENTRAL SOUTH UNIVERSITY. MEDICAL SCIENCES 2023; 48:1793-1803. [PMID: 38448372 PMCID: PMC10930753 DOI: 10.11817/j.issn.1672-7347.2023.230208] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Indexed: 03/08/2024]
Abstract
OBJECTIVES Sepsis-associated cognitive dysfunction is a common complication in patients with sepsis and lack of effective treatment. Its pathological mechanisms remain unclear. Salt-induced kinase (SIK) is an important molecule in the regulation of metabolism, immunity, and inflammatory response. It is associated with the development of many neurological diseases. This study aims to investigate the expression of SIK in the hippocampus of septic mice, and to evaluate the role and mechanism of the SIK inhibitor HG-9-91-01 in sepsis-associated cognitive dysfunction. METHODS Firstly, C57BL/6 mice were randomly divided into a control group (Con group) and a sepsis model group [lipopolysaccharide (LPS) group]. The model group was injected intraperitoneally with LPS at a dose of 8 mg/kg and the Con group was injected with an equal volume of normal saline. Hippocampal tissues were harvested at 1, 3, and 6 days after injection and the expressions of SIK1, SIK2, and SIK3 were detected by real-time fluorescence quantitative PCR (qPCR) and Western blotting. Secondly, C57BL/6 mice were randomly divided into a Con group, a LPS group, and a SIK inhibitor group (HG group). The LPS and HG groups were injected with LPS to establish a sepsis model; in the HG group, HG-9-91-01 (10 mg/kg) was injected intraperitoneally at 3-6 days after LPS injection, and the LPS group was injected with the same volume of vehicle. Cognitive function was assessed at 7-11 days after LPS injection using the Morris water maze (MWM). Hippocampal tissues were harvested after the behavioral tests, and the mRNA levels of inflammatory factors and microglial markers were assessed by qPCR. The protein levels of inducible nitric oxide synthase (iNOS), CD68, ionized calcium binding adaptor molecule 1 (Iba-1), N-methyl-D-aspartate (NMDA) receptor (NR) subunit, cAMP response element-binding protein (CREB)-regulated transcription coactivator 1 (CRTC1), and insulin-like growth factor 1 (IGF-1) were detected by Western blotting. Immunohistochemistry (IHC) was used to detect the expression of Iba-1 positive cells in the CA1, CA3 and dentate gyrus (DG) of the hippocampus, followed by Sholl analysis. RESULTS Compared with the Con group, the mRNA and protein levels of SIK1, SIK2, and SIK3 in the hippocampus were increased in the LPS group (all P<0.05). Compared with the Con group, mice in the LPS group had a significantly longer escape latency, a lower percentage of target quadrant dwell time and a reduced locomotor speed (all P<0.05); the HG group had a decreased escape latency and an increased percentage of time spent in the target quadrant in comparison with the LPS group (both P<0.05). The mRNA levels of inflammatory factors [tumor necrosis factor-α (TNF-α), interleukin-1β (IL-1β), interleukin-6 (IL-6)], and the M1-type microglial markers iNOS and CD68 in the hippocampus of the LPS group were increased in comparison with the Con group, while the M2-type microglial markers CD206 and arginase-1 (Arg-1) were decreased. Compared with the LPS group, the mRNA levels of TNF-α, IL-1β, IL-6, and iNOS were downregulated, while the levels of CD206 and Arg-1 were upregulated in the HG group (all P<0.05). The protein levels of iNOS, CD68, and Iba-1 in the hippocampus of the LPS group were increased in comparison with the Con group, but they were downregulated in the HG group in comparison with the LPS group (all P<0.05). The number of Iba-1 positive cells in CA1, CA3, and DG of the hippocampus was increased in the LPS group in comparison with the Con group, but they were decreased in the HG group in comparison with the LPS group (all P<0.05). Sholl analysis showed that the number of intersections at all radii between 8-38 µm from the microglial soma was decreased in the LPS group in comparison with the Con group (all P<0.05). Compared with the LPS group, the number of intersections at all radii between 14-20 µm was significantly increased in the HG group (all P<0.05). The protein levels of NR subunit NR1, NR2A, NR2B, and IGF-1 were downregulated in the hippocampus of the LPS group in comparison with the Con group, while the expression of phosphorylated CRTC1 (p-CRTC1) was increased. Compared with the LPS group, the levels of NR1, NR2A, NR2B, and IGF-1 were upregulated, while p-CRTC1 was downregulated in the HG group (all P<0.05). CONCLUSIONS SIK expression is upregulated in the hippocampus of septic mice. The SIK inhibitor HG-9-91-01 ameliorates sepsis-associated cognitive dysfunction in mice, and the mechanism may involve in the activation of the CRTC1/IGF-1 pathway, inhibition of neuroinflammation, and enhancement of synaptic plasticity.
Collapse
Affiliation(s)
- Xueqin Wang
- Department of Anatomy and Neurobiology, School of Basic Medical Science, Central South University, Changsha 410013.
- Hunan Provincial University Key Laboratory of the Fundamental and Clinical Research on Neurodegenerative Diseases, Changsha Medical University, Changsha 410219.
| | - Shuang Wang
- Department of Clinical Laboratory, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Yanhui Cui
- Department of Anatomy and Neurobiology, School of Basic Medical Science, Central South University, Changsha 410013.
| |
Collapse
|
13
|
Rodriguez Esquivel M, Hayes E, Lakomy O, Hassan M, Foretz M, Stocco C. Salt-inducible kinases regulate androgen synthesis in theca cells by enhancing CREB signaling. Mol Cell Endocrinol 2023; 577:112030. [PMID: 37499999 PMCID: PMC10592241 DOI: 10.1016/j.mce.2023.112030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Revised: 07/18/2023] [Accepted: 07/24/2023] [Indexed: 07/29/2023]
Abstract
Ovulation is the pinnacle of folliculogenesis, a process that requires an interplay between the oocyte, the granulosa cells, and the theca cells (TCs). TCs are the only source of ovarian androgens, which play a vital role in female fertility. However, abnormally elevated androgen levels reduce fertility. Therefore, uncovering novel mechanisms regulating androgen synthesis in TCs is of great significance. We have shown that salt-inducible kinases (SIKs) regulate granulosa cell steroidogenesis. Here, we investigated whether SIKs regulate androgen production in TCs. SIK2 and SIK3 were detected in the TCs of mouse ovaries and isolated TCs. Next, TCs in culture were treated with luteinizing hormone (LH) in the presence or absence of a highly specific SIK inhibitor. SIK inhibition enhanced the stimulatory effect of LH on steroidogenic gene expression and androgen production in a concentration-dependent manner. SIK inhibition alone stimulated the expression of steroidogenic genes and increased androgen production. Activation of adenylyl cyclase with forskolin or emulation of increased intracellular cyclic AMP levels stimulated steroidogenesis, an effect that was enhanced by the inhibition of SIK activity. The stimulatory effect of downstream targets of cyclic AMP was also significantly augmented by SIK inhibition, suggesting that SIKs control targets downstream cyclic AMP. Finally, it is shown that SIK2 knockout mice have higher circulating testosterone than controls. This evidence shows that TCs express SIKs and reveal novel roles for SIKs in the regulation of TC function and androgen production. This information could contribute to uncovering therapeutic targets to treat hyperandrogenic diseases.
Collapse
Affiliation(s)
| | - Emily Hayes
- Department of Physiology and Biophysics, University of Illinois at Chicago, Chicago, IL, 60612, USA
| | - Oliwia Lakomy
- Department of Physiology and Biophysics, University of Illinois at Chicago, Chicago, IL, 60612, USA
| | - Mariam Hassan
- Department of Physiology and Biophysics, University of Illinois at Chicago, Chicago, IL, 60612, USA
| | - Marc Foretz
- Université Paris Cité, Institut Cochin, CNRS, INSERM, F-75014, Paris, France
| | - Carlos Stocco
- Department of Physiology and Biophysics, University of Illinois at Chicago, Chicago, IL, 60612, USA.
| |
Collapse
|
14
|
Shadab A, Mahjoor M, Abbasi-Kolli M, Afkhami H, Moeinian P, Safdarian AR. Divergent functions of NLRP3 inflammasomes in cancer: a review. Cell Commun Signal 2023; 21:232. [PMID: 37715239 PMCID: PMC10503066 DOI: 10.1186/s12964-023-01235-9] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Accepted: 07/17/2023] [Indexed: 09/17/2023] Open
Abstract
The cancer is a serious health problem, which is The cancer death rate (cancer mortality) is 158.3 per 100,000 men and women per year (based on 2013-2017 deaths). Both clinical and translational studies have demonstrated that chronic inflammation is associated with Cancer progression. However, the precise mechanisms of inflammasome, and the pathways that mediate this phenomenon are not fully characterized. One of the most recently identified signaling pathways, whose activation seems to affect many metabolic disorders, is the "inflammasome" a multiprotein complex composed of NLRP3 (nucleotide-binding domain and leucine-rich repeat protein 3), ASC (apoptosis associated speck-like protein containing a CARD), and procaspase-1. NLRP3 inflammasome activation leads to the processing and secretion of the proinflammatory cytokines interleukin-1β (IL-1β) and IL-18. The goal of this paper is to review new insights on the effects of the NLRP3 inflammasome activation in the complex mechanisms of crosstalk between different organs, for a better understanding of the role of chronic inflammation in cancer pathogenesis. We will provide here a perspective on the current research on NLRP3 inflammasome, which may represent an innovative therapeutic target to reverse the malignancy condition consequences of the inflammation. Video Abstract.
Collapse
Affiliation(s)
- Alireza Shadab
- Department of Immunology, School of Medicine, Semnan University of Medical Sciences, Semnan, Iran
- Iran University of Medical Sciences, Deputy of Health, Tehran, Iran
| | - Mohamad Mahjoor
- Cellular and Molecular Research Center, Qom University of Medical Sciences, Qom, Iran
- Department of Immunology, Faculty of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Mohammad Abbasi-Kolli
- Iran University of Medical Sciences, Deputy of Health, Tehran, Iran
- Department of Medical Genetics, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Hamed Afkhami
- Cellular and Molecular Research Center, Qom University of Medical Sciences, Qom, Iran
- Nervous System Stem Cells Research Center, Semnan University of Medical Sciences, Semnan, Iran
- Department of Medical Microbiology, Faculty of Medicine, Shahed University, Tehran, Iran
| | - Parisa Moeinian
- Department of Medical Genetics and Molecular Biology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Amir-Reza Safdarian
- Immunology Board for Transplantation and Cell-Based Therapeutics (Immuno TACT), Universal Scientific Education and Research Network (USERN) Chicago, Chicago, IL, USA.
- Department of Immunology and Microbiology, Faculty of Veterinary Medicine, Tehran University, Tehran, Iran.
| |
Collapse
|
15
|
Huang J, Fan H, Chen YM, Wang CN, Guan W, Li WY, Shi TS, Chen WJ, Zhu BL, Liu JF, Jiang B. The salt-inducible kinases inhibitor HG-9-91-01 exhibits antidepressant-like actions in mice exposed to chronic unpredictable mild stress. Neuropharmacology 2023; 227:109437. [PMID: 36702294 DOI: 10.1016/j.neuropharm.2023.109437] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Revised: 01/16/2023] [Accepted: 01/21/2023] [Indexed: 01/24/2023]
Abstract
Major depressive disorder is a frequently occurring neuropsychiatric disorder throughout the world. However, the limited and delayed therapeutic efficacy of monoaminergic medications has led to intensive research efforts to develop novel antidepressants. We have previously demonstrated that hippocampal salt-inducible kinase 2 (SIK2) plays a role in the pathogenesis of depression via regulating the downstream CREB-regulated transcription coactivator 1 (CRTC1)-cAMP response element-binding protein (CREB)-brain derived neurotrophic factor (BDNF) pathway. HG-9-91-01 is a potent and selective inhibitor of salt-inducible kinases (SIKs). The present study aims to explore whether HG-9-91-01 has antidepressant-like actions in male C57BL/6J mice. The chronic unpredictable mild stress (CUMS) model of depression, various behavioral tests, western blotting, co-immunoprecipitation, immunofluorescence, stereotactic infusion, and viral-mediated genetic knockdown were used together. It was found that hippocampal infusion of HG-9-91-01 induced significant antidepressant-like effects in the CUMS model, accompanied with preventing the enhancement of CUMS on the hippocampal SIK2 expression and cytoplasmic translocation of CRTC1. HG-9-91-01 treatment also reversed the decreasing effects of CUMS on the BDNF signaling cascade and adult neurogenesis in the hippocampus. Moreover, the antidepressant-like actions of HG-9-91-01 in mice required the hippocampal CRTC1-CREB-BDNF pathway. In conclusion, HG-9-91-01 has potential of being a novel antidepressant candidate.
Collapse
Affiliation(s)
- Jie Huang
- Department of Pharmacology, School of Pharmacy, Nantong University, Nantong, 226001, Jiangsu, China
| | - Hua Fan
- The First Affiliated Hospital, College of Clinical Medicine of Henan University of Science and Technology, Luoyang, 471000, Henan, China
| | - Yan-Mei Chen
- Department of Pharmacology, School of Pharmacy, Nantong University, Nantong, 226001, Jiangsu, China
| | - Cheng-Niu Wang
- Basic Medical Research Centre, Medical College, Nantong University, Nantong, 226001, Jiangsu, China
| | - Wei Guan
- Department of Pharmacology, School of Pharmacy, Nantong University, Nantong, 226001, Jiangsu, China
| | - Wei-Yu Li
- Department of Pharmacology, School of Pharmacy, Nantong University, Nantong, 226001, Jiangsu, China
| | - Tian-Shun Shi
- Department of Pharmacology, School of Pharmacy, Nantong University, Nantong, 226001, Jiangsu, China
| | - Wei-Jia Chen
- Department of Pharmacology, School of Pharmacy, Nantong University, Nantong, 226001, Jiangsu, China
| | - Bao-Lun Zhu
- Department of Pharmacology, School of Pharmacy, Nantong University, Nantong, 226001, Jiangsu, China
| | - Jian-Feng Liu
- Department of Pharmacology, School of Pharmacy, Nantong University, Nantong, 226001, Jiangsu, China.
| | - Bo Jiang
- Department of Pharmacology, School of Pharmacy, Nantong University, Nantong, 226001, Jiangsu, China.
| |
Collapse
|
16
|
Shi FL, Yuan LS, Wong TS, Li Q, Li YP, Xu R, You YP, Yuan T, Zhang HR, Shi ZJ, Zha QB, Hu B, He XH, Ouyang DY. Dimethyl fumarate inhibits necroptosis and alleviates systemic inflammatory response syndrome by blocking the RIPK1-RIPK3-MLKL axis. Pharmacol Res 2023; 189:106697. [PMID: 36796462 DOI: 10.1016/j.phrs.2023.106697] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Revised: 02/10/2023] [Accepted: 02/13/2023] [Indexed: 02/16/2023]
Abstract
Necroptosis has been implicated in various inflammatory diseases including tumor-necrosis factor-α (TNF-α)-induced systemic inflammatory response syndrome (SIRS). Dimethyl fumarate (DMF), a first-line drug for treating relapsing-remitting multiple sclerosis (RRMS), has been shown to be effective against various inflammatory diseases. However, it is still unclear whether DMF can inhibit necroptosis and confer protection against SIRS. In this study, we found that DMF significantly inhibited necroptotic cell death in macrophages induced by different necroptotic stimulations. Both the autophosphorylation of receptor-interacting serine/threonine kinase 1 (RIPK1) and RIPK3 and the downstream phosphorylation and oligomerization of MLKL were robustly suppressed by DMF. Accompanying the suppression of necroptotic signaling, DMF blocked the mitochondrial reverse electron transport (RET) induced by necroptotic stimulation, which was associated with its electrophilic property. Several well-known anti-RET reagents also markedly inhibited the activation of the RIPK1-RIPK3-MLKL axis accompanied by decreased necrotic cell death, indicating a critical role of RET in necroptotic signaling. DMF and other anti-RET reagents suppressed the ubiquitination of RIPK1 and RIPK3, and they attenuated the formation of necrosome. Moreover, oral administration of DMF significantly alleviated the severity of TNF-α-induced SIRS in mice. Consistent with this, DMF mitigated TNF-α-induced cecal, uterine, and lung damage accompanied by diminished RIPK3-MLKL signaling. Collectively, DMF represents a new necroptosis inhibitor that suppresses the RIPK1-RIPK3-MLKL axis through blocking mitochondrial RET. Our study highlights DMF's potential therapeutic applications for treating SIRS-associated diseases.
Collapse
Affiliation(s)
- Fu-Li Shi
- Department of Immunobiology, College of Life Science and Technology, Jinan University, Guangzhou 510632, China
| | - Li-Sha Yuan
- Department of Immunobiology, College of Life Science and Technology, Jinan University, Guangzhou 510632, China
| | - Tak-Sui Wong
- Department of Nephrology, the First Affiliated Hospital of Jinan University, Guangzhou 510630, China
| | - Qing Li
- Department of Immunobiology, College of Life Science and Technology, Jinan University, Guangzhou 510632, China
| | - Ya-Ping Li
- Department of Immunobiology, College of Life Science and Technology, Jinan University, Guangzhou 510632, China
| | - Rong Xu
- Department of Immunobiology, College of Life Science and Technology, Jinan University, Guangzhou 510632, China
| | - Yi-Ping You
- Department of Immunobiology, College of Life Science and Technology, Jinan University, Guangzhou 510632, China
| | - Tao Yuan
- Department of Immunobiology, College of Life Science and Technology, Jinan University, Guangzhou 510632, China
| | - Hong-Rui Zhang
- Department of Immunobiology, College of Life Science and Technology, Jinan University, Guangzhou 510632, China
| | - Zi-Jian Shi
- Department of Fetal Medicine, the First Affiliated Hospital of Jinan University, Guangzhou 510630, China
| | - Qing-Bing Zha
- Department of Fetal Medicine, the First Affiliated Hospital of Jinan University, Guangzhou 510630, China; Department of Clinical Laboratory, the Fifth Affiliated Hospital of Jinan University, Heyuan 517000, China
| | - Bo Hu
- Department of Nephrology, the First Affiliated Hospital of Jinan University, Guangzhou 510630, China.
| | - Xian-Hui He
- Department of Immunobiology, College of Life Science and Technology, Jinan University, Guangzhou 510632, China; Department of Clinical Laboratory, the Fifth Affiliated Hospital of Jinan University, Heyuan 517000, China.
| | - Dong-Yun Ouyang
- Department of Immunobiology, College of Life Science and Technology, Jinan University, Guangzhou 510632, China.
| |
Collapse
|
17
|
Zhang Q, Hu XM, Zhao WJ, Ban XX, Li Y, Huang YX, Wan H, He Y, Liao LS, Shang L, Jiang B, Qing GP, Xiong K. Targeting Necroptosis: A Novel Therapeutic Option for Retinal Degenerative Diseases. Int J Biol Sci 2023; 19:658-674. [PMID: 36632450 PMCID: PMC9830514 DOI: 10.7150/ijbs.77994] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2022] [Accepted: 12/15/2022] [Indexed: 01/04/2023] Open
Abstract
The discovery of the necroptosis, a form of regulated necrosis that is mediated by receptor-interacting protein kinase 1 (RIPK1), RIPK3, and mixed-lineage kinase domain-like pseudokinase (MLKL), represents a major breakthrough that has dramatically altered the conception of necrosis - traditionally thought of as uncontrolled cell death - in various human diseases. Retinal cell death is a leading cause of blindness and has been identified in most retinal diseases, e.g., age-related macular degeneration, glaucoma, retinal detachment, retinitis pigmentosa, etc. Increasing evidence demonstrates that retinal degenerative diseases also share a common mechanism in necroptosis. Exacerbated necroptotic cell death hinders the treatment for retinal degenerative diseases. In this review, we highlight recent advances in identifying retinal necroptosis, summarize the underlying mechanisms of necroptosis in retinal degenerative diseases, and discuss potential anti-necroptosis strategies, such as selective inhibitors and chemical agents, for treating retinal degenerative diseases.
Collapse
Affiliation(s)
- Qi Zhang
- Department of Anatomy and Neurobiology, School of Basic Medical Sciences, Central South University, Changsha, China.,Key Laboratory of Emergency and Trauma, Ministry of Education, College of Emergency and Trauma, Hainan Medical University, Haikou, China
| | - Xi-min Hu
- Department of Anatomy and Neurobiology, School of Basic Medical Sciences, Central South University, Changsha, China
| | - Wen-juan Zhao
- Department of Anatomy and Neurobiology, School of Basic Medical Sciences, Central South University, Changsha, China
| | - Xiao-xia Ban
- Department of Anatomy and Neurobiology, School of Basic Medical Sciences, Central South University, Changsha, China
| | - Yan Li
- Department of Anatomy and Neurobiology, School of Basic Medical Sciences, Central South University, Changsha, China
| | - Yan-xia Huang
- Department of Anatomy and Neurobiology, School of Basic Medical Sciences, Central South University, Changsha, China
| | - Hao Wan
- Department of Anatomy and Neurobiology, School of Basic Medical Sciences, Central South University, Changsha, China
| | - Ye He
- Changsha Aier Eye Hospital, Changsha, China
| | - Lv-shuang Liao
- School of Physical Education, Hunan Institute of Science and Technology, Yueyang, China
| | - Lei Shang
- Affiliated Eye Hospital of Nanchang University, Jiangxi Research Institute of Ophthalmology and Visual Science, Jiangxi Clinical Research Center for Ophthalmic Disease, Nanchang, China
| | - Bin Jiang
- Department of Ophthalmology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Guo-ping Qing
- Beijing Tongren Eye Center, Beijing Tongren Hospital, Capital Medical University, Beijing, China.,Beijing Ophthalmology and Visual Sciences Key Laboratory, Beijing, China
| | - Kun Xiong
- Department of Anatomy and Neurobiology, School of Basic Medical Sciences, Central South University, Changsha, China.,Key Laboratory of Emergency and Trauma, Ministry of Education, College of Emergency and Trauma, Hainan Medical University, Haikou, China.,Hunan Key Laboratory of Ophthalmology, Changsha, China.,✉ Corresponding author: E-mail:
| |
Collapse
|
18
|
Goode RA, Hum JM, Kalwat MA. Therapeutic Strategies Targeting Pancreatic Islet β-Cell Proliferation, Regeneration, and Replacement. Endocrinology 2022; 164:6836713. [PMID: 36412119 PMCID: PMC9923807 DOI: 10.1210/endocr/bqac193] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Revised: 11/16/2022] [Accepted: 11/18/2022] [Indexed: 11/23/2022]
Abstract
Diabetes results from insufficient insulin production by pancreatic islet β-cells or a loss of β-cells themselves. Restoration of regulated insulin production is a predominant goal of translational diabetes research. Here, we provide a brief overview of recent advances in the fields of β-cell proliferation, regeneration, and replacement. The discovery of therapeutic targets and associated small molecules has been enabled by improved understanding of β-cell development and cell cycle regulation, as well as advanced high-throughput screening methodologies. Important findings in β-cell transdifferentiation, neogenesis, and stem cell differentiation have nucleated multiple promising therapeutic strategies. In particular, clinical trials are underway using in vitro-generated β-like cells from human pluripotent stem cells. Significant challenges remain for each of these strategies, but continued support for efforts in these research areas will be critical for the generation of distinct diabetes therapies.
Collapse
Affiliation(s)
- Roy A Goode
- Division of Biomedical Sciences, College of Osteopathic Medicine, Marian University, Indianapolis, IN, USA
| | - Julia M Hum
- Division of Biomedical Sciences, College of Osteopathic Medicine, Marian University, Indianapolis, IN, USA
| | - Michael A Kalwat
- Correspondence: Michael A. Kalwat, PhD, Lilly Diabetes Center of Excellence, Indiana Biosciences Research Institute, 1210 Waterway Blvd, Suite 2000, Indianapolis, IN 46202, USA. or
| |
Collapse
|
19
|
Shiau JP, Chuang YT, Tang JY, Yang KH, Chang FR, Hou MF, Yen CY, Chang HW. The Impact of Oxidative Stress and AKT Pathway on Cancer Cell Functions and Its Application to Natural Products. Antioxidants (Basel) 2022; 11:1845. [PMID: 36139919 PMCID: PMC9495789 DOI: 10.3390/antiox11091845] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Revised: 09/14/2022] [Accepted: 09/16/2022] [Indexed: 01/10/2023] Open
Abstract
Oxidative stress and AKT serine-threonine kinase (AKT) are responsible for regulating several cell functions of cancer cells. Several natural products modulate both oxidative stress and AKT for anticancer effects. However, the impact of natural product-modulating oxidative stress and AKT on cell functions lacks systemic understanding. Notably, the contribution of regulating cell functions by AKT downstream effectors is not yet well integrated. This review explores the role of oxidative stress and AKT pathway (AKT/AKT effectors) on ten cell functions, including apoptosis, autophagy, endoplasmic reticulum stress, mitochondrial morphogenesis, ferroptosis, necroptosis, DNA damage response, senescence, migration, and cell-cycle progression. The impact of oxidative stress and AKT are connected to these cell functions through cell function mediators. Moreover, the AKT effectors related to cell functions are integrated. Based on this rationale, natural products with the modulating abilities for oxidative stress and AKT pathway exhibit the potential to regulate these cell functions, but some were rarely reported, particularly for AKT effectors. This review sheds light on understanding the roles of oxidative stress and AKT pathway in regulating cell functions, providing future directions for natural products in cancer treatment.
Collapse
Affiliation(s)
- Jun-Ping Shiau
- Division of Breast Oncology and Surgery, Department of Surgery, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 80708, Taiwan or
| | - Ya-Ting Chuang
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
| | - Jen-Yang Tang
- School of Post-Baccalaureate Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
- Department of Radiation Oncology, Kaohsiung Medical University Hospital, Kaoshiung Medical University, Kaohsiung 80708, Taiwan
| | - Kun-Han Yang
- Graduate Institute of Natural Products, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
| | - Fang-Rong Chang
- Graduate Institute of Natural Products, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
| | - Ming-Feng Hou
- Division of Breast Oncology and Surgery, Department of Surgery, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 80708, Taiwan or
- Department of Biomedical Science and Environmental Biology, College of Life Science, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
| | - Ching-Yu Yen
- Department of Oral and Maxillofacial Surgery, Chi-Mei Medical Center, Tainan 71004, Taiwan
- School of Dentistry, Taipei Medical University, Taipei 11031, Taiwan
| | - Hsueh-Wei Chang
- Department of Biomedical Science and Environmental Biology, College of Life Science, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
- Center for Cancer Research, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
| |
Collapse
|