1
|
Wu X, Yang Z, Zou J, Gao H, Shao Z, Li C, Lei P. Protein kinases in neurodegenerative diseases: current understandings and implications for drug discovery. Signal Transduct Target Ther 2025; 10:146. [PMID: 40328798 PMCID: PMC12056177 DOI: 10.1038/s41392-025-02179-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2024] [Revised: 01/03/2025] [Accepted: 02/12/2025] [Indexed: 05/08/2025] Open
Abstract
Neurodegenerative diseases (e.g., Alzheimer's, Parkinson's, Huntington's disease, and Amyotrophic Lateral Sclerosis) are major health threats for the aging population and their prevalences continue to rise with the increasing of life expectancy. Although progress has been made, there is still a lack of effective cures to date, and an in-depth understanding of the molecular and cellular mechanisms of these neurodegenerative diseases is imperative for drug development. Protein phosphorylation, regulated by protein kinases and protein phosphatases, participates in most cellular events, whereas aberrant phosphorylation manifests as a main cause of diseases. As evidenced by pharmacological and pathological studies, protein kinases are proven to be promising therapeutic targets for various diseases, such as cancers, central nervous system disorders, and cardiovascular diseases. The mechanisms of protein phosphatases in pathophysiology have been extensively reviewed, but a systematic summary of the role of protein kinases in the nervous system is lacking. Here, we focus on the involvement of protein kinases in neurodegenerative diseases, by summarizing the current knowledge on the major kinases and related regulatory signal transduction pathways implicated in diseases. We further discuss the role and complexity of kinase-kinase networks in the pathogenesis of neurodegenerative diseases, illustrate the advances of clinical applications of protein kinase inhibitors or novel kinase-targeted therapeutic strategies (such as antisense oligonucleotides and gene therapy) for effective prevention and early intervention.
Collapse
Affiliation(s)
- Xiaolei Wu
- Department of Neurology and State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Zhangzhong Yang
- Department of Neurology and State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Jinjun Zou
- Department of Neurology and State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Huile Gao
- Key Laboratory of Drug Targeting and Drug Delivery Systems, West China School of Pharmacy, Sichuan University, Chengdu, China
| | - Zhenhua Shao
- Division of Nephrology and Kidney Research Institute, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Chuanzhou Li
- Department of Medical Genetics, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Peng Lei
- Department of Neurology and State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan, China.
| |
Collapse
|
2
|
Lu X, Chen D, Wang M, Song X, Ermine K, Hao S, Jha A, Huang Y, Kang Y, Qiu H, Lenz HJ, Li S, Jin Z, Yu J, Zhang L. Depletion of oxysterol-binding proteins by OSW-1 triggers RIP1/RIP3-independent necroptosis and sensitization to cancer immunotherapy. Cell Death Differ 2025:10.1038/s41418-025-01521-8. [PMID: 40329104 DOI: 10.1038/s41418-025-01521-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2024] [Revised: 04/18/2025] [Accepted: 04/30/2025] [Indexed: 05/08/2025] Open
Abstract
Oxysterol-binding proteins (OSBPs), lipid transfer proteins functioning at intracellular membrane contact sites, are recently found to be dysregulated in cancer and promote cancer cell survival. However, their role as potential targets in cancer therapy remains largely unexplored. In this study, we found OSW-1, a natural compound and OSBP inhibitor, potently and selectively kills colon cancer cells by activating a previously unknown necroptosis pathway that is independent of receptor-interacting protein 1 (RIP1) and RIP3. OSW-1 stabilizes p53 and degrades OSBPs to promote endoplasmic reticulum (ER) stress and glycogen synthase kinase 3β (GSK3β)/Tip60-mediated p53 acetylation at Lysine 120, which selectively induces its target PUMA. PUMA-mediated mitochondrial calcium influx activates calcium/calmodulin-dependent protein kinase IIδ (CamKIIδ) to promote mixed lineage kinase domain-like (MLKL) phosphorylation and necroptotic cell death. Furthermore, OSW-1-induced necroptosis is highly immunogenic and sensitizes syngeneic colorectal tumors to anti-PD-1 immunotherapy. Together, our results identified a novel RIP1/RIP3-independent necroptosis pathway underlying the extremely potent anticancer activity of OSW-1, which can be harnessed to develop new anticancer therapies by selectively stimulating antitumor immunity.
Collapse
Affiliation(s)
- Xinyan Lu
- Department of Medicine, Keck School of Medicine of University of Southern California (USC), Los Angeles, CA, USA
- Norris Comprehensive Cancer Center, Keck School of Medicine of USC, Los Angeles, CA, USA
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- UPMC Hillman Cancer Center, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Dongshi Chen
- Department of Medicine, Keck School of Medicine of University of Southern California (USC), Los Angeles, CA, USA
- Norris Comprehensive Cancer Center, Keck School of Medicine of USC, Los Angeles, CA, USA
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- UPMC Hillman Cancer Center, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Min Wang
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- UPMC Hillman Cancer Center, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Xiangping Song
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- UPMC Hillman Cancer Center, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Kaylee Ermine
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- UPMC Hillman Cancer Center, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Suisui Hao
- Department of Medicine, Keck School of Medicine of University of Southern California (USC), Los Angeles, CA, USA
- Norris Comprehensive Cancer Center, Keck School of Medicine of USC, Los Angeles, CA, USA
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- UPMC Hillman Cancer Center, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Anupma Jha
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- UPMC Hillman Cancer Center, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Yixian Huang
- UPMC Hillman Cancer Center, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Department of Pharmaceutical Sciences, Center for Pharmacogenetics, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA, USA
| | - Ying Kang
- Department of Pharmaceutical Sciences and Experimental Therapeutics, College of Pharmacy, University of Iowa, Iowa City, IA, USA
| | - Haibo Qiu
- Department of Pharmaceutical Sciences and Experimental Therapeutics, College of Pharmacy, University of Iowa, Iowa City, IA, USA
| | - Heinz-Josef Lenz
- Department of Medicine, Keck School of Medicine of University of Southern California (USC), Los Angeles, CA, USA
- Norris Comprehensive Cancer Center, Keck School of Medicine of USC, Los Angeles, CA, USA
| | - Song Li
- UPMC Hillman Cancer Center, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Department of Pharmaceutical Sciences, Center for Pharmacogenetics, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA, USA
| | - Zhendong Jin
- Department of Pharmaceutical Sciences and Experimental Therapeutics, College of Pharmacy, University of Iowa, Iowa City, IA, USA
| | - Jian Yu
- Department of Medicine, Keck School of Medicine of University of Southern California (USC), Los Angeles, CA, USA
- Norris Comprehensive Cancer Center, Keck School of Medicine of USC, Los Angeles, CA, USA
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- UPMC Hillman Cancer Center, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Lin Zhang
- Department of Medicine, Keck School of Medicine of University of Southern California (USC), Los Angeles, CA, USA.
- Norris Comprehensive Cancer Center, Keck School of Medicine of USC, Los Angeles, CA, USA.
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.
- UPMC Hillman Cancer Center, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.
| |
Collapse
|
3
|
Kuo YC, Lin CW, Tai CK. Etoposide-loaded lipopolymer nanoparticles promote Smac minetic activity against inhibitor of apoptosis protein for glioblastoma treatment. BIOMATERIALS ADVANCES 2025; 170:214185. [PMID: 39879864 DOI: 10.1016/j.bioadv.2025.214185] [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: 11/25/2024] [Revised: 12/25/2024] [Accepted: 01/10/2025] [Indexed: 01/31/2025]
Abstract
Encapsulated BV6 and SM164, two bivalent second mitochondria-derived activator of caspase (Smac) mimetics, in etoposide (ETO)-lipopolymer nanoparticles (NPs) have been developed to deplete inhibitor of apoptosis proteins (IAP), impair DNA, and produce antagonistic effects on glioblastoma multiforme (GBM) in nude mice. The NPs, composed of cocoa butter (CB) and polyvinyl alcohol (PVA), were stabilized by glycerol monostearate and Pluronic F-127, and grafted with transferrin (Tf) and wheat germ agglutinin (WGA) to dock the blood-brain barrier (BBB) and degenerated dopaminergic neurons. The dual-targeting NPs increased the BBB permeability of BV6, SM164 and ETO via recognizing Tf receptor (TfR) and N-acetylglucosamine that are abundantly expressed on brain microvascular endothelial cells. The sustained release of BV6, SM164 and ETO from CB-PVA-NPs for 48 h resulted in a reduction of about 40 % in the viability of U87MG cells and human brain cancer stem cells. Hematoxylin and eosin staining of the brain in GBM mice revealed atypical mitosis of cancer cells and a considerable decrease in tumor cell density after treatment with Tf-WGA-BV6-SM164-ETO-NPs. Compared to untreated mice, the current ETO preparation carrying Smac mimetics reduced cellular IAP-1 expression to about 33 % and X-linked IAP expression to about 42 %, while enhanced about 3.8-fold caspase-3, indicating the effectiveness of the nanocarriers in accelerating apoptosis of GBM cells. Tf-WGA-CB-PVA-NPs can be promising to upgrade BV6 and SM164 activity by ETO in clinical trials for GBM management.
Collapse
Affiliation(s)
- Yung-Chih Kuo
- Department of Chemical Engineering, National Chung Cheng University, Chia-Yi 62102, Taiwan, ROC; Advanced Institute of Manufacturing with High-tech Innovations, National Chung Cheng University, Chia-Yi 62102, Taiwan, ROC.
| | - Chia-Wei Lin
- Department of Chemical Engineering, National Chung Cheng University, Chia-Yi 62102, Taiwan, ROC
| | - Chien-Kuo Tai
- Department of Biomedical Sciences, National Chung Cheng University, Chia-Yi 62102, Taiwan, ROC
| |
Collapse
|
4
|
Zhang Z, Yang Z, Wang S, Wang X, Mao J. Natural products and ferroptosis: A novel approach for heart failure management. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 142:156783. [PMID: 40286752 DOI: 10.1016/j.phymed.2025.156783] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/13/2025] [Revised: 03/23/2025] [Accepted: 04/17/2025] [Indexed: 04/29/2025]
Abstract
BACKGROUND The discovery of ferroptosis has brought a revolutionary breakthrough in heart failure treatment, and natural products, as a significant source of drug discovery, are gradually demonstrating their extraordinary potential in regulating ferroptosis and alleviating heart failure symptoms. In addition to chemically synthesized small molecule compounds, natural products have attracted attention as an important source for discovering compounds that target ferroptosis in treating heart failure. PURPOSE Systematically summarize and analyze the research progress on improving heart failure through natural products' modulation of the ferroptosis pathway. METHODS By comprehensively searching authoritative databases like PubMed, Web of Science, and China National Knowledge Infrastructure with keywords such as "heart failure", "cardiovascular disease", "heart disease", "ferroptosis", "natural products", "active compounds", "traditional Chinese medicine formulas", "traditional Chinese medicine", and "acupuncture", we aim to systematically review the mechanism of ferroptosis and its link with heart failure. We also want to explore natural small-molecule compounds, traditional Chinese medicine formulas, and acupuncture therapies that can inhibit ferroptosis to improve heart failure. RESULTS In this review, we not only trace the evolution of the concept of ferroptosis and clearly distinguish it from other forms of cell death but also establish a comprehensive theoretical framework encompassing core mechanisms such as iron overload and system xc-/GSH/GPX4 imbalance, along with multiple auxiliary pathways. On this basis, we innovatively link ferroptosis with various types of heart failure, covering classic heart failure types and extending our research to pre-heart failure conditions such as arrhythmia and aortic aneurysm, providing new insights for early intervention in heart failure. Importantly, this article systematically integrates multiple strategies of natural products for interfering with ferroptosis, ranging from monomeric compounds and bioactive components to crude extracts and further to traditional Chinese medicine formulae. In addition, non-pharmacological means such as acupuncture are also included. CONCLUSION This study fills the gap in the systematic description of the relationship between ferroptosis and heart failure and the therapeutic strategies of natural products, aiming to provide patients with more diverse treatment options and promote the development of the heart failure treatment field.
Collapse
Affiliation(s)
- Zeyu Zhang
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, No.88 Changling Road, Xiqing District, Tianjin 300381, PR China; Tianjin University of Traditional Chinese Medicine, Tianjin 301617, PR China
| | - Zhihua Yang
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, No.88 Changling Road, Xiqing District, Tianjin 300381, PR China; Tianjin University of Traditional Chinese Medicine, Tianjin 301617, PR China
| | - Shuai Wang
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, No.88 Changling Road, Xiqing District, Tianjin 300381, PR China
| | - Xianliang Wang
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, No.88 Changling Road, Xiqing District, Tianjin 300381, PR China.
| | - Jingyuan Mao
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, No.88 Changling Road, Xiqing District, Tianjin 300381, PR China.
| |
Collapse
|
5
|
Hu T, Lai X, Li L, Li Y, Wang M, Zhang H, Yang Y, Zhang C, Yan Y, Wang B. UVB-Induced necroptosis of the skin cells via RIPK3-MLKL activation independent of RIPK1 kinase activity. Cell Death Discov 2025; 11:167. [PMID: 40221399 PMCID: PMC11993685 DOI: 10.1038/s41420-025-02471-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2024] [Revised: 03/29/2025] [Accepted: 04/02/2025] [Indexed: 04/14/2025] Open
Abstract
Ultraviolet B (UVB) is recognized for inducing inflammation and death of keratinocytes through the activation of death receptors and pattern recognition receptors (PRRs). Receptor-interacting protein kinase 1 (RIPK1) and RIPK3 play pivotal roles in mediating necroptosis, which can be triggered by the activation of specific death receptors and PRRs. In this study, we observed a reduction of RIPK1 protein after UVB exposure which led to activation of Nuclear factor-kappa B (NF-κB) in HaCaT cells. This activation, in turn, promoted the production of IL-1β and TNF-α. However, RIPK1 kinase remained inactive and did not participate in cell death. Interestingly, UVB radiation triggered the activation of RIPK3 independently of RIPK1 kinase activity and subsequently induced phosphorylation of mixed-lineage kinase domain-like protein (MLKL), culminating in necroptosis and inflammation of the skin. At the same time, UVB-induced activation of RIPK3 also played a role in promoting the mitochondrial apoptotic pathway of Keratinocytes. In conclusion, UVB irradiation initiates an inflammatory response via RIPK1 pathway without necessitating its enzymatic activity. Simultaneously, RIPK3 can be activated by UVB exposure independently of RIPK1's activity, resulting in necroptosis and inflammation of the skin.
Collapse
Affiliation(s)
- Tairan Hu
- Department of Dermatology, Plastic Surgery Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xiaodong Lai
- Department of Dermatology, Plastic Surgery Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Li Li
- Department of Dermatology, Plastic Surgery Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yi Li
- Department of Dermatology, Plastic Surgery Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Meng Wang
- Department of Dermatology, Plastic Surgery Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Haini Zhang
- Department of Dermatology, Plastic Surgery Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yan Yang
- Department of Dermatology, Plastic Surgery Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Chong Zhang
- Department of Dermatology, Plastic Surgery Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yan Yan
- Department of Dermatology, Plastic Surgery Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
| | - Baoxi Wang
- Department of Dermatology, Plastic Surgery Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
| |
Collapse
|
6
|
Alenad AMH, Khan MS, Al-Twaijry N, Alokail MS, Shano LB, Karthikeyan S, Naz H, Jali BR. Suppression of necroptosis-driven cell death and inflammation in hypoxic neuroblastoma (SH-SY5Y) cells by necrostatin-1. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025:10.1007/s00210-025-04023-z. [PMID: 40095052 DOI: 10.1007/s00210-025-04023-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/30/2024] [Accepted: 03/05/2025] [Indexed: 03/19/2025]
Abstract
Neuroblastoma (NB) is the most typical malignant extracranial solid tumor in the pediatric population. The advent of drug resistance is an essential deterrent in treating high-risk NB patients despite a multi-modality remedy. Inflammation-induced early neuronal degeneration plays a leading part in the pathogenesis of NB via necroptosis; however, the mechanisms remained cryptic. Our current investigation determines the anti-inflammatory and neuroprotective effect of necroptosis inhibitor necrostatin-1 (Nec-1) in receptor-interacting proteins 1 and 3 (RIP1/3)-induced cell death pathway and inflammation caused by hypoxia mimetic agent cobalt chloride (CoCl2). Our biomolecular study illustrates that necroptosis marker RIP1/3 and mixed-lineage kinase domain-like pseudokinase (MLKL) protein expression was increased after treatment with CoCl2 in SH-SY5Y cells. Subsequently, elevated expression levels of RIP1/3 and MLKL further contributed to the inflammation by activating transcription factors extracellular signal-regulated protein kinase (ERK1/2), nuclear factor kappa-B (NF-κB), and releasing high levels of proinflammatory cytokines, such as vascular endothelial growth factor (VEGF) and monocyte chemoattractant protein-1 (MCP-1/CCL2). At the same time, Nec-1 treatment reduced the RIP1/3 and MLKL, phospho-ERK1/2, p65 subunit of NF-κB expression, and VEGF and MCP-1 levels. Molecular docking analysis of RIP1/3-necrostatin-1 complex highlights a significant interaction between necrostatin-1 and specific amino acid residues within the protein. Based on our promising results, necrostatin-1 could be exploited as a therapeutic agent during neuroblastoma's pathogenesis and its molecular therapy.
Collapse
Affiliation(s)
- Amal Majed H Alenad
- Department of Biochemistry, College of Science, King Saud University, 11451, Riyadh, Saudi Arabia
| | - Mohd Shahnawaz Khan
- Department of Biochemistry, College of Science, King Saud University, 11451, Riyadh, Saudi Arabia.
| | - Nojood Al-Twaijry
- Department of Biochemistry, College of Science, King Saud University, 11451, Riyadh, Saudi Arabia
| | - Majed S Alokail
- Department of Biochemistry, College of Science, King Saud University, 11451, Riyadh, Saudi Arabia
| | - Leon Bernet Shano
- Division of Physics, School of Advanced Science, Vellore Institute of Technology (VIT) Chennai Campus, Vandalur- Kelambakkam Road, Chennai, Tamil Nadu, 600 127, India
| | - Subramani Karthikeyan
- Centre for Healthcare Advancement, Innovation and Research, Vellore Institute of Technology University, Vandalur- Kelambakkam Road, Vellore, Tamil Nadu, 600 127, India
| | - Huma Naz
- Department of Internal Medicine, University of Missouri, Mizzou, Columbia, MO, 65211, USA
| | - Bigyan Ranjan Jali
- Department of Chemistry, Veer Surendra Sai University of Technology Burla Sambalpur Odisha, Burla, 768018, India.
| |
Collapse
|
7
|
Sun Z, Ye J, Sun W, Jiang L, Shan B, Zhang M, Xu J, Li W, Liu J, Jing H, Zhang T, Hou M, Xie C, Wu R, Pan H, Yuan J. Cooperation of TRADD- and RIPK1-dependent cell death pathways in maintaining intestinal homeostasis. Nat Commun 2025; 16:1890. [PMID: 39987261 PMCID: PMC11846980 DOI: 10.1038/s41467-025-57211-z] [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: 09/11/2024] [Accepted: 02/12/2025] [Indexed: 02/24/2025] Open
Abstract
Dysfunctional NF-κB signaling is critically involved in inflammatory bowel disease (IBD). We investigated the mechanism by which RIPK1 and TRADD, two key mediators of NF-κB signaling, in mediating intestinal pathology using TAK1 IEC deficient model. We show that phosphorylation of TRADD by TAK1 modulates RIPK1-dependent apoptosis. TRADD and RIPK1 act cooperatively to mediate cell death regulated by TNF and TLR signaling. We demonstrate the pathological evolution from RIPK1-dependent ileitis to RIPK1- and TRADD-co-dependent colitis in TAK1 IEC deficient condition. Combined RIPK1 inhibition and TRADD knockout completely protect against intestinal pathology and lethality in TAK1 IEC KO mice. Furthermore, we identify distinctive microbiota dysbiosis biomarkers for RIPK1-dependent ileitis and TRADD-dependent colitis. These findings reveal the cooperation between RIPK1 and TRADD in mediating cell death and inflammation in IBD with NF-κB deficiency and suggest the possibility of combined inhibition of RIPK1 kinase and TRADD as a new therapeutic strategy for IBD.
Collapse
Affiliation(s)
- Ziyu Sun
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, 201203, Shanghai, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
- Shanghai Key Laboratory of Aging Studies, Shanghai, 201210, China
| | - Jianyu Ye
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, 201203, Shanghai, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
- Shanghai Key Laboratory of Aging Studies, Shanghai, 201210, China
| | - Weimin Sun
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, 201203, Shanghai, China
| | - Libo Jiang
- School of Life Sciences and Medicine, Shandong University of Technology, Zibo, Shandong, 255000, China
| | - Bing Shan
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, 201203, Shanghai, China
| | - Mengmeng Zhang
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, 201203, Shanghai, China
| | - Jingyi Xu
- University of Chinese Academy of Sciences, Beijing, 100049, China
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, P.R. China
| | - Wanjin Li
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, 201203, Shanghai, China
| | - Jianping Liu
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, 201203, Shanghai, China
| | - Hongyang Jing
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, 201203, Shanghai, China
| | - Tian Zhang
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, 201203, Shanghai, China
| | - Meiling Hou
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, 201203, Shanghai, China
| | - Cen Xie
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, P.R. China
| | - Rongling Wu
- Beijing Key Laboratory of Topological Statistics and Applications for Complex Systems, Beijing Institute of Mathematical Sciences and Applications, Beijing, 101408, China
- Yau Mathematical Sciences Center, Tsinghua University, Beijing, 100084, China
- Shanghai Institute for Mathematics and Interdisciplinary Sciences, Shanghai, 200433, China
| | - Heling Pan
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, 201203, Shanghai, China
- Shanghai Key Laboratory of Aging Studies, Shanghai, 201210, China
| | - Junying Yuan
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, 201203, Shanghai, China.
- Shanghai Key Laboratory of Aging Studies, Shanghai, 201210, China.
| |
Collapse
|
8
|
Yuan Y, Hu X, Guo C, Xu Y, Li S, Wen W, Hu X, Zeng F, Cui W, Chen W, Sun X, Hou N, Wang J, Xiao RP, Zhang X. Reduction of intestinal RIPK1 ameliorates HFD-induced metabolic disorders in female mice. iScience 2025; 28:111906. [PMID: 40028283 PMCID: PMC11869535 DOI: 10.1016/j.isci.2025.111906] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2024] [Revised: 11/27/2024] [Accepted: 01/23/2025] [Indexed: 03/05/2025] Open
Abstract
In modern society, excessive nutrient intake from food is a major factor contributing to the development of a series of metabolic disorders and cardiovascular diseases. Further investigation of the mechanisms underlying nutrient absorption in the intestine will help to better understand and develop preventive or therapeutic strategies. In this study, using receptor-interacting protein kinase 1 (Ripk1) intestine-specific heterozygous knockout mice (Ripk1 IEC+/-) and high-fat diet (HFD)-feeding mouse model, we report that HFD-induced shift in the transcriptional profile of the ileum toward that of the jejunum, characterized by increased expression of jejunal feature genes in the ileum, are attenuated in Ripk1 IEC+/- female mice, but not in males. Accordingly, HFD-induced metabolic disorders, including obesity, impaired glucose tolerance, insulin resistance, and dyslipidemia, are significantly ameliorated in the Ripk1 IEC+/- female mice. These findings demonstrate a new, sex-specific intestinal regulatory mechanism and highlight the critical role of intestinal RIPK1 in regulating HFD-induced metabolic disorders in females.
Collapse
Affiliation(s)
- Ye Yuan
- Institute of Molecular Medicine, College of Future Technology, Peking University, Beijing 100871, China
- Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Peking University, Beijing 100871, China
- National Biomedical Imaging Center, Peking University, Beijing 100871, China
| | - Xiaomin Hu
- Institute of Molecular Medicine, College of Future Technology, Peking University, Beijing 100871, China
- Department of Medical Research Center, Peking Union Medical College Hospital, Chinese Academy of Medical Science & Peking Union Medical College, Beijing 100730, China
| | - Chunguang Guo
- Institute of Molecular Medicine, College of Future Technology, Peking University, Beijing 100871, China
- Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Peking University, Beijing 100871, China
| | - Yihua Xu
- Institute of Molecular Medicine, College of Future Technology, Peking University, Beijing 100871, China
- Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Peking University, Beijing 100871, China
| | - Shihan Li
- Institute of Molecular Medicine, College of Future Technology, Peking University, Beijing 100871, China
- Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Peking University, Beijing 100871, China
| | - Wei Wen
- Institute of Molecular Medicine, College of Future Technology, Peking University, Beijing 100871, China
- Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Peking University, Beijing 100871, China
- PKU-Nanjing Institute of Translational Medicine, Nanjing 211800, China
| | - Xinli Hu
- Institute of Molecular Medicine, College of Future Technology, Peking University, Beijing 100871, China
- Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Peking University, Beijing 100871, China
| | - Fanxin Zeng
- Institute of Molecular Medicine, College of Future Technology, Peking University, Beijing 100871, China
- Department of Clinical Research Center, Dazhou Central Hospital, Dazhou 635000, China
| | - Weiyi Cui
- Institute of Molecular Medicine, College of Future Technology, Peking University, Beijing 100871, China
- Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Peking University, Beijing 100871, China
| | - Wenli Chen
- Institute of Molecular Medicine, College of Future Technology, Peking University, Beijing 100871, China
- Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Peking University, Beijing 100871, China
| | - Xueting Sun
- Institute of Molecular Medicine, College of Future Technology, Peking University, Beijing 100871, China
- Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Peking University, Beijing 100871, China
| | - Ning Hou
- Institute of Molecular Medicine, College of Future Technology, Peking University, Beijing 100871, China
- Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Peking University, Beijing 100871, China
| | - Jue Wang
- Institute of Molecular Medicine, College of Future Technology, Peking University, Beijing 100871, China
- Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Peking University, Beijing 100871, China
| | - Rui-Ping Xiao
- Institute of Molecular Medicine, College of Future Technology, Peking University, Beijing 100871, China
- Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Peking University, Beijing 100871, China
- State Key Laboratory of Biomembrane and Membrane Biotechnology, Peking-Tsinghua Center for Life Sciences, Beijing 100871, China
| | - Xiuqin Zhang
- Institute of Molecular Medicine, College of Future Technology, Peking University, Beijing 100871, China
- Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Peking University, Beijing 100871, China
- National Biomedical Imaging Center, Peking University, Beijing 100871, China
| |
Collapse
|
9
|
Liu S, Zhang G, Li N, Wang Z, Lu L. The Interplay of Aging and PANoptosis in Osteoarthritis Pathogenesis: Implications for Novel Therapeutic Strategies. J Inflamm Res 2025; 18:1951-1967. [PMID: 39959642 PMCID: PMC11829118 DOI: 10.2147/jir.s489613] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Accepted: 01/22/2025] [Indexed: 02/18/2025] Open
Abstract
Osteoarthritis (OA) is a common degenerative joint disease characterized by the progressive degradation of articular cartilage, synovial inflammation, and subchondral bone remodeling. This review explores the interplay between aging, PANoptosis, and inflammation in OA progression. Age-related cellular and immune dysfunctions, including cellular senescence, senescence-associated secretory phenotypes (SASPs), and immunosenescence, significantly contribute to joint degeneration. In OA, dysregulated apoptosis, necroptosis, and pyroptosis, particularly in chondrocytes, exacerbate cartilage damage. Apoptosis, mediated by the JNK pathway, reduces chondrocyte density, while necroptosis and pyroptosis, involving RIPK-1/RIPK-3 and the NLRP3 inflammasome, respectively, amplify inflammation and cartilage destruction. Inflammatory cytokines and damage-associated molecular patterns (DAMPs) further enhance these PANoptotic pathways. Current therapeutic strategies primarily focus on anti-inflammatory agents such as non-steroidal anti-inflammatory drugs (NSAIDs) and corticosteroids, with growing interest in anti-senescence drugs targeting cellular senescence and SASP. Additionally, exploring PANoptosis mechanisms offers potential for innovative OA treatments.
Collapse
Affiliation(s)
- Shaoshan Liu
- Department of Joint Surgery, Liaocheng Traditional Chinese Medicine Hospital, Liaocheng, 252000, People's Republic of China
| | - Guifeng Zhang
- Department of Neurology, Liaocheng People's Hospital and Liaocheng Hospital Affiliated to Shandong First Medical University, Liaocheng, 252000, People's Republic of China
| | - Nan Li
- Department of Trauma Orthopedics, Liaocheng Traditional Chinese Medicine Hospital, Liaocheng, 252000, People's Republic of China
| | - Zheng Wang
- Department of Neurosurgery, Liaocheng Traditional Chinese Medicine Hospital, Liaocheng, 252000, People's Republic of China
| | - Liaodong Lu
- Department of Joint Surgery, Liaocheng Traditional Chinese Medicine Hospital, Liaocheng, 252000, People's Republic of China
| |
Collapse
|
10
|
Zhang W, Liu H, Zhang D, Yi Y, Tao L, Zhu Y, Huang S, Zhao X, Shao Q, Li P, Weng Y, Lu W, Zhang J, Zhang H, Chen Y, Weng D. Role of hepatocyte RIPK1 in maintaining liver homeostasis during metabolic challenges. eLife 2025; 13:RP96798. [PMID: 39886919 PMCID: PMC11785375 DOI: 10.7554/elife.96798] [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: 02/01/2025] Open
Abstract
As a central hub for metabolism, the liver exhibits strong adaptability to maintain homeostasis in response to food fluctuations throughout evolution. However, the mechanisms governing this resilience remain incompletely understood. In this study, we identified Receptor interacting protein kinase 1 (RIPK1) in hepatocytes as a critical regulator in preserving hepatic homeostasis during metabolic challenges, such as short-term fasting or high-fat dieting. Our results demonstrated that hepatocyte-specific deficiency of RIPK1 sensitized the liver to short-term fasting-induced liver injury and hepatocyte apoptosis in both male and female mice. Despite being a common physiological stressor that typically does not induce liver inflammation, short-term fasting triggered hepatic inflammation and compensatory proliferation in hepatocyte-specific RIPK1-deficient (Ripk1-hepKO) mice. Transcriptomic analysis revealed that short-term fasting oriented the hepatic microenvironment into an inflammatory state in Ripk1-hepKO mice, with up-regulated expression of inflammation and immune cell recruitment-associated genes. Single-cell RNA sequencing further confirmed the altered cellular composition in the liver of Ripk1-hepKO mice during fasting, highlighting the increased recruitment of macrophages to the liver. Mechanically, our results indicated that ER stress was involved in fasting-induced liver injury in Ripk1-hepKO mice. Overall, our findings revealed the role of RIPK1 in maintaining liver homeostasis during metabolic fluctuations and shed light on the intricate interplay between cell death, inflammation, and metabolism.
Collapse
Affiliation(s)
- Weigao Zhang
- School of Environmental and Biological Engineering, Key Laboratory of Metabolic Engineering and Biosynthesis Technology, Ministry of Industry and Information Technology, Nanjing University of Science and TechnologyNanjingChina
| | - Hu Liu
- School of Environmental and Biological Engineering, Key Laboratory of Metabolic Engineering and Biosynthesis Technology, Ministry of Industry and Information Technology, Nanjing University of Science and TechnologyNanjingChina
- Department of Laboratory Medicine, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing UniversityNanjingChina
| | - Danyang Zhang
- School of Environmental and Biological Engineering, Key Laboratory of Metabolic Engineering and Biosynthesis Technology, Ministry of Industry and Information Technology, Nanjing University of Science and TechnologyNanjingChina
| | - Yuguo Yi
- School of Medicine, Shenzhen Campus of Sun Yat-sen University, Sun Yat-sen UniversityShenzhenChina
| | - Liang Tao
- The First Affiliated Hospital, Basic Medical Sciences, University of South ChinaHengyangChina
| | - Yunfeng Zhu
- School of Environmental and Biological Engineering, Key Laboratory of Metabolic Engineering and Biosynthesis Technology, Ministry of Industry and Information Technology, Nanjing University of Science and TechnologyNanjingChina
| | - Shuxian Huang
- School of Environmental and Biological Engineering, Key Laboratory of Metabolic Engineering and Biosynthesis Technology, Ministry of Industry and Information Technology, Nanjing University of Science and TechnologyNanjingChina
| | - Xunan Zhao
- School of Environmental and Biological Engineering, Key Laboratory of Metabolic Engineering and Biosynthesis Technology, Ministry of Industry and Information Technology, Nanjing University of Science and TechnologyNanjingChina
| | - Qianchao Shao
- School of Environmental and Biological Engineering, Key Laboratory of Metabolic Engineering and Biosynthesis Technology, Ministry of Industry and Information Technology, Nanjing University of Science and TechnologyNanjingChina
| | - Peiqi Li
- School of Environmental and Biological Engineering, Key Laboratory of Metabolic Engineering and Biosynthesis Technology, Ministry of Industry and Information Technology, Nanjing University of Science and TechnologyNanjingChina
| | - Yiwen Weng
- Internal Medicine Department, Chengdu Jinniu District People's HospitalChengduChina
| | - Wei Lu
- Affiliated Hospital of Nanjing University of Chinese MedicineNanjingChina
| | - Jianfa Zhang
- School of Environmental and Biological Engineering, Key Laboratory of Metabolic Engineering and Biosynthesis Technology, Ministry of Industry and Information Technology, Nanjing University of Science and TechnologyNanjingChina
| | - Haibing Zhang
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of SciencesShanghaiChina
| | - Yuxin Chen
- Department of Laboratory Medicine, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing UniversityNanjingChina
| | - Dan Weng
- School of Environmental and Biological Engineering, Key Laboratory of Metabolic Engineering and Biosynthesis Technology, Ministry of Industry and Information Technology, Nanjing University of Science and TechnologyNanjingChina
| |
Collapse
|
11
|
Walsh RB, McNaughton P, Nademi Z, Laberko A, Balashov D, Al-Mousa H, Arkwright PD, Wynn RF, Flood T, Williams E, Cant A, Abinun M, Hambleton S, Slatter M, Gennery AR, Lum SH, Owens S. Outcomes of Hematopoietic Stem Cell Transplantation in 5 Patients with Autosomal Recessive RIPK1-Deficiency. J Clin Immunol 2025; 45:65. [PMID: 39762600 PMCID: PMC11703983 DOI: 10.1007/s10875-024-01850-2] [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: 12/28/2023] [Accepted: 12/13/2024] [Indexed: 01/11/2025]
Abstract
Receptor Interacting Serine/Threonine Kinase 1 (RIPK1) is widely expressed and integral to inflammatory and cell death responses. Autosomal recessive RIPK1-deficiency, due to biallelic loss of function mutations in RIPK1, is a rare inborn error of immunity (IEI) resulting in uncontrolled necroptosis, apoptosis and inflammation. Although hematopoietic stem cell transplantation (HSCT) has been suggested as a potential curative therapy, the extent to which disease may be driven by extra-hematopoietic effects of RIPK1-deficiency, which are non-amenable to HSCT, is not clear. We present a multi-centre, international review of an additional 5 RIPK1-deficient children who underwent HSCT. All patients presented with very early onset inflammatory bowel disease, 2 also suffered from inflammatory arthritis. Median age at transplant was 3 years (range 1-5 years); 1 received matched sibling marrow, 1 matched unrelated peripheral blood stem cells (PBSC), 2 TCRαβ/CD19-depleted PBSC from maternal-haploidentical donors, and 1 had TCRαβ/CD19-depleted PBSC from a mismatched unrelated donor. All received reduced-toxicity conditioning, based on treosulfan (n = 4) or busulfan (n = 1); 1 patient underwent a successful second transplant following autologous reconstitution. Four of five patients (80%) survived; 1 child died due to multi-drug resistant pseudomonas infection and multi-organ failure. With a median duration of 14 months follow-up, 2 survivors were disease-free, and 2 had substantially improving enteropathy. These findings demonstrated that HSCT is a potential curative therapy for RIPK1-deficiency.
Collapse
Affiliation(s)
- Rebecca B Walsh
- Royal Victoria Infirmary, Newcastle-Upon-Tyne, UK
- Population Health Sciences Institute, Newcastle University, Newcastle-Upon-Tyne, UK
| | | | - Zohreh Nademi
- Great North Children's Hospital, Newcastle-Upon-Tyne, UK
- Translational and Clinical Research Institute, Newcastle University, Newcastle-Upon-Tyne, UK
| | - Alexandra Laberko
- Hematopoietic Stem Cell Transplantation, Dmitry Rogachev National Medical Research Centre of Paediatric Haematology, Oncology and Immunology, Moscow, Russia
| | - Dmitry Balashov
- Hematopoietic Stem Cell Transplantation, Dmitry Rogachev National Medical Research Centre of Paediatric Haematology, Oncology and Immunology, Moscow, Russia
| | - Hamoud Al-Mousa
- Section of Paediatric Allergy and Immunology, Department of Paediatrics, King Faisal Specialist Hospital and Research Centre, Riyadh, Saudi Arabia
| | - Peter D Arkwright
- Lydia Becker Institute of Immunology and Inflammation, University of Manchester & Royal Manchester Children's Hospital, Manchester, UK
| | - Robert F Wynn
- Blood and Marrow Transplant Unit, Royal Manchester Children's Hospital, Central Manchester University Hospitals NHS Foundation Trust, Manchester, UK
| | - Terry Flood
- Great North Children's Hospital, Newcastle-Upon-Tyne, UK
| | - Eleri Williams
- Great North Children's Hospital, Newcastle-Upon-Tyne, UK
| | - Andrew Cant
- Great North Children's Hospital, Newcastle-Upon-Tyne, UK
- Translational and Clinical Research Institute, Newcastle University, Newcastle-Upon-Tyne, UK
| | - Mario Abinun
- Great North Children's Hospital, Newcastle-Upon-Tyne, UK
- Translational and Clinical Research Institute, Newcastle University, Newcastle-Upon-Tyne, UK
| | - Sophie Hambleton
- Great North Children's Hospital, Newcastle-Upon-Tyne, UK
- Translational and Clinical Research Institute, Newcastle University, Newcastle-Upon-Tyne, UK
| | - Mary Slatter
- Great North Children's Hospital, Newcastle-Upon-Tyne, UK
- Translational and Clinical Research Institute, Newcastle University, Newcastle-Upon-Tyne, UK
| | - Andrew R Gennery
- Great North Children's Hospital, Newcastle-Upon-Tyne, UK
- Translational and Clinical Research Institute, Newcastle University, Newcastle-Upon-Tyne, UK
| | - Su Han Lum
- Great North Children's Hospital, Newcastle-Upon-Tyne, UK
- Translational and Clinical Research Institute, Newcastle University, Newcastle-Upon-Tyne, UK
| | - Stephen Owens
- Population Health Sciences Institute, Newcastle University, Newcastle-Upon-Tyne, UK.
- Great North Children's Hospital, Newcastle-Upon-Tyne, UK.
| |
Collapse
|
12
|
Milosevic M, Magnutzki A, Braun T, Hussain S, Jakschitz T, Kragl M, Soeberdt M, Nausch B, Bonn GK, Huber LA, Valovka T. Anti-inflammatory and cytoprotective polypharmacology of Canephron N reveals targeting of the IKK-NF-κB and p38-MK2-RIPK1 axes. Biomed Pharmacother 2025; 182:117747. [PMID: 39671726 DOI: 10.1016/j.biopha.2024.117747] [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: 10/08/2024] [Revised: 12/06/2024] [Accepted: 12/08/2024] [Indexed: 12/15/2024] Open
Abstract
Urinary tract infections are among the most frequently occurring forms of infection, and inflammation and tissue damage contribute significantly to symptoms, e.g., dysuria and urge. Canephron N is an orally bioavailable herbal medicine with anti-inflammatory, spasmolytic, anti-adhesive, and anti-nociceptive therapeutic effects that is approved for the treatment of uncomplicated urinary tract infections. Here, we used renal tubular epithelial HK-2 cells to study the anti-inflammatory and cytoprotective effects and molecular mechanisms of its active component, BNO 2103. BNO 2103 suppressed nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) activation by lipopolysaccharide (LPS) and tumor necrosis factor alpha (TNFα) and prevented inhibitory κB kinase (IKK)-dependent phosphorylation and degradation of inhibitor of nuclear factor kappa B alpha (IκBα). BNO 2103 also suppressed the inflammation-specific S536 phosphorylation of the NF-κB subunit p65 and the production of a specific set of inflammatory cytokines. Unlike other NF-κB inhibitors, BNO 2103 demonstrated cytoprotection against TNFα-induced cytotoxicity. Our data suggest that BNO 2103 acts primarily through the mitogen-activated protein kinase p38 (p38 MAPK)-MAPK-activated protein kinase 2 (MK2) axis by promoting receptor-interacting serine/threonine protein kinase 1 (RIPK1) phosphorylation at S320. Simultaneously, it suppresses S166 autophosphorylation and subsequent activation of RIPK1, which is required for apoptotic and necroptotic responses to TNFα. This study confirms Canephron N as an effective alternative to traditional anti-inflammatory drugs and provides initial evidence of its ability to inhibit apoptosis and necroptosis in the urogenital system. It also presents a detailed pathway investigation that identifies the specific targets of Canephron N within the NF-κB signaling cascade.
Collapse
Affiliation(s)
- Marija Milosevic
- ADSI-Austrian Drug Screening Institute, Leopold-Franzens University of Innsbruck, Innsbruck 6020, Austria
| | - Alexander Magnutzki
- ADSI-Austrian Drug Screening Institute, Leopold-Franzens University of Innsbruck, Innsbruck 6020, Austria
| | - Theodor Braun
- ADSI-Austrian Drug Screening Institute, Leopold-Franzens University of Innsbruck, Innsbruck 6020, Austria
| | - Shah Hussain
- ADSI-Austrian Drug Screening Institute, Leopold-Franzens University of Innsbruck, Innsbruck 6020, Austria
| | - Thomas Jakschitz
- ADSI-Austrian Drug Screening Institute, Leopold-Franzens University of Innsbruck, Innsbruck 6020, Austria
| | | | | | | | - Günther K Bonn
- ADSI-Austrian Drug Screening Institute, Leopold-Franzens University of Innsbruck, Innsbruck 6020, Austria.
| | - Lukas A Huber
- Institute of Cell Biology, Biocenter, Medical University of Innsbruck, Innsbruck 6020, Austria; ADSI-Austrian Drug Screening Institute, Leopold-Franzens University of Innsbruck, Innsbruck 6020, Austria.
| | - Taras Valovka
- Institute of Cell Biology, Biocenter, Medical University of Innsbruck, Innsbruck 6020, Austria; Department of Pediatrics I, Medical University of Innsbruck, Innsbruck 6020, Austria.
| |
Collapse
|
13
|
Tak J, Kim YS, Kim SG. Roles of X-box binding protein 1 in liver pathogenesis. Clin Mol Hepatol 2025; 31:1-31. [PMID: 39355873 PMCID: PMC11791611 DOI: 10.3350/cmh.2024.0441] [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: 06/10/2024] [Revised: 09/06/2024] [Accepted: 09/27/2024] [Indexed: 10/03/2024] Open
Abstract
The prevalence of drug-induced liver injury (DILI) and viral liver infections presents significant challenges in modern healthcare and contributes to considerable morbidity and mortality worldwide. Concurrently, metabolic dysfunctionassociated steatotic liver disease (MASLD) has emerged as a major public health concern, reflecting the increasing rates of obesity and leading to more severe complications such as fibrosis and hepatocellular carcinoma. X-box binding protein 1 (XBP1) is a distinct transcription factor with a basic-region leucine zipper structure, whose activity is regulated by alternative splicing in response to disruptions in endoplasmic reticulum (ER) homeostasis and the unfolded protein response (UPR) activation. XBP1 interacts with a key signaling component of the highly conserved UPR and is critical in determining cell fate when responding to ER stress in liver diseases. This review aims to elucidate the emerging roles and molecular mechanisms of XBP1 in liver pathogenesis, focusing on its involvement in DILI, viral liver infections, MASLD, fibrosis/cirrhosis, and liver cancer. Understanding the multifaceted functions of XBP1 in these liver diseases offers insights into potential therapeutic strategies to restore ER homeostasis and mitigate liver damage.
Collapse
Affiliation(s)
- Jihoon Tak
- College of Pharmacy and Integrated Research Institute for Drug Development, Dongguk University-Seoul, Goyang, Korea
| | - Yun Seok Kim
- Department of Clinical Pharmacology and Therapeutics, Seoul National University College of Medicine, Seoul, Korea
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, Korea
| | - Sang Geon Kim
- College of Pharmacy and Integrated Research Institute for Drug Development, Dongguk University-Seoul, Goyang, Korea
| |
Collapse
|
14
|
Chen J, Zhao L, Xu MF, Huang D, Sun XL, Zhang YX, Li HM, Wu CZ. Novel isobavachalcone derivatives induce apoptosis and necroptosis in human non-small cell lung cancer H1975 cells. J Enzyme Inhib Med Chem 2024; 39:2292006. [PMID: 38086769 PMCID: PMC11721617 DOI: 10.1080/14756366.2023.2292006] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Revised: 11/17/2023] [Accepted: 12/03/2023] [Indexed: 12/18/2023] Open
Abstract
In this study, seventeen isobavachalcone (IBC) derivatives (1-17) were synthesised, and evaluated for their cytotoxic activity against three human lung cancer cell lines. Among these derivatives, compound 16 displayed the most potent cytotoxic activity against H1975 and A549 cells, with IC50 values of 4.35 and 14.21 μM, respectively. Compared with IBC, compound 16 exhibited up to 4.11-fold enhancement of cytotoxic activity on human non-small cell lung cancer H1975 cells. In addition, we found that compound 16 suppressed H1975 cells via inducing apoptosis and necroptosis. The initial mechanism of compound 16 induced cell death in H1975 cells involves the increasing of Bax/Bcl-2 ratio and Cyt C protein level, down-regulating of Akt protein level, and cleaving caspase-9 and -3 induced apoptosis; the up-regulation of RIP3, p-RIP3, MLKL, and p-MLKL levels induced necroptosis. Moreover, compound 16 also caused mitochondrial dysfunction, thereby decreasing cellular ATP levels, and resulting in excessive reactive oxygen species (ROS) accumulation.
Collapse
Affiliation(s)
- Jie Chen
- School of Pharmacy, Bengbu Medical College, Bengbu, Anhui, China
| | - Long Zhao
- School of Pharmacy, Bengbu Medical College, Bengbu, Anhui, China
- Anhui Province Biochemical Pharmaceutical Engineering Technology Research Center, Bengbu, Anhui, China
| | - Meng-Fan Xu
- School of Pharmacy, Bengbu Medical College, Bengbu, Anhui, China
| | - Di Huang
- School of Pharmacy, Bengbu Medical College, Bengbu, Anhui, China
| | - Xiao-Long Sun
- School of Pharmacy, Bengbu Medical College, Bengbu, Anhui, China
| | - Yu-Xin Zhang
- Anhui Province Biochemical Pharmaceutical Engineering Technology Research Center, Bengbu, Anhui, China
- School of Laboratory Medicine, Bengbu Medical College, Bengbu, Anhui, China
| | - Hong-Mei Li
- School of Pharmacy, Bengbu Medical College, Bengbu, Anhui, China
- Anhui Province Biochemical Pharmaceutical Engineering Technology Research Center, Bengbu, Anhui, China
| | - Cheng-Zhu Wu
- School of Pharmacy, Bengbu Medical College, Bengbu, Anhui, China
- Anhui Province Biochemical Pharmaceutical Engineering Technology Research Center, Bengbu, Anhui, China
| |
Collapse
|
15
|
He R, Liu Y, Fu W, He X, Liu S, Xiao D, Tao Y. Mechanisms and cross-talk of regulated cell death and their epigenetic modifications in tumor progression. Mol Cancer 2024; 23:267. [PMID: 39614268 PMCID: PMC11606237 DOI: 10.1186/s12943-024-02172-y] [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: 08/21/2024] [Accepted: 11/07/2024] [Indexed: 12/01/2024] Open
Abstract
Cell death is a fundamental part of life for metazoans. To maintain the balance between cell proliferation and metabolism of human bodies, a certain number of cells need to be removed regularly. Hence, the mechanisms of cell death have been preserved during the evolution of multicellular organisms. Tumorigenesis is closely related with exceptional inhibition of cell death. Mutations or defects in cell death-related genes block the elimination of abnormal cells and enhance the resistance of malignant cells to chemotherapy. Therefore, the investigation of cell death mechanisms enables the development of drugs that directly induce tumor cell death. In the guidelines updated by the Cell Death Nomenclature Committee (NCCD) in 2018, cell death was classified into 12 types according to morphological, biochemical and functional classification, including intrinsic apoptosis, extrinsic apoptosis, mitochondrial permeability transition (MPT)-driven necrosis, necroptosis, ferroptosis, pyroptosis, PARP-1 parthanatos, entotic cell death, NETotic cell death, lysosome-dependent cell death, autophagy-dependent cell death, immunogenic cell death, cellular senescence and mitotic catastrophe. The mechanistic relationships between epigenetic controls and cell death in cancer progression were previously unclear. In this review, we will summarize the mechanisms of cell death pathways and corresponding epigenetic regulations. Also, we will explore the extensive interactions between these pathways and discuss the mechanisms of cell death in epigenetics which bring benefits to tumor therapy.
Collapse
Affiliation(s)
- Ruimin He
- Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, 410078, China
- Cancer Research Institute and School of Basic Medicine, Central South University, Changsha, Hunan, 410078, China
- Department of Pathology, Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Xiangya Hospital, Central South University, Hunan, 410078, China
| | - Yifan Liu
- Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, 410078, China
- Cancer Research Institute and School of Basic Medicine, Central South University, Changsha, Hunan, 410078, China
- Department of Pathology, Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Xiangya Hospital, Central South University, Hunan, 410078, China
| | - Weijie Fu
- Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, 410078, China
- Cancer Research Institute and School of Basic Medicine, Central South University, Changsha, Hunan, 410078, China
- Department of Pathology, Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Xiangya Hospital, Central South University, Hunan, 410078, China
| | - Xuan He
- Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, 410078, China
- Cancer Research Institute and School of Basic Medicine, Central South University, Changsha, Hunan, 410078, China
- Department of Pathology, Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Xiangya Hospital, Central South University, Hunan, 410078, China
| | - Shuang Liu
- Department of Oncology, Institute of Medical Sciences, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China.
| | - Desheng Xiao
- Department of Pathology, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China.
| | - Yongguang Tao
- Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, 410078, China.
- Cancer Research Institute and School of Basic Medicine, Central South University, Changsha, Hunan, 410078, China.
- Department of Pathology, Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Xiangya Hospital, Central South University, Hunan, 410078, China.
- Department of Pathology, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China.
- Department of Thoracic Surgery, Hunan Key Laboratory of Early Diagnosis and Precision Therapy in Lung Cancer, Second Xiangya Hospital, Central South University, Changsha, 410011, China.
- Furong Laboratory, Xiangya School of Medicine, Central South University, Hunan, 410078, China.
| |
Collapse
|
16
|
Inuzuka H, Qian C, Qi Y, Xiong Y, Wang C, Wang Z, Zhang D, Zhang C, Jin J, Wei W. Targeted Degradation of Receptor-Interacting Protein Kinase 1 to Modulate the Necroptosis Pathway. ACS Pharmacol Transl Sci 2024; 7:3518-3526. [PMID: 39539258 PMCID: PMC11555510 DOI: 10.1021/acsptsci.4c00421] [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: 07/17/2024] [Revised: 10/02/2024] [Accepted: 10/04/2024] [Indexed: 11/16/2024]
Abstract
Necroptosis is a highly regulated form of necrotic cell death that plays an essential role in pathogen defense and tissue homeostasis. Abnormal regulation of the necroptotic pathway has been implicated in the pathogenesis of various human diseases, including cancer, inflammatory, and neurodegenerative diseases. Receptor-interacting protein kinase 1 (RIPK1) serves as a crucial regulator of the necroptotic signaling pathway and has been identified as a potential therapeutic target. Mechanistically, RIPK1 serves as both a protein kinase and a scaffolding protein, fulfilling its dual function through a combination of kinase activity-dependent and kinase activity-independent mechanisms. Thus, employing a targeted RIPK1 knockdown strategy is a highly effective means of inhibiting RIPK1 functions. To achieve a targeted RIPK1 knockdown, we generated a RIPK1-PROTAC, MS2031, by connecting the ZB-R-55 RIPK1 binder to the VHL ligand, thereby recruiting the CUL2-RING-VHL (CRL2VHL) E3 ubiquitin ligase complex for targeted degradation of RIPK1 through the 26S proteasome. Notably, MS2031 treatment effectively reduced the abundance of RIPK1 protein in the nanomolar range in various cell lines we examined, including HT-29 and T47D cells, and modulated the necroptosis signaling pathway. These results suggest that MS2031 may hold potential for the treatment of human diseases resulting from aberrant regulation of RIPK1.
Collapse
Affiliation(s)
- Hiroyuki Inuzuka
- Department
of Pathology, Beth Israel Deaconess Medical
Center, Harvard Medical School, Boston, Massachusetts 02215, United States
| | - Chao Qian
- Mount
Sinai Center for Therapeutics Discovery, Departments of Pharmacological
Sciences, Oncological Sciences and Neuroscience, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, New York 10029, United States
| | - Yihang Qi
- Department
of Pathology, Beth Israel Deaconess Medical
Center, Harvard Medical School, Boston, Massachusetts 02215, United States
| | - Yan Xiong
- Mount
Sinai Center for Therapeutics Discovery, Departments of Pharmacological
Sciences, Oncological Sciences and Neuroscience, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, New York 10029, United States
| | - Chaoyu Wang
- Department
of Pathology, Beth Israel Deaconess Medical
Center, Harvard Medical School, Boston, Massachusetts 02215, United States
| | - Zhen Wang
- Department
of Pathology, Beth Israel Deaconess Medical
Center, Harvard Medical School, Boston, Massachusetts 02215, United States
| | - Dingpeng Zhang
- Department
of Pathology, Beth Israel Deaconess Medical
Center, Harvard Medical School, Boston, Massachusetts 02215, United States
| | - Can Zhang
- Genetics
and Aging Research Unit, McCance Center for Brain Health, Mass General
Institute for Neurodegenerative Disease, Department of Neurology, Massachusetts General Hospital, Harvard Medical School, 114 16th Street, Charlestown, Massachusetts 02129, United States
| | - Jian Jin
- Mount
Sinai Center for Therapeutics Discovery, Departments of Pharmacological
Sciences, Oncological Sciences and Neuroscience, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, New York 10029, United States
| | - Wenyi Wei
- Department
of Pathology, Beth Israel Deaconess Medical
Center, Harvard Medical School, Boston, Massachusetts 02215, United States
| |
Collapse
|
17
|
Zhao H, Wang C, Liu B, Weng Z, Shi Y, Zhang C. RIP1 inhibition reduces chondrocyte apoptosis through downregulating nuclear factor-kappa B signaling in a mouse osteoarthritis model. Mol Biol Rep 2024; 51:1132. [PMID: 39514126 DOI: 10.1007/s11033-024-10080-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Accepted: 10/29/2024] [Indexed: 11/16/2024]
Abstract
BACKGROUND Excessive chondrocyte death is a critical player in the process of osteoarthritis (OA). The present study was aimed to study the role of receptor-interacting serine/threonine kinase (RIP) 1-mediated signaling for programmed cell death in OA. METHODS In the present study, RIP1 protein expression was evaluated in mouse OA cartilage and cultured primary murine chondrocytes exposed to tumor necrosis factor-alpha (TNF-α). Protein expression involved in necroptosis and apoptosis and chondrocyte-derived extracellular matrix were examined. Inhibition of RIP1 was conducted using the RNAi technique and pharmacological inhibition. Western blot, immunohistochemistry, and immunofluorescence examination were applied. RESULTS The protein presence of RIP1, but not RIP3, was increased in the mouse OA tissue and cultured chondrocytes exposed to TNF-α. Knockdown of RIP1 increased protein expression of collagen II and sex-determining region Y-box transcription factor 9, and reduced protein expression of matrix metallopeptidases 13 and a disintegrin and metalloproteinase with thrombospondin motifs 5. Inhibition of RIP1 reduced the phosphorylated NF-κB signals, decreased cell apoptosis, and restored extracellular matrix expression in cultured chondrocytes. Both RNAi and pharmacological inhibition of RIP1 decelerated the progress of OA in mice. CONCLUSION RIP1 regulates chondrocyte apoptosis through NF-κB signaling. Inhibition of RIP1 provides a novel therapeutic approach for OA therapy.
Collapse
Affiliation(s)
- Hong Zhao
- Department of Orthopedic Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Chenzhong Wang
- Department of Orthopedic Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Bo Liu
- Department of Orthopedic Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Ziyu Weng
- Department of Orthopedic Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Yi Shi
- Department of Kidney Transplantation, Zhongshan Hospital, Fudan University, Shanghai, 200032, China.
- Shanghai Key Laboratory of Organ Transplantation, Zhongshan Hospital, Fudan University, Shanghai, China.
| | - Chi Zhang
- Department of Orthopedic Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200032, China.
| |
Collapse
|
18
|
Liu H, Sheng Q, Dan J, Xie X. Crosstalk and Prospects of TBK1 in Inflammation. Immunol Invest 2024; 53:1205-1233. [PMID: 39194013 DOI: 10.1080/08820139.2024.2392587] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/29/2024]
Abstract
BACKGROUND TANK-binding kinase 1 (TBK1) is a pivotal mediator of innate immunity, activated by receptors such as mitochondrial antiviral signaling protein (MAVS), stimulator of interferon genes (STING), and TIR-domain-containing adaptor inducing interferon-β (TRIF). It modulates immune responses by exerting influence on the type I interferons (IFN-Is) signaling and the nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) pathways, Over the past few years, TBK1 multifaceted role in both immune and inflammatory responses is increasingly recognized. METHODS AND RESULTS This review aims to scrutinize how TBK1 operates within the NF-κB pathway and the interferon regulatory transcription factor 3 (IRF3)-dependent IFN-I pathways, highlighting the kinases and other molecules involved in these processes. This analysis reveals the distinctive characteristics of TBK1's involvement in these pathways. Furthermore, it has been observed that the role of TBK1 in exerting anti-inflammatory or pro-inflammatory effects is contingent upon varying pathological conditions, indicating a multifaceted role in immune regulation. DISCUSSION TBK1's evolving role in various diseases and the potential of TBK1 inhibitors as therapeutic agents are explored. Targeting TBK1 may provide new strategies for treating inflammatory disorders and autoimmune diseases associated with IFN-Is, warranting further investigation.
Collapse
Affiliation(s)
- Huan Liu
- Laboratory of Molecular Genetics of Aging and Tumor, Medical School, Kunming University of Science and Technology, Kunming, China
| | - Qihuan Sheng
- Laboratory of Molecular Genetics of Aging and Tumor, Medical School, Kunming University of Science and Technology, Kunming, China
| | - Juhua Dan
- Laboratory of Molecular Genetics of Aging and Tumor, Medical School, Kunming University of Science and Technology, Kunming, China
| | - Xiaoli Xie
- Laboratory of Molecular Genetics of Aging and Tumor, Medical School, Kunming University of Science and Technology, Kunming, China
| |
Collapse
|
19
|
Hushmandi K, Einollahi B, Aow R, Suhairi SB, Klionsky DJ, Aref AR, Reiter RJ, Makvandi P, Rabiee N, Xu Y, Nabavi N, Saadat SH, Farahani N, Kumar AP. Investigating the interplay between mitophagy and diabetic neuropathy: Uncovering the hidden secrets of the disease pathology. Pharmacol Res 2024; 208:107394. [PMID: 39233055 PMCID: PMC11934918 DOI: 10.1016/j.phrs.2024.107394] [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: 05/12/2024] [Revised: 08/18/2024] [Accepted: 08/30/2024] [Indexed: 09/06/2024]
Abstract
Mitophagy, the cellular process of selectively eliminating damaged mitochondria, plays a crucial role in maintaining metabolic balance and preventing insulin resistance, both key factors in type 2 diabetes mellitus (T2DM) development. When mitophagy malfunctions in diabetic neuropathy, it triggers a cascade of metabolic disruptions, including reduced energy production, increased oxidative stress, and cell death, ultimately leading to various complications. Thus, targeting mitophagy to enhance the process may have emerged as a promising therapeutic strategy for T2DM and its complications. Notably, plant-derived compounds with β-cell protective and mitophagy-stimulating properties offer potential as novel therapeutic agents. This review highlights the intricate mechanisms linking mitophagy dysfunction to T2DM and its complications, particularly neuropathy, elucidating potential therapeutic interventions for this debilitating disease.
Collapse
Affiliation(s)
- Kiavash Hushmandi
- Nephrology and Urology Research Center, Clinical Sciences Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran.
| | - Behzad Einollahi
- Nephrology and Urology Research Center, Clinical Sciences Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Rachel Aow
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore; NUS Center for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Suhana Binte Suhairi
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore; NUS Center for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Daniel J Klionsky
- Life Sciences Institute, University of Michigan, Ann Arbor, MI 48109, USA
| | - Amir Reza Aref
- Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Russel J Reiter
- Department of Cell Systems and Anatomy, UT Health San Antonio, Long School of Medicine, San Antonio, TX, USA
| | - Pooyan Makvandi
- Department of Biomaterials, Saveetha Dental College and Hospitals, SIMATS, Saveetha University, Chennai 600077, India; University Centre for Research & Development, Chandigarh University, Mohali, Punjab 140413, India
| | - Navid Rabiee
- Department of Biomaterials, Saveetha Dental College and Hospitals, SIMATS, Saveetha University, Chennai 600077, India
| | - Yi Xu
- Department of Science & Technology, Department of Urology, NanoBioMed Group, The Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou People's Hospital, Quzhou 324000, China
| | - Noushin Nabavi
- Independent Researcher, Victoria, British Columbia V8V 1P7, Canada
| | - Seyed Hassan Saadat
- Nephrology and Urology Research Center, Clinical Sciences Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Najma Farahani
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran.
| | - Alan Prem Kumar
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore; NUS Center for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.
| |
Collapse
|
20
|
Zhang Z, Yang Z, Wang S, Wang X, Mao J. Overview of pyroptosis mechanism and in-depth analysis of cardiomyocyte pyroptosis mediated by NF-κB pathway in heart failure. Biomed Pharmacother 2024; 179:117367. [PMID: 39214011 DOI: 10.1016/j.biopha.2024.117367] [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: 07/17/2024] [Revised: 08/14/2024] [Accepted: 08/26/2024] [Indexed: 09/04/2024] Open
Abstract
The pyroptosis of cardiomyocytes has become an essential topic in heart failure research. The abnormal accumulation of these biological factors, including angiotensin II, advanced glycation end products, and various growth factors (such as connective tissue growth factor, vascular endothelial growth factor, transforming growth factor beta, among others), activates the nuclear factor-κB (NF-κB) signaling pathway in cardiovascular diseases, ultimately leading to pyroptosis of cardiomyocytes. Therefore, exploring the underlying molecular biological mechanisms is essential for developing novel drugs and therapeutic strategies. However, our current understanding of the precise regulatory mechanism of this complex signaling pathway in cardiomyocyte pyroptosis is still limited. Given this, this study reviews the milestone discoveries in the field of pyroptosis research since 1986, analyzes in detail the similarities, differences, and interactions between pyroptosis and other cell death modes (such as apoptosis, necroptosis, autophagy, and ferroptosis), and explores the deep connection between pyroptosis and heart failure. At the same time, it depicts in detail the complete pathway of the activation, transmission, and eventual cardiomyocyte pyroptosis of the NF-κB signaling pathway in the process of heart failure. In addition, the study also systematically summarizes various therapeutic approaches that can inhibit NF-κB to reduce cardiomyocyte pyroptosis, including drugs, natural compounds, small molecule inhibitors, gene editing, and other cutting-edge technologies, aiming to provide solid scientific support and new research perspectives for the prevention and treatment of heart failure.
Collapse
Affiliation(s)
- Zeyu Zhang
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin 300381, China; Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Zhihua Yang
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin 300381, China; Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Shuai Wang
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin 300381, China
| | - Xianliang Wang
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin 300381, China.
| | - Jingyuan Mao
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin 300381, China.
| |
Collapse
|
21
|
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
|
22
|
Park JM, Park JE, Park JS, Leem YH, Kim DY, Hyun JW, Kim HS. Anti-inflammatory and antioxidant mechanisms of coniferaldehyde in lipopolysaccharide-induced neuroinflammation: Involvement of AMPK/Nrf2 and TAK1/MAPK/NF-κB signaling pathways. Eur J Pharmacol 2024; 979:176850. [PMID: 39059571 DOI: 10.1016/j.ejphar.2024.176850] [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: 05/30/2024] [Revised: 07/09/2024] [Accepted: 07/24/2024] [Indexed: 07/28/2024]
Abstract
Microglia are primarily involved in inflammatory reactions and oxidative stress in the brain; as such reducing microglial activation has been proposed as a potential therapeutic strategy for neurodegenerative disorders. Herein, we investigated the anti-inflammatory and antioxidant activities of coniferaldehyde (CFA), a naturally occurring cinnamaldehyde derivative, on activated microglia to evaluate its therapeutic potential. CFA inhibited the production of nitric oxide (NO) and proinflammatory cytokines, such as tumor necrosis factor-α, interleukin (IL)-1β, and IL-6, in lipopolysaccharide (LPS)-stimulated BV2 microglial cells. CFA also inhibited intracellular reactive oxygen species levels and oxidative stress markers such as 4-HNE and 8-OHdG. Detailed mechanistic studies showed that CFA exerted anti-inflammatory effects by inhibiting TAK1-mediated MAP kinase/NF-κB activation and upregulating AMPK signaling pathways. In addition, CFA exerted antioxidant effects by inhibiting the NADPH oxidase subunits and by increasing the expression of antioxidant enzymes such as HO-1, NQO1, and catalase by upregulating Nrf2 signaling. Finally, we confirmed the effects of CFA on the brains of the LPS-injected mice. CFA inhibited microglial activation and the expression of proinflammatory markers and increased Nrf2-driven antioxidant enzymes. Furthermore, CFA inhibited the production of 4-HNE and 8-OHdG in the brains of LPS-injected mice. As a result, CFA's significant anti-inflammatory and antioxidant properties may have therapeutic applications in neuroinflammatory disorders related with oxidative stress and microglial activation.
Collapse
Affiliation(s)
- Jae-Min Park
- Department of Molecular Medicine, Inflammation-Cancer Microenvironment Research Center, School of Medicine, Ewha Womans University, Seoul, South Korea
| | - Jung-Eun Park
- Department of Molecular Medicine, Inflammation-Cancer Microenvironment Research Center, School of Medicine, Ewha Womans University, Seoul, South Korea
| | - Jin-Sun Park
- Department of Molecular Medicine, Inflammation-Cancer Microenvironment Research Center, School of Medicine, Ewha Womans University, Seoul, South Korea
| | - Yea-Hyun Leem
- Department of Molecular Medicine, Inflammation-Cancer Microenvironment Research Center, School of Medicine, Ewha Womans University, Seoul, South Korea
| | - Do-Yeon Kim
- Department of Molecular Medicine, Inflammation-Cancer Microenvironment Research Center, School of Medicine, Ewha Womans University, Seoul, South Korea
| | - Jin-Won Hyun
- Department of Biochemistry, College of Medicine, and Jeju Research Center for Natural Medicine, Jeju National University, Jeju, South Korea
| | - Hee-Sun Kim
- Department of Molecular Medicine, Inflammation-Cancer Microenvironment Research Center, School of Medicine, Ewha Womans University, Seoul, South Korea.
| |
Collapse
|
23
|
Deng Q, Chen L, Zhang G, Liu L, Luo SM, Gao X. TRIAL-based combination therapies in cancers. Int Immunopharmacol 2024; 138:112570. [PMID: 38971105 DOI: 10.1016/j.intimp.2024.112570] [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: 05/20/2024] [Revised: 06/23/2024] [Accepted: 06/24/2024] [Indexed: 07/08/2024]
Abstract
Tumor necrosis factor (TNF)-related apoptosis-inducing ligand (TRAIL) shows promising therapeutic potential in cancer treatment as it is able to trigger extrinsic apoptotic pathways by binding to the cognate death receptor, causing broad-spectrum apoptosis in cancer cells with negligible toxicity to normal cells. However, the majority of cancers display resistance to TRAIL, limiting its clinical utility. Overcoming resistance to TRAIL therapies remains a challenge in the development of effective anti-cancer strategies. To address the limitations of TRAIL therapy, a viable alternative approach involves combining TRAIL with more potent drugs compared to monotherapy. This combination strategy aims to induce synergistic effects or sensitize drug-resistant cancer cells. This review provides an overview of relevant modalities of TRAIL combination therapy, highlighting different drug classes. The findings demonstrate that combining TRAIL with other agents can effectively counteract resistance observed with TRAIL therapies in cancer. These findings lay a foundation for future advancements in TRAIL-based therapies for treating various cancers.
Collapse
Affiliation(s)
- Qiumin Deng
- MOE Key Laboratory of Tumor Molecular Biology and State Key Laboratory of Bioactive Molecules and Druggability Assessment, Institute of Life and Health Engineering, College of Life Science and Technology, Jinan University, Guangzhou 510632, China
| | - Luxuan Chen
- MOE Key Laboratory of Tumor Molecular Biology and State Key Laboratory of Bioactive Molecules and Druggability Assessment, Institute of Life and Health Engineering, College of Life Science and Technology, Jinan University, Guangzhou 510632, China
| | - Gui Zhang
- MOE Key Laboratory of Tumor Molecular Biology and State Key Laboratory of Bioactive Molecules and Druggability Assessment, Institute of Life and Health Engineering, College of Life Science and Technology, Jinan University, Guangzhou 510632, China
| | - Langxia Liu
- MOE Key Laboratory of Tumor Molecular Biology and State Key Laboratory of Bioactive Molecules and Druggability Assessment, Institute of Life and Health Engineering, College of Life Science and Technology, Jinan University, Guangzhou 510632, China
| | - Shi-Ming Luo
- Guangzhou Key Laboratory of Metabolic Diseases and Reproductive Health, Guangdong-Hong Kong Metabolism & Reproduction Joint Laboratory, Reproductive Medicine Center, Guangdong Second Provincial General Hospital, Guangzhou, Guangdong 510317, China.
| | - Xuejuan Gao
- MOE Key Laboratory of Tumor Molecular Biology and State Key Laboratory of Bioactive Molecules and Druggability Assessment, Institute of Life and Health Engineering, College of Life Science and Technology, Jinan University, Guangzhou 510632, China.
| |
Collapse
|
24
|
Xu Y, Lin F, Liao G, Sun J, Chen W, Zhang L. Ripks and Neuroinflammation. Mol Neurobiol 2024; 61:6771-6787. [PMID: 38349514 DOI: 10.1007/s12035-024-03981-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Accepted: 01/20/2024] [Indexed: 08/22/2024]
Abstract
Neuroinflammation is an immune response in the central nervous system and poses a significant threat to human health. Studies have shown that the receptor serine/threonine protein kinase family (RIPK) family, a popular research target in inflammation, has been shown to play an essential role in neuroinflammation. It is significant to note that the previous reviews have only examined the link between RIPK1 and neuroinflammation. However, it has yet to systematically analyze the relationship between the RIPK family and neuroinflammation. Activation of RIPK1 promotes neuroinflammation. RIPK1 and RIPK3 are responsible for the control of cell death, including apoptosis, necrosis, and inflammation. RIPK1 and RIPK3 regulate inflammatory responses through the release of damage in necroptosis. RIPK1 and RIPK3 regulate inflammatory responses by releasing damage-associated molecular patterns (DAMPs) during necrosis. In addition, activated RIPK1 nuclear translocation and its interaction with the BAF complex leads to upregulation of chromatin modification and inflammatory gene expression, thereby triggering inflammation. Although RIPK2 is not directly involved in regulating cell death, it is considered an essential target for treating neurological inflammation. When the peptidoglycan receptor detects peptidoglycan IE-DAP or MDP in bacteria, it prompts NOD1 and NOD2 to recruit RIPK2 and activate the XIAP E3 ligase. This leads to the K63 ubiquitination of RIPK2. This is followed by LUBAC-mediated linear ubiquitination, which activates NF-KB and MAPK pathways to produce cytokines and chemokines. In conclusion, there are seven known members of the RIPK family, but RIPK4, RIPK5, RIPK6, and RIPK7 have not been linked to neuroinflammation. This article seeks to explore the potential of RIPK1, RIPK2, and RIPK3 kinases as therapeutic interventions for neuroinflammation, which is associated with Alzheimer's disease (AD), amyotrophic lateral sclerosis (ALS), ischemic stroke, Parkinson's disease (PD), multiple sclerosis (MS), and traumatic brain injury (TBI).
Collapse
Affiliation(s)
- Yue Xu
- Department of Cerebrovascular Disease, Sun Yat-Sen University, The Fifth Affiliated Hospital, Zhuhai, 519000, Guangdong, People's Republic of China
| | - Feng Lin
- Department of Cerebrovascular Disease, Sun Yat-Sen University, The Fifth Affiliated Hospital, Zhuhai, 519000, Guangdong, People's Republic of China
| | - Guolei Liao
- Department of Cerebrovascular Disease, Sun Yat-Sen University, The Fifth Affiliated Hospital, Zhuhai, 519000, Guangdong, People's Republic of China
| | - Jiaxing Sun
- Department of Cerebrovascular Disease, Sun Yat-Sen University, The Fifth Affiliated Hospital, Zhuhai, 519000, Guangdong, People's Republic of China
| | - Wenli Chen
- Department of Pharmacy, Sun Yat-Sen University, The Fifth Affiliated Hospital, Zhuhai, 519000, Guangdong, People's Republic of China.
| | - Lei Zhang
- Department of Cerebrovascular Disease, Sun Yat-Sen University, The Fifth Affiliated Hospital, Zhuhai, 519000, Guangdong, People's Republic of China.
| |
Collapse
|
25
|
Peng L. Necroptosis and autoimmunity. Clin Immunol 2024; 266:110313. [PMID: 39002793 DOI: 10.1016/j.clim.2024.110313] [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: 06/17/2024] [Accepted: 07/10/2024] [Indexed: 07/15/2024]
Abstract
Autoimmunity is a normal physiological state that requires immunological homeostasis and surveillance, whereas necroptosis is a type of inflammatory cell death. When necroptosis occurs, various immune system cells must perform their appropriate duties to preserve immunological homeostasis, whether the consequence is expanding or limiting the inflammatory response and the pathological condition is cleared or progresses to the autoimmune disease stage. This article discusses necroptosis based on RIP homotypic interaction motif (RHIM) interaction under various physiological and pathological situations, with the RIPK1-RIPK3-MLKL necrosome serving as the regulatory core. In addition, the cell biology of necroptosis involved in autoimmunity and its application in autoimmune diseases were also reviewed.
Collapse
Affiliation(s)
- Lin Peng
- National Clinical Research Center for Kidney Disease, Affiliated Jinling Hospital, Medical School of Nanjing University, Zhongshan East Road No.305, Nanjing, Jiangsu 210002, China.
| |
Collapse
|
26
|
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
|
27
|
Jetton D, Muendlein HI, Connolly WM, Magri Z, Smirnova I, Batorsky R, Mecsas J, Degterev A, Poltorak A. Non-canonical autophosphorylation of RIPK1 drives timely pyroptosis to control Yersinia infection. Cell Rep 2024; 43:114641. [PMID: 39154339 PMCID: PMC11465231 DOI: 10.1016/j.celrep.2024.114641] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Revised: 06/17/2024] [Accepted: 07/31/2024] [Indexed: 08/20/2024] Open
Abstract
Caspase-8-dependent pyroptosis has been shown to mediate host protection from Yersinia infection. For this mode of cell death, the kinase activity of receptor-interacting protein kinase 1 (RIPK1) is required, but the autophosphorylation sites required to drive caspase-8 activation have not been determined. Here, we show that non-canonical autophosphorylation of RIPK1 at threonine 169 (T169) is necessary for caspase-8-mediated pyroptosis. Mice with alanine in the T169 position are highly susceptible to Yersinia dissemination. Mechanistically, the delayed formation of a complex containing RIPK1, ZBP1, Fas-associated protein with death domain (FADD), and caspase-8 abrogates caspase-8 maturation in T169A mice and leads to the eventual activation of RIPK3-dependent necroptosis in vivo; however, this is insufficient to protect the host, suggesting that timely pyroptosis during early response is specifically required to control infection. These results position RIPK1 T169 phosphorylation as a driver of pyroptotic cell death critical for host defense.
Collapse
Affiliation(s)
- David Jetton
- Graduate Program in Immunology, Tufts Graduate School of Biomedical Sciences, Boston, MA 02111, USA
| | - Hayley I Muendlein
- Department of Immunology, Tufts University School of Medicine, Boston, MA 02111, USA
| | - Wilson M Connolly
- Department of Immunology, Tufts University School of Medicine, Boston, MA 02111, USA
| | - Zoie Magri
- Graduate Program in Immunology, Tufts Graduate School of Biomedical Sciences, Boston, MA 02111, USA
| | - Irina Smirnova
- Department of Immunology, Tufts University School of Medicine, Boston, MA 02111, USA
| | - Rebecca Batorsky
- Data Intensive Studies Center, Tufts University, Medford, MA 02155, USA
| | - Joan Mecsas
- Department of Molecular Biology and Microbiology, Tufts University School of Medicine, Boston, MA 02111, USA
| | - Alexei Degterev
- Department of Cell, Molecular & Developmental Biology, Tufts University School of Medicine, Boston, MA 02111, USA
| | - Alexander Poltorak
- Graduate Program in Immunology, Tufts Graduate School of Biomedical Sciences, Boston, MA 02111, USA; Department of Immunology, Tufts University School of Medicine, Boston, MA 02111, USA.
| |
Collapse
|
28
|
Liu J, Wu XL, Zhang J, Li B, Wang HY, Wang J, Lu JX. The structure of mouse RIPK1 RHIM-containing domain as a homo-amyloid and in RIPK1/RIPK3 complex. Nat Commun 2024; 15:6975. [PMID: 39143113 PMCID: PMC11325021 DOI: 10.1038/s41467-024-51303-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Accepted: 08/05/2024] [Indexed: 08/16/2024] Open
Abstract
Receptor-interacting protein kinase 1 (RIPK1) is a therapeutic target in treating neurodegenerative diseases and cancers. RIPK1 has three distinct functional domains, with the center domain containing a receptor-interacting protein homotypic interaction motif (RHIM), which mediates amyloid formation. The functional amyloid formed by RIPK1 and/or RIPK3 is a crucial intermediate in regulating cell necroptosis. In this study, the amyloid structure of mouse RIPK1, formed by an 82-residue sequence centered at RHIM, is presented. It reveals the "N"-shaped folding of the protein subunit in the fibril with four β-strands. The folding pattern is shared by several amyloid structures formed by proteins with RHIM, with the central β-strand formed by the most conserved tetrad sequence I/VQI/VG. However, the solid-state NMR results indicate a structural difference between mouse RIPK1 and mouse RIPK3. A change in the structural rigidity is also suggested by the observation of weakened signals for mouse RIPK3 upon mixing with RIPK1 to form the RIPK1/RIPK3 complex fibrils. Our results provide vital information to understand the interactions between different proteins with RHIM, which will help us further comprehend the regulation mechanism in cell necroptosis.
Collapse
Affiliation(s)
- Jing Liu
- School of Life Science and Technology, ShanghaiTech University, Shanghai, 201210, China
- Interdisciplinary Institute of NMR and Molecular Sciences, School of Chemistry and Chemical Engineering, The State Key Laboratory of Refractories and Metallurgy, Wuhan University of Science and Technology, Wuhan, 430081, China
| | - Xia-Lian Wu
- Interdisciplinary Institute of NMR and Molecular Sciences, School of Chemistry and Chemical Engineering, The State Key Laboratory of Refractories and Metallurgy, Wuhan University of Science and Technology, Wuhan, 430081, China
| | - Jing Zhang
- School of Life Science and Technology, ShanghaiTech University, Shanghai, 201210, China
| | - Bing Li
- School of Life Science and Technology, ShanghaiTech University, Shanghai, 201210, China
| | | | - Jian Wang
- School of Life Science and Technology, ShanghaiTech University, Shanghai, 201210, China.
| | - Jun-Xia Lu
- School of Life Science and Technology, ShanghaiTech University, Shanghai, 201210, China.
- Interdisciplinary Institute of NMR and Molecular Sciences, School of Chemistry and Chemical Engineering, The State Key Laboratory of Refractories and Metallurgy, Wuhan University of Science and Technology, Wuhan, 430081, China.
| |
Collapse
|
29
|
Yadav S, El Hamra R, Alturki NA, Ariana A, Bhan A, Hurley K, Gaestel M, Blackshear PJ, Blais A, Sad S. Regulation of Zfp36 by ISGF3 and MK2 restricts the expression of inflammatory cytokines during necroptosis stimulation. Cell Death Dis 2024; 15:574. [PMID: 39117638 PMCID: PMC11310327 DOI: 10.1038/s41419-024-06964-4] [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: 01/22/2024] [Revised: 07/29/2024] [Accepted: 07/31/2024] [Indexed: 08/10/2024]
Abstract
Necrosome activation following TLR- or cytokine receptor-signaling results in cell death by necroptosis which is characterized by the rupture of cell membranes and the consequent release of intracellular contents to the extracellular milieu. While necroptosis exacerbates various inflammatory diseases, the mechanisms through which the inflammatory responses are regulated are not clear. We show that the necrosome activation of macrophages results in an upregulation of various pathways, including the mitogen-activated protein kinase (MAPK) cascade, which results in an elevation of the inflammatory response and consequent expression of several cytokines and chemokines. Programming for this upregulation of inflammatory response occurs during the early phase of necrosome activation and proceeds independently of cell death but depends on the activation of the receptor-interacting protein kinase-1 (RipK1). Interestingly, necrosome activation also results in an upregulation of IFNβ, which in turn exerts an inhibitory effect on the maintenance of inflammatory response through the repression of MAPK-signaling and an upregulation of Zfp36. Activation of the interferon-induced gene factor-3 (ISGF3) results in the expression of ZFP36 (TTP), which induces the post-transcriptional degradation of mRNAs of various inflammatory cytokines and chemokines through the recognition of AU-rich elements in their 3'UTR. Furthermore, ZFP-36 inhibits IFNβ-, but not TNFα- induced necroptosis. Overall, these results reveal the molecular mechanism through which IFNβ, a pro-inflammatory cytokine, induces the expression of ZFP-36, which in turn inhibits necroptosis and halts the maintenance of the inflammatory response.
Collapse
Affiliation(s)
- Sahil Yadav
- Department of Biochemistry, Microbiology, and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada
| | - Rayan El Hamra
- Department of Biochemistry, Microbiology, and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada
| | - Norah A Alturki
- Clinical Laboratory Science Department, College of Applied Medical Sciences, King Saud University, Riyadh, Saudi Arabia
| | - Ardeshir Ariana
- Department of Biochemistry, Microbiology, and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada
| | - Avni Bhan
- Department of Biochemistry, Microbiology, and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada
| | - Kate Hurley
- Department of Biochemistry, Microbiology, and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada
| | - Matthias Gaestel
- Institute of Cell Biochemistry, Hannover Medical School, Hannover, Germany
| | - Perry J Blackshear
- Signal Transduction Laboratory, National Institute of Environmental Health Sciences, Research Triangle Park, Durham, North Carolina, United States of America
| | - Alexandre Blais
- Department of Biochemistry, Microbiology, and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada
- Ottawa Institute of Systems Biology, Ottawa, ON, Canada
- University of Ottawa, Centre for Infection Immunity and Inflammation, Ottawa, ON, Canada
- University of Ottawa Brain and Mind Research Institute, Ottawa, ON, Canada
| | - Subash Sad
- Department of Biochemistry, Microbiology, and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada.
- University of Ottawa, Centre for Infection Immunity and Inflammation, Ottawa, ON, Canada.
| |
Collapse
|
30
|
Dimopoulou C, Guerra PR, Mortensen MS, Kristensen KA, Pedersen M, Bahl MI, Sommer MAO, Licht TR, Laursen MF. Potential of using an engineered indole lactic acid producing Escherichia coli Nissle 1917 in a murine model of colitis. Sci Rep 2024; 14:17542. [PMID: 39080343 PMCID: PMC11289411 DOI: 10.1038/s41598-024-68412-9] [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: 10/05/2023] [Accepted: 07/23/2024] [Indexed: 08/02/2024] Open
Abstract
The gut microbiome is a significant factor in the pathophysiology of ulcerative colitis (UC), prompting investigations into the use of probiotic therapies to counter gastrointestinal inflammation. However, while much attention has been given to the therapeutic potential of microbes at the species and strain level, the discovery and application of their metabolic products may offer more precise and controlled solutions in battling disease. In this work, we examined the therapeutic potential of indole lactic acid (ILA) to alleviate inflammation in a murine model of colitis. A previously constructed ILA-producing Escherichia coli Nissle 1917 strain (EcN aldh) and its isogenic non-ILA producing counterpart (EcN) were studied in a murine model of Dextran Sodium Sulfate (DSS) induced colitis. The colitic animals suffered from severe colitic symptoms, with no differentiation between the groups in body weight loss and disease activity index. However, three days after cessation of DSS treatment the EcN aldh-treated mice showed signs of reduced intestinal inflammation, as manifested by lower concentrations of fecal lipocalin-2. Additionally, expression analysis of the inflamed tissue revealed distinct effects of the EcN aldh strain on proteins associated with intestinal health, such as TFF3, occludin and IL-1β expression. These results show no impact of EcN or EcN aldh on acute DSS-induced colitis, but suggest that in particular EcN aldh may assist recovery from intestinal inflammation.
Collapse
Affiliation(s)
| | | | | | | | - Mikael Pedersen
- National Food Institute, Technical University of Denmark, Kgs. Lyngby, Denmark
| | - Martin Iain Bahl
- National Food Institute, Technical University of Denmark, Kgs. Lyngby, Denmark
| | | | - Tine Rask Licht
- National Food Institute, Technical University of Denmark, Kgs. Lyngby, Denmark
| | | |
Collapse
|
31
|
Zhang Z, Yang Z, Wang S, Wang X, Mao J. Decoding ferroptosis: Revealing the hidden assassin behind cardiovascular diseases. Biomed Pharmacother 2024; 176:116761. [PMID: 38788596 DOI: 10.1016/j.biopha.2024.116761] [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: 03/25/2024] [Revised: 05/09/2024] [Accepted: 05/17/2024] [Indexed: 05/26/2024] Open
Abstract
The discovery of regulatory cell death processes has driven innovation in cardiovascular disease (CVD) therapeutic strategies. Over the past decade, ferroptosis, an iron-dependent form of regulated cell death driven by excessive lipid peroxidation, has been shown to drive the development of multiple CVDs. This review provides insights into the evolution of the concept of ferroptosis, the similarities and differences with traditional modes of programmed cell death (e.g., apoptosis, autophagy, and necrosis), as well as the core regulatory mechanisms of ferroptosis (including cystine/glutamate transporter blockade, imbalance of iron metabolism, and lipid peroxidation). In addition, it provides not only a detailed review of the role of ferroptosis and its therapeutic potential in widely studied CVDs such as coronary atherosclerotic heart disease, myocardial infarction, myocardial ischemia/reperfusion injury, heart failure, cardiomyopathy, and aortic aneurysm but also an overview of the phenomenon and therapeutic perspectives of ferroptosis in lesser-addressed CVDs such as cardiac valvulopathy, pulmonary hypertension, and sickle cell disease. This article aims to integrate this knowledge to provide a comprehensive view of ferroptosis in a wide range of CVDs and to drive innovation and progress in therapeutic strategies in this field.
Collapse
Affiliation(s)
- Zeyu Zhang
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin 300381, China; Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Zhihua Yang
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin 300381, China; Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Shuai Wang
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin 300381, China
| | - Xianliang Wang
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin 300381, China.
| | - Jingyuan Mao
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin 300381, China.
| |
Collapse
|
32
|
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
|
33
|
Peng L, Wang P, Xu X, Chen D, Xu F, Yang F, Yang S, Xia H, Liu ZH, Qin W. Inhibition of receptor interacting protein kinase-1 (RIPK1) in the treatment of murine lupus. Lupus Sci Med 2024; 11:e001146. [PMID: 38906550 PMCID: PMC11191810 DOI: 10.1136/lupus-2024-001146] [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: 01/08/2024] [Accepted: 06/07/2024] [Indexed: 06/23/2024]
Abstract
OBJECTIVE Systemic lupus erythematosus (SLE) is a type of autoimmune disease that involves multiple organs involved as well as cytokine dysregulation. The treatment of SLE is still challenging due to the side effects of the different drugs used. Receptor-interacting protein kinase 1 (RIPK1) is a kinase involved in T cell homeostasis and autoinflammation. Although clinical trials have shown that RIPK1 inhibition exhibits significant efficacy in different autoimmune diseases, its role in SLE remains unclear. METHODS MRL/lpr lupus-prone mice received RIPK1 inhibitor ZJU37 or vehicle intraperitoneally for 10 weeks. A BM12-induced chronic graft-versus-host-disease (cGVHD) lupus-like model was introduced in RIPK1 D138N mice or C57BL/6 mice. Nephritis, serum autoantibody levels, dysregulation of adaptive immune response and cytokines were compared in treated and untreated mice. RESULTS ZJU37 alleviated the clinical features of the MRL/lpr mice including nephritis and anti-dsDNA antibody production. In addition, ZJU37 treatment reduced the proportion of double-negative T cells in the spleen and the cytokines of TNFα, IFN-γ, IL-6, IL-17 and IL-1β in the serum. Moreover, RIPK1 D138N mice were able to prevent the cGVHD lupus-like model from SLE attack, manifesting as anti-dsDNA antibody production, the proliferation of germinal centre B cells, plasma cells, and T follicular helper cells as well as IgG and C3 deposits in kidneys. CONCLUSION RIPK1 inhibition has a protective effect in the mouse model of SLE and can potentially become a new therapeutic target for SLE in humans.
Collapse
Affiliation(s)
- Lin Peng
- National Clinical Research Center for Kidney Disease, Affiliated Jinling Hospital, Medical School of Nanjing University, Nanjing, Jiangsu, China
| | - Pengcheng Wang
- National Clinical Research Center for Kidney Disease, Affiliated Jinling Hospital, Medical School of Nanjing University, Nanjing, Jiangsu, China
| | - Xiaodong Xu
- National Clinical Research Center for Kidney Disease, Affiliated Jinling Hospital, Medical School of Nanjing University, Nanjing, Jiangsu, China
| | - Dacheng Chen
- National Clinical Research Center for Kidney Disease, Affiliated Jinling Hospital, Medical School of Nanjing University, Nanjing, Jiangsu, China
| | - Feng Xu
- National Clinical Research Center for Kidney Disease, Affiliated Jinling Hospital, Medical School of Nanjing University, Nanjing, Jiangsu, China
| | - Fan Yang
- National Clinical Research Center for Kidney Disease, Affiliated Jinling Hospital, Medical School of Nanjing University, Nanjing, Jiangsu, China
| | - Shuying Yang
- Department of Biochemistry and Molecular Medical Center, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Hongguang Xia
- Department of Biochemistry and Molecular Medical Center, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Zhi-Hong Liu
- National Clinical Research Center for Kidney Disease, Affiliated Jinling Hospital, Medical School of Nanjing University, Nanjing, Jiangsu, China
| | - Weisong Qin
- National Clinical Research Center for Kidney Disease, Affiliated Jinling Hospital, Medical School of Nanjing University, Nanjing, Jiangsu, China
| |
Collapse
|
34
|
Bozgeyik E, Elek A, Gocer Z, Bozgeyik I. The fate and function of non-coding RNAs during necroptosis. Epigenomics 2024; 16:901-915. [PMID: 38884366 PMCID: PMC11370912 DOI: 10.1080/17501911.2024.2354653] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Accepted: 05/07/2024] [Indexed: 06/18/2024] Open
Abstract
Necroptosis is a novel form of cell death which is activated when apoptotic cell death signals are disrupted. Accumulating body of observations suggests that noncoding RNAs, which are the lately discovered mystery of the human genome, are significantly associated with necroptotic signaling circuitry. The fate and function of miRNAs have been well documented in human disease, especially cancer. Recently, lncRNAs have gained much attention due to their diverse regulatory functions. Although available studies are currently based on bioinformatic analysis, predicted interactions desires further attention, as these hold significant promise and should not be overlooked. In the light of these, here we comprehensively review and discuss noncoding RNA molecules that play significant roles during execution of necroptotic cell death.
Collapse
Affiliation(s)
- Esra Bozgeyik
- Department of Medical Services & Techniques, Vocational School of Health Services, Adiyaman University, Adiyaman, Turkey
| | - Alperen Elek
- Faculty of Medicine, Ege University, Izmir, Turkey
| | - Zekihan Gocer
- Faculty of Medicine, Gaziantep University, Gaziantep, Turkey
| | - Ibrahim Bozgeyik
- Department of Medical Biology, Faculty of Medicine, Adiyaman University, Adiyaman, Turkey
| |
Collapse
|
35
|
Zhang C, Ma J, Zhang X, Zhou D, Cao Z, Qiao L, Chen G, Yang L, Ding BS. Processing of angiocrine alarmin IL-1α in endothelial cells promotes lung and liver fibrosis. Int Immunopharmacol 2024; 134:112176. [PMID: 38723369 DOI: 10.1016/j.intimp.2024.112176] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2024] [Revised: 04/21/2024] [Accepted: 04/27/2024] [Indexed: 06/03/2024]
Abstract
BACKGROUND Fibrosis results from excessive scar formation after tissue injury. Injured cells release alarmins such as interleukin 1 (IL-1) α and β as primary mediators initiating tissue repair. However, how alarmins from different cell types differentially regulate fibrosis remains to be explored. METHODS Here, we used tissue specific knockout strategy to illustrate a unique contribution of endothelial cell-derived IL-1α to lung and liver fibrosis. The two fibrotic animal model triggered by bleomycin and CCl4 were used to study the effects of endothelial paracrine/angiocrine IL-1α in fibrotic progression. Human umbilical vein endothelial cells (HUVEC) were performed to explore the production of angiocrine IL-1α at both transcriptional and post-transcriptional levels in vitro. RESULTS We found that endothelial paracrine/angiocrine IL-1α primarily promotes lung and liver fibrosis during the early phase of organ repair. By contrast, myeloid cell-specific ablation of IL-1α in mice resulted in little influence on fibrosis, suggesting the specific pro-fibrotic role of IL-1α from endothelial cell but not macrophage. In vitro study revealed a coordinated regulation of IL-1α production in human primary endothelial cells at both transcriptional and post-transcriptional levels. Specifically, the transcription of IL-1α is regulated by RIPK1, and after caspase-8 (CASP8) cleaves the precursor form of IL-1α, its secretion is triggered by ion channel Pannexin 1 upon CASP8 cleavage. CONCLUSIONS Endothelial cell-produced IL-1α plays a unique role in promoting organ fibrosis. Furthermore, the release of this angiocrine alarmin relies on a unique molecular mechanism involving RIPK1, CASP8, and ion channel Pannexin 1.
Collapse
Affiliation(s)
- Chunxue Zhang
- Key Laboratory of Birth Defects of MOE, State Key Laboratory of Biotherapy, West China Second Hospital, College of Life Sciences, Sichuan University, Chengdu 610041, China
| | - Jie Ma
- Key Laboratory of Birth Defects of MOE, State Key Laboratory of Biotherapy, West China Second Hospital, College of Life Sciences, Sichuan University, Chengdu 610041, China
| | - Xu Zhang
- Department of Pathophysiology, Harbin Medical University, Harbin 150081, China
| | - Dengcheng Zhou
- Key Laboratory of Birth Defects of MOE, State Key Laboratory of Biotherapy, West China Second Hospital, College of Life Sciences, Sichuan University, Chengdu 610041, China
| | - Zhongwei Cao
- Key Laboratory of Birth Defects of MOE, State Key Laboratory of Biotherapy, West China Second Hospital, College of Life Sciences, Sichuan University, Chengdu 610041, China
| | - Lina Qiao
- Key Laboratory of Birth Defects of MOE, State Key Laboratory of Biotherapy, West China Second Hospital, College of Life Sciences, Sichuan University, Chengdu 610041, China.
| | - Guo Chen
- Department of Anesthesiology, The Research Units of West China(2018RU012)-Chinese Academy of Medical Sciences, West China Hospital, Sichuan University, China.
| | - Liming Yang
- Department of Pathophysiology, Harbin Medical University, Harbin 150081, China.
| | - Bi-Sen Ding
- Key Laboratory of Birth Defects of MOE, State Key Laboratory of Biotherapy, West China Second Hospital, College of Life Sciences, Sichuan University, Chengdu 610041, China.
| |
Collapse
|
36
|
Yu X, Lin H, Li F, Wang J, Lu D. Development of Biochemical and Cellular Probes to Study RIPK1 Target Engagement. ACS Med Chem Lett 2024; 15:906-916. [PMID: 38894934 PMCID: PMC11181498 DOI: 10.1021/acsmedchemlett.4c00104] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2024] [Revised: 05/07/2024] [Accepted: 05/09/2024] [Indexed: 06/21/2024] Open
Abstract
RIPK1 inhibitors have emerged as promising candidates for treating diverse diseases, including inflammatory diseases, autoimmune disorders, Alzheimer's disease, and cancer. However, the previously reported binding assays have limited sensitivity and stability, impeding high-throughput screening and robust characterization of the RIPK1 inhibitors. To address this challenge, we introduced two probes, T2-BDP-FL and T3-BDP-FL, derived from distinct RIPK1 inhibitors with different binding modes to establish time-resolved fluorescence resonance energy transfer (TR-FRET) displacement assays. Employing our TR-FRET displacement assays, we quantified the biochemical binding affinities of a series of RIPK1 inhibitors with diverse structural and binding modes for human RIPK1. Consistent results were obtained with these two probes in the TR-FRET displacement assay. Furthermore, we developed a RIPK1 fluorescent probe, T2-BDP589, for the NanoBRET assay. This assay enabled the characterization of RIPK1 target engagement by various RIPK1 inhibitors for both human and mouse RIPK1 in live cells. Our developed fluorescent probe displacement assays offer a sensitive and high-throughput approach to identify RIPK1 inhibitors based on both biochemical and cellular activities.
Collapse
Affiliation(s)
- Xin Yu
- Department
of Biochemistry and Molecular Pharmacology, Baylor College of Medicine, Houston, Texas 77030, United States
| | - Hanfeng Lin
- Department
of Biochemistry and Molecular Pharmacology, Baylor College of Medicine, Houston, Texas 77030, United States
| | - Feng Li
- Center
for Drug Discovery, Baylor College of Medicine, Houston, Texas 77030, United States
| | - Jin Wang
- Department
of Biochemistry and Molecular Pharmacology, Baylor College of Medicine, Houston, Texas 77030, United States
- Department
of Molecular and Cellular Biology, Baylor
College of Medicine, Houston, Texas 77030, United States
| | - Dong Lu
- Department
of Biochemistry and Molecular Pharmacology, Baylor College of Medicine, Houston, Texas 77030, United States
| |
Collapse
|
37
|
Wu L, Chang E, Zhao H, Ma D. Regulated cell death in hypoxic-ischaemic encephalopathy: recent development and mechanistic overview. Cell Death Discov 2024; 10:277. [PMID: 38862503 PMCID: PMC11167026 DOI: 10.1038/s41420-024-02014-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Revised: 05/05/2024] [Accepted: 05/07/2024] [Indexed: 06/13/2024] Open
Abstract
Hypoxic-ischaemic encephalopathy (HIE) in termed infants remains a significant cause of morbidity and mortality worldwide despite the introduction of therapeutic hypothermia. Depending on the cell type, cellular context, metabolic predisposition and insult severity, cell death in the injured immature brain can be highly heterogenous. A continuum of cell death exists in the H/I-injured immature brain. Aside from apoptosis, emerging evidence supports the pathological activation of necroptosis, pyroptosis and ferroptosis as alternative regulated cell death (RCD) in HIE to trigger neuroinflammation and metabolic disturbances in addition to cell loss. Upregulation of autophagy and mitophagy in HIE represents an intrinsic neuroprotective strategy. Molecular crosstalk between RCD pathways implies one RCD mechanism may compensate for the loss of function of another. Moreover, mitochondrion was identified as the signalling "hub" where different RCD pathways converge. The highly-orchestrated nature of RCD makes them promising therapeutic targets. Better understanding of RCD mechanisms and crosstalk between RCD subtypes likely shed light on novel therapy development for HIE. The identification of a potential RCD converging node may open up the opportunity for simultaneous and synergistic inhibition of cell death in the immature brain.
Collapse
Affiliation(s)
- Lingzhi Wu
- Division of Anaesthetics, Pain Medicine and Intensive Care, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, Chelsea and Westminster Hospital, London, UK
| | - Enqiang Chang
- Division of Anaesthetics, Pain Medicine and Intensive Care, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, Chelsea and Westminster Hospital, London, UK
| | - Hailin Zhao
- Division of Anaesthetics, Pain Medicine and Intensive Care, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, Chelsea and Westminster Hospital, London, UK
| | - Daqing Ma
- Division of Anaesthetics, Pain Medicine and Intensive Care, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, Chelsea and Westminster Hospital, London, UK.
- Perioperative and Systems Medicine Laboratory, The Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, 310052, China.
| |
Collapse
|
38
|
Song Q, Fan Y, Zhang H, Wang N. Z-DNA binding protein 1 orchestrates innate immunity and inflammatory cell death. Cytokine Growth Factor Rev 2024; 77:15-29. [PMID: 38548490 DOI: 10.1016/j.cytogfr.2024.03.005] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Revised: 03/17/2024] [Accepted: 03/20/2024] [Indexed: 06/22/2024]
Abstract
Innate immunity is not only the first line of host defense against microbial infections but is also crucial for the host responses against a variety of noxious stimuli. Z-DNA binding protein 1 (ZBP1) is a cytosolic nucleic acid sensor that can induce inflammatory cell death in both immune and nonimmune cells upon sensing of incursive virus-derived Z-form nucleic acids and self-nucleic acids via its Zα domain. Mechanistically, aberrantly expressed or activated ZBP1 induced by pathogens or noxious stimuli enables recruitment of TANK binding kinase 1 (TBK1), interferon regulatory factor 3 (IRF3), receptor-interacting serine/threonine-protein kinase 1 (RIPK1) and RIPK3 to drive type I interferon (IFN-I) responses and activation of nuclear factor kappa B (NF-κB) signaling. Meanwhile, ZBP1 promotes the assembly of ZBP1- and absent in melanoma 2 (AIM2)-PANoptosome, which ultimately triggers PANoptosis through caspase 3-mediated apoptosis, mixed lineage kinase domain like pseudokinase (MLKL)-mediated necroptosis, and gasdermin D (GSDMD)-mediated pyroptosis. In response to damaged mitochondrial DNA, ZBP1 can interact with cyclic GMP-AMP synthase to augment IFN-I responses but inhibits toll like receptor 9-mediated inflammatory responses. This review summarizes the structure and expression pattern of ZBP1, discusses its roles in human diseases through immune-dependent (e.g., the production of IFN-I and pro-inflammatory cytokines) and -independent (e.g., the activation of cell death) functions, and highlights the attractive prospect of manipulating ZBP1 as a promising therapeutic target in diseases.
Collapse
Affiliation(s)
- Qixiang Song
- Department of Pathophysiology, School of Basic Medical Science, Central South University, 110 Xiangya Road, Changsha 410083, China; Key Laboratory of Sepsis Translational Medicine of Hunan, Central South University, 110 Xiangya Road, Changsha 410083, China
| | - Yuhang Fan
- Department of Pathophysiology, School of Basic Medical Science, Central South University, 110 Xiangya Road, Changsha 410083, China; Key Laboratory of Sepsis Translational Medicine of Hunan, Central South University, 110 Xiangya Road, Changsha 410083, China
| | - Huali Zhang
- Department of Pathophysiology, School of Basic Medical Science, Central South University, 110 Xiangya Road, Changsha 410083, China; Key Laboratory of Sepsis Translational Medicine of Hunan, Central South University, 110 Xiangya Road, Changsha 410083, China.
| | - Nian Wang
- Department of Pathophysiology, School of Basic Medical Science, Central South University, 110 Xiangya Road, Changsha 410083, China; Key Laboratory of Sepsis Translational Medicine of Hunan, Central South University, 110 Xiangya Road, Changsha 410083, China.
| |
Collapse
|
39
|
Mago E, Zhao X, Zhang W, Shao Q, Li P, Huang S, Ding X, Liu H, Sun T, He F, Weng D. RIP1 kinase inactivation protects against LPS-induced acute respiratory distress syndrome in mice. Int Immunopharmacol 2024; 133:112060. [PMID: 38652970 DOI: 10.1016/j.intimp.2024.112060] [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: 12/26/2023] [Revised: 03/22/2024] [Accepted: 04/08/2024] [Indexed: 04/25/2024]
Abstract
Acute respiratory distress syndrome (ARDS) is characterized by lung tissue oedema and inflammatory cell infiltration, with limited therapeutic interventions available. Receptor-interacting protein kinase 1 (RIPK1), a critical regulator of cell death and inflammation implicated in many diseases, is not fully understood in the context of ARDS. In this study, we employed RIP1 kinase-inactivated (Rip1K45A/K45A) mice and two distinct RIPK1 inhibitors to investigate the contributions of RIP1 kinase activity in lipopolysaccharide (LPS)-induced ARDS pathology. Our results indicated that RIPK1 kinase inactivation, achieved through both genetic and chemical approaches, significantly attenuated LPS-induced ARDS pathology, as demonstrated by reduced polymorphonuclear neutrophil percentage (PMN%) in alveolar lavage fluid, expression of inflammatory and fibrosis-related factors in lung tissues, as well as histological examination. Results by tunnel staining and qRT-PCR analysis indicated that RIPK1 kinase activity played a role in regulating cell apoptosis and inflammation induced by LPS administration in lung tissue. In summary, employing both pharmacological and genetic approaches, this study demonstrated that targeted RIPK1 kinase inactivation attenuates the pathological phenotype induced by LPS inhalation in an ARDS mouse model. This study enhances our understanding of the therapeutic potential of RIPK1 kinase modulation in ARDS, providing insights for the pathogenesis of ARDS.
Collapse
Affiliation(s)
- Emmauel Mago
- School of Environmental and Biological Engineering, Nanjing University of Science & Technology, 200 Xiaolingwei Street, Nanjing 210094, China
| | - Xunan Zhao
- School of Environmental and Biological Engineering, Nanjing University of Science & Technology, 200 Xiaolingwei Street, Nanjing 210094, China
| | - Weigao Zhang
- School of Environmental and Biological Engineering, Nanjing University of Science & Technology, 200 Xiaolingwei Street, Nanjing 210094, China
| | - Qianchao Shao
- School of Environmental and Biological Engineering, Nanjing University of Science & Technology, 200 Xiaolingwei Street, Nanjing 210094, China
| | - Peiqi Li
- School of Environmental and Biological Engineering, Nanjing University of Science & Technology, 200 Xiaolingwei Street, Nanjing 210094, China
| | - Shuxian Huang
- School of Environmental and Biological Engineering, Nanjing University of Science & Technology, 200 Xiaolingwei Street, Nanjing 210094, China
| | - Xinyu Ding
- School of Environmental and Biological Engineering, Nanjing University of Science & Technology, 200 Xiaolingwei Street, Nanjing 210094, China
| | - Hu Liu
- School of Environmental and Biological Engineering, Nanjing University of Science & Technology, 200 Xiaolingwei Street, Nanjing 210094, China
| | - Tingzhe Sun
- School of Life Sciences, Anqing Normal University, Anqing 246133, Anhui, China
| | - Fei He
- Department of Emergency Medicine, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing 210008, China.
| | - Dan Weng
- School of Environmental and Biological Engineering, Nanjing University of Science & Technology, 200 Xiaolingwei Street, Nanjing 210094, China.
| |
Collapse
|
40
|
Zhou Y, Tao L, Qiu J, Xu J, Yang X, Zhang Y, Tian X, Guan X, Cen X, Zhao Y. Tumor biomarkers for diagnosis, prognosis and targeted therapy. Signal Transduct Target Ther 2024; 9:132. [PMID: 38763973 PMCID: PMC11102923 DOI: 10.1038/s41392-024-01823-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Revised: 03/07/2024] [Accepted: 04/02/2024] [Indexed: 05/21/2024] Open
Abstract
Tumor biomarkers, the substances which are produced by tumors or the body's responses to tumors during tumorigenesis and progression, have been demonstrated to possess critical and encouraging value in screening and early diagnosis, prognosis prediction, recurrence detection, and therapeutic efficacy monitoring of cancers. Over the past decades, continuous progress has been made in exploring and discovering novel, sensitive, specific, and accurate tumor biomarkers, which has significantly promoted personalized medicine and improved the outcomes of cancer patients, especially advances in molecular biology technologies developed for the detection of tumor biomarkers. Herein, we summarize the discovery and development of tumor biomarkers, including the history of tumor biomarkers, the conventional and innovative technologies used for biomarker discovery and detection, the classification of tumor biomarkers based on tissue origins, and the application of tumor biomarkers in clinical cancer management. In particular, we highlight the recent advancements in biomarker-based anticancer-targeted therapies which are emerging as breakthroughs and promising cancer therapeutic strategies. We also discuss limitations and challenges that need to be addressed and provide insights and perspectives to turn challenges into opportunities in this field. Collectively, the discovery and application of multiple tumor biomarkers emphasized in this review may provide guidance on improved precision medicine, broaden horizons in future research directions, and expedite the clinical classification of cancer patients according to their molecular biomarkers rather than organs of origin.
Collapse
Affiliation(s)
- Yue Zhou
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Lei Tao
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Jiahao Qiu
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Jing Xu
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Xinyu Yang
- West China School of Pharmacy, Sichuan University, Chengdu, 610041, China
| | - Yu Zhang
- West China School of Pharmacy, Sichuan University, Chengdu, 610041, China
- School of Medicine, Tibet University, Lhasa, 850000, China
| | - Xinyu Tian
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Xinqi Guan
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Xiaobo Cen
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
- National Chengdu Center for Safety Evaluation of Drugs, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Yinglan Zhao
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China.
| |
Collapse
|
41
|
De Meyer GRY, Zurek M, Puylaert P, Martinet W. Programmed death of macrophages in atherosclerosis: mechanisms and therapeutic targets. Nat Rev Cardiol 2024; 21:312-325. [PMID: 38163815 DOI: 10.1038/s41569-023-00957-0] [Citation(s) in RCA: 15] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 11/02/2023] [Indexed: 01/03/2024]
Abstract
Atherosclerosis is a progressive inflammatory disorder of the arterial vessel wall characterized by substantial infiltration of macrophages, which exert both favourable and detrimental functions. Early in atherogenesis, macrophages can clear cytotoxic lipoproteins and dead cells, preventing cytotoxicity. Efferocytosis - the efficient clearance of dead cells by macrophages - is crucial for preventing secondary necrosis and stimulating the release of anti-inflammatory cytokines. In addition, macrophages can promote tissue repair and proliferation of vascular smooth muscle cells, thereby increasing plaque stability. However, advanced atherosclerotic plaques contain large numbers of pro-inflammatory macrophages that secrete matrix-degrading enzymes, induce death in surrounding cells and contribute to plaque destabilization and rupture. Importantly, macrophages in the plaque can undergo apoptosis and several forms of regulated necrosis, including necroptosis, pyroptosis and ferroptosis. Regulated necrosis has an important role in the formation and expansion of the necrotic core during plaque progression, and several triggers for necrosis are present within atherosclerotic plaques. This Review focuses on the various forms of programmed macrophage death in atherosclerosis and the pharmacological interventions that target them as a potential means of stabilizing vulnerable plaques and improving the efficacy of currently available anti-atherosclerotic therapies.
Collapse
Affiliation(s)
- Guido R Y De Meyer
- Laboratory of Physiopharmacology, University of Antwerp, Antwerp, Belgium.
| | - Michelle Zurek
- Laboratory of Physiopharmacology, University of Antwerp, Antwerp, Belgium
| | - Pauline Puylaert
- Laboratory of Physiopharmacology, University of Antwerp, Antwerp, Belgium
| | - Wim Martinet
- Laboratory of Physiopharmacology, University of Antwerp, Antwerp, Belgium
| |
Collapse
|
42
|
Song M, Kang K, Wang S, Zhang C, Zhao X, Song F. Elevated intracellular Ca 2+ functions downstream of mitodysfunction to induce Wallerian-like degeneration and necroptosis in organophosphorus-induced delayed neuropathy. Toxicology 2024; 504:153812. [PMID: 38653376 DOI: 10.1016/j.tox.2024.153812] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Revised: 04/06/2024] [Accepted: 04/19/2024] [Indexed: 04/25/2024]
Abstract
Neurotoxic organophosphorus compounds can induce a type of delayed neuropathy in humans and sensitive animals, known as organophosphorus-induced delayed neuropathy (OPIDN). OPIDN is characterized by axonal degeneration akin to Wallerian-like degeneration, which is thought to be caused by increased intra-axonal Ca2+ concentrations. This study was designed to investigate that deregulated cytosolic Ca2+ may function downstream of mitodysfunction in activating Wallerian-like degeneration and necroptosis in OPIDN. Adult hens were administrated a single dosage of 750 mg/kg tri-ortho-cresyl phosphate (TOCP), and then sacrificed at 1 day, 5 day, 10 day and 21 day post-exposure, respectively. Sciatic nerves and spinal cords were examined for pathological changes and proteins expression related to Wallerian-like degeneration and necroptosis. In vitro experiments using differentiated neuro-2a (N2a) cells were conducted to investigate the relationship among mitochondrial dysfunction, Ca2+ influx, axonal degeneration, and necroptosis. The cells were co-administered with the Ca2+-chelator BAPTA-AM, the TRPA1 channel inhibitor HC030031, the RIPK1 inhibitor Necrostatin-1, and the mitochondrial-targeted antioxidant MitoQ along with TOCP. Results demonstrated an increase in cytosolic calcium concentration and key proteins associated with Wallerian degeneration and necroptosis in both in vivo and in vitro models after TOCP exposure. Moreover, co-administration with BATPA-AM or HC030031 significantly attenuated the loss of NMNAT2 and STMN2 in N2a cells, as well as the upregulation of SARM1, RIPK1 and p-MLKL. In contrast, Necrostatin-1 treatment only inhibited the TOCP-induced elevation of p-MLKL. Notably, pharmacological protection of mitochondrial function with MitoQ effectively alleviated the increase in intracellular Ca2+ following TOCP and mitigated axonal degeneration and necroptosis in N2a cells, supporting mitochondrial dysfunction as an upstream event of the intracellular Ca2+ imbalance and neuronal damage in OPIDN. These findings suggest that mitochondrial dysfunction post-TOCP intoxication leads to an elevated intracellular Ca2+ concentration, which plays a pivotal role in the initiation and development of OPIDN through inducing SARM1-mediated axonal degeneration and activating the necroptotic signaling pathway.
Collapse
Affiliation(s)
- Mingxue Song
- Department of Toxicology and Nutrition, School of Public Health, Cheeloo College of Medicine, Shandong University, 44 West Wenhua Road, Jinan, Shandong 250012, PR China
| | - Kang Kang
- Qingdao Municipal Center for Disease Control & Prevention, Qingdao, Shandong 266033, PR China
| | - Shuai Wang
- Department of Toxicology and Nutrition, School of Public Health, Cheeloo College of Medicine, Shandong University, 44 West Wenhua Road, Jinan, Shandong 250012, PR China
| | - Cuiqin Zhang
- Department of Toxicology and Nutrition, School of Public Health, Cheeloo College of Medicine, Shandong University, 44 West Wenhua Road, Jinan, Shandong 250012, PR China
| | - Xiulan Zhao
- Department of Toxicology and Nutrition, School of Public Health, Cheeloo College of Medicine, Shandong University, 44 West Wenhua Road, Jinan, Shandong 250012, PR China
| | - Fuyong Song
- Department of Toxicology and Nutrition, School of Public Health, Cheeloo College of Medicine, Shandong University, 44 West Wenhua Road, Jinan, Shandong 250012, PR China.
| |
Collapse
|
43
|
Luo T, Sang N, Liu Y, Zhou Y, Wu R, Bagdasarian FA, Wey HY, Lang J, Wang C, Bai P. Synthesis and preclinical evaluation of 11C-labeled 7-Oxo-2,4,5,7-tetrahydro-6H-pyrazolo[3,4-c]pyridine radioligands for RIPK1 positron emission tomography imaging. Bioorg Chem 2024; 146:107279. [PMID: 38513325 DOI: 10.1016/j.bioorg.2024.107279] [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: 12/28/2023] [Accepted: 03/08/2024] [Indexed: 03/23/2024]
Abstract
Targeting receptor-interacting protein kinase 1 (RIPK1) has emerged as a promising therapeutic strategy for various neurodegenerative disorders. The development of a positron emission tomography (PET) probe for brain RIPK1 imaging could offer a valuable tool to assess therapeutic effectiveness and uncover the neuropathology associated with RIPK1. In this study, we present the development and characterization of two new PET radioligands, [11C]PB218 and [11C]PB220, which have the potential to facilitate brain RIPK1 imaging. [11C]PB218 and [11C]PB220 were successfully synthesized with a high radiochemical yield (34 % - 42 %) and molar activity (293 - 314 GBq/µmol). PET imaging characterization of two radioligands was conducted in rodents, demonstrating that both newly developed tracers have good brain penetration (maximum SUV = 0.9 - 1.0) and appropriate brain clearance kinetic profiles. Notably, [11C]PB218 has a more favorable binding specificity than [11C]PB220. A PET/MR study of [11C]PB218 in a non-human primate exhibited good brain penetration, desirable kinetic properties, and a safe profile, thus supporting the translational applicability of our new probe. These investigations enable further translational exploration of [11C]PB218 for drug discovery and PET probe development targeting RIPK1.
Collapse
Affiliation(s)
- Tianwen Luo
- Department of Respiratory and Critical Care Medicine, Targeted Tracer Research and Development Laboratory, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China; Institute of Respiratory Health, Targeted Tracer Research and Development Laboratory, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China; Precision Medicine Center, Precision Medicine Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China; The Research Units of West China, Chinese Academy of Medical Sciences, West China Hospital, Chengdu, Sichuan, 610041, China; State Key Laboratory of Respiratory Health and Multimorbidity, West China Hospital, Chengdu, Sichuan, 610041, China
| | - Na Sang
- Department of Radiation Oncology, Radiation Oncology Key Laboratory of Sichuan Province, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu, China
| | - Yan Liu
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA 02129, United States
| | - Yanting Zhou
- Department of Respiratory and Critical Care Medicine, Targeted Tracer Research and Development Laboratory, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China; Institute of Respiratory Health, Targeted Tracer Research and Development Laboratory, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China; Precision Medicine Center, Precision Medicine Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China; The Research Units of West China, Chinese Academy of Medical Sciences, West China Hospital, Chengdu, Sichuan, 610041, China; State Key Laboratory of Respiratory Health and Multimorbidity, West China Hospital, Chengdu, Sichuan, 610041, China
| | - Rui Wu
- Department of Respiratory and Critical Care Medicine, Targeted Tracer Research and Development Laboratory, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China; Institute of Respiratory Health, Targeted Tracer Research and Development Laboratory, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China; Precision Medicine Center, Precision Medicine Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China; The Research Units of West China, Chinese Academy of Medical Sciences, West China Hospital, Chengdu, Sichuan, 610041, China; State Key Laboratory of Respiratory Health and Multimorbidity, West China Hospital, Chengdu, Sichuan, 610041, China
| | - Frederick A Bagdasarian
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA 02129, United States
| | - Hsiao-Ying Wey
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA 02129, United States
| | - Jinyi Lang
- Department of Radiation Oncology, Radiation Oncology Key Laboratory of Sichuan Province, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu, China
| | - Changning Wang
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA 02129, United States.
| | - Ping Bai
- Department of Respiratory and Critical Care Medicine, Targeted Tracer Research and Development Laboratory, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China; Institute of Respiratory Health, Targeted Tracer Research and Development Laboratory, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China; Precision Medicine Center, Precision Medicine Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China; The Research Units of West China, Chinese Academy of Medical Sciences, West China Hospital, Chengdu, Sichuan, 610041, China; State Key Laboratory of Respiratory Health and Multimorbidity, West China Hospital, Chengdu, Sichuan, 610041, China.
| |
Collapse
|
44
|
Divandari M, Javadifar A, Moghadam AB, Janatabadi AA. RIPK3 and RIPK1 gene expression in pterygium: unveiling molecular insights into pathogenesis. Mol Biol Rep 2024; 51:524. [PMID: 38630344 DOI: 10.1007/s11033-024-09368-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2024] [Accepted: 02/20/2024] [Indexed: 04/19/2024]
Abstract
BACKGROUND Pterygium, characterized by the abnormal proliferation of epithelial cells, matrix remodeling, vascularization, and lesion migration, is a prevalent ocular surface disease involving the growth of fibrovascular tissue on the cornea. Despite the unclear underlying causes of pterygium, numerous investigations have indicated the involvement of cell death pathways in the regulation of cell cycle dynamics. Consequently, the objective of this study was to assess the expression levels of necroptosis markers in individuals diagnosed with pterygium, aiming to shed light on the potential role of necroptosis in the pathogenesis of this condition. METHODS This study aimed to investigate the expression patterns of receptor-interacting serine/threonine kinase 3 (RIPK3) and receptor-interacting serine/threonine kinase 1 (RIPK1) genes in pterygium tissues. 41 patients undergoing pterygium excision surgery were recruited. Resected pterygium samples and normal conjunctival tissues were collected, and RIPK3 and RIPK1 mRNA levels were measured using quantitative real-time PCR. RESULTS Our findings reveal that the expression of RIPK3 is significantly increased in samples obtained from individuals with pterygium. However, no significant alterations were observed in the expression of RIPK1 in these samples. Results showed significantly higher RIPK3 expression in pterygium tissues compared to controls. Moreover, increased RIPK3 levels correlated negatively with pterygium recurrence rates. CONCLUSIONS These findings suggest RIPK3 may play a protective role against pterygium recurrence through necroptosis.
Collapse
Affiliation(s)
- Mahnaz Divandari
- Department of Biology, Sabzevar Branch, Islamic Azad University, Sabzevar, Iran
| | - Amin Javadifar
- Immunology Research Center, Inflammation and Inflammatory Diseases Division, Mashhad University of medical sciences, Mashhad, Iran
| | | | - Ali Akbar Janatabadi
- Department of Biology, Sabzevar Branch, Islamic Azad University, Sabzevar, Iran.
| |
Collapse
|
45
|
Li Z, Shang W, Mei T, Fu D, Xi F, Shao Y, Song X, Wang Z, Qi K, Tu J. Outer membrane vesicles of avian pathogenic Escherichia coli induce necroptosis and NF-κB activation in chicken macrophages via RIPK1 mediation. Res Vet Sci 2024; 170:105185. [PMID: 38422838 DOI: 10.1016/j.rvsc.2024.105185] [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: 08/10/2023] [Revised: 12/20/2023] [Accepted: 02/11/2024] [Indexed: 03/02/2024]
Abstract
Outer membrane vesicles (OMVs) are soluble mediators secreted by Gram-negative bacteria that are involved in communication. They can carry a variety of harmful molecules, which induce cytotoxic responses and inflammatory reactions in the absence of direct host cell-bacterium interactions. We previously reported the isolation of OMVs from avian pathogenic Escherichia coli (APEC) culture medium by ultracentrifugation, and characterized them as a substance capable of inducing the production of pro-inflammatory cytokines and causing tissue damage. However, the specific mechanisms by which APEC-secreted OMVs activate host cell death signaling and inflammation are poorly understood. Here, we show that OMVs are involved in the pathogenesis of APEC disease. In an APEC/chicken macrophage (HD11) coculture system, APEC significantly promoted HD11 cell death and inflammatory responses by secreting OMVs. Using western blotting analysis and specific pathway inhibitors, we demonstrated that the induction of HD11 death by APEC OMVs is associated with the activation of receptor interacting serine/threonine kinase 1 (RIPK1)-, receptor interacting serine/threonine kinase 3 (RIPK3)-, and mixed lineage kinase like pseudokinase (MLKL)-induced necroptosis. Notably, necroptosis inhibitor-1 (Nec-1), an RIPK1 inhibitor, reversed these effects. We also showed that APEC OMVs promote the activation of the NF-κB signaling pathway, leading to the phosphorylation of IκB-α and p65, the increased nuclear translocation of p65, and the significant upregulation of interleukin 1β (IL-1β) and IL-6 transcription. Importantly, APEC OMVs-induced IL-1β and IL-6 mRNA expression and the activation of the NF-κB signaling pathway were similarly significantly inhibited by a RIPK1-specific inhibitor. Based on these findings, we have established that RIPK1 plays a dual role in HD11 cells necroptosis and the proinflammatory cytokine (IL-1β and IL-6) expression induced by APEC OMVs. RIPK1 mediated the induction of necroptosis and the activation of the NF-κB in HD11 cells via APEC OMVs. The results of this study provide a basis for further investigation of the contribution of OMVs to the pathogenesis of APEC.
Collapse
Affiliation(s)
- Zhe Li
- Anhui Province Key Laboratory of Veterinary Pathobiology and Disease Control, College of Animal Science and Technology, Anhui Agricultural University, Hefei 230036, China; Anhui Province Engineering Laboratory for Animal Food Quality and Bio-safety, College of Animal Science and Technology, Anhui Agricultural University, Hefei 230036, China
| | - Wenbin Shang
- Anhui Province Key Laboratory of Veterinary Pathobiology and Disease Control, College of Animal Science and Technology, Anhui Agricultural University, Hefei 230036, China; Anhui Province Engineering Laboratory for Animal Food Quality and Bio-safety, College of Animal Science and Technology, Anhui Agricultural University, Hefei 230036, China
| | - Ting Mei
- Anhui Province Key Laboratory of Veterinary Pathobiology and Disease Control, College of Animal Science and Technology, Anhui Agricultural University, Hefei 230036, China; Anhui Province Engineering Laboratory for Animal Food Quality and Bio-safety, College of Animal Science and Technology, Anhui Agricultural University, Hefei 230036, China
| | - Dandan Fu
- Anhui Province Key Laboratory of Veterinary Pathobiology and Disease Control, College of Animal Science and Technology, Anhui Agricultural University, Hefei 230036, China; Anhui Province Engineering Laboratory for Animal Food Quality and Bio-safety, College of Animal Science and Technology, Anhui Agricultural University, Hefei 230036, China
| | - Feng Xi
- Anhui Province Key Laboratory of Veterinary Pathobiology and Disease Control, College of Animal Science and Technology, Anhui Agricultural University, Hefei 230036, China; Anhui Province Engineering Laboratory for Animal Food Quality and Bio-safety, College of Animal Science and Technology, Anhui Agricultural University, Hefei 230036, China
| | - Ying Shao
- Anhui Province Key Laboratory of Veterinary Pathobiology and Disease Control, College of Animal Science and Technology, Anhui Agricultural University, Hefei 230036, China; Anhui Province Engineering Laboratory for Animal Food Quality and Bio-safety, College of Animal Science and Technology, Anhui Agricultural University, Hefei 230036, China
| | - Xiangjun Song
- Anhui Province Key Laboratory of Veterinary Pathobiology and Disease Control, College of Animal Science and Technology, Anhui Agricultural University, Hefei 230036, China; Anhui Province Engineering Laboratory for Animal Food Quality and Bio-safety, College of Animal Science and Technology, Anhui Agricultural University, Hefei 230036, China
| | - Zhenyu Wang
- Anhui Province Key Laboratory of Veterinary Pathobiology and Disease Control, College of Animal Science and Technology, Anhui Agricultural University, Hefei 230036, China; Anhui Province Engineering Laboratory for Animal Food Quality and Bio-safety, College of Animal Science and Technology, Anhui Agricultural University, Hefei 230036, China
| | - Kezong Qi
- Anhui Province Key Laboratory of Veterinary Pathobiology and Disease Control, College of Animal Science and Technology, Anhui Agricultural University, Hefei 230036, China; Anhui Province Engineering Laboratory for Animal Food Quality and Bio-safety, College of Animal Science and Technology, Anhui Agricultural University, Hefei 230036, China
| | - Jian Tu
- Anhui Province Key Laboratory of Veterinary Pathobiology and Disease Control, College of Animal Science and Technology, Anhui Agricultural University, Hefei 230036, China; Anhui Province Engineering Laboratory for Animal Food Quality and Bio-safety, College of Animal Science and Technology, Anhui Agricultural University, Hefei 230036, China.
| |
Collapse
|
46
|
Park KA, Jung CS, Sohn KC, Ju E, Shin S, Park I, Na M, Hur GM. Eupatolide, isolated from Liriodendron tulipifera, sensitizes TNF-mediated dual modes of apoptosis and necroptosis by disrupting RIPK1 ubiquitination. Heliyon 2024; 10:e28092. [PMID: 38533031 PMCID: PMC10963378 DOI: 10.1016/j.heliyon.2024.e28092] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Revised: 03/11/2024] [Accepted: 03/12/2024] [Indexed: 03/28/2024] Open
Abstract
Ubiquitination of RIPK1 plays an essential role in the recruitment of the IKK complex, an upstream component of pro-survival NF-κB. It also limits TNF-induced programmed cell death by inhibiting the spatial transition from TNFR1-associated complex-I to RIPK1-dependent death-inducing complex-II or necrosome. Thus, the targeted disruption of RIPK1 ubiquitination, which induces RIPK1-dependent cell death, has proven to be a useful strategy for improving the therapeutic efficacy of TNF. In this study, we found that eupatolide, isolated from Liriodendron tulipifera, is a potent activator of the cytotoxic potential of RIPK1 by disrupting the ubiquitination of RIPK1 upon TNFR1 ligation. Analysis of events upstream of NF-κB signaling revealed that eupatolide inhibited IKKβ-mediated NF-κB activation while having no effect on IKKα-mediated non-canonical NF-κB activation. Pretreatment with eupatolide drastically interfered with RIPK1 recruitment to the TNFR1 complex-I by disrupting RIPK1 ubiquitination. Moreover, eupatolide was sufficient to upregulate the activation of RIPK1, facilitating the TNF-mediated dual modes of apoptosis and necroptosis. Thus, we propose a novel mechanism by which eupatolide activates the cytotoxic potential of RIPK1 at the TNFR1 level and provides a promising anti-cancer therapeutic approach to overcome TNF resistance.
Collapse
Affiliation(s)
- Kyeong Ah Park
- Department of Pharmacology and Department of Medical Science, College of Medicine, Chungnam National University, 266 Munhwa-ro, Daejeon, 35015, Republic of Korea
| | - Chan Seok Jung
- Department of Pharmacology and Department of Medical Science, College of Medicine, Chungnam National University, 266 Munhwa-ro, Daejeon, 35015, Republic of Korea
| | - Kyung-Cheol Sohn
- Department of Pharmacology and Department of Medical Science, College of Medicine, Chungnam National University, 266 Munhwa-ro, Daejeon, 35015, Republic of Korea
| | - Eunjin Ju
- Department of Pharmacology and Department of Medical Science, College of Medicine, Chungnam National University, 266 Munhwa-ro, Daejeon, 35015, Republic of Korea
| | - Sanghee Shin
- Department of Pharmacology and Department of Medical Science, College of Medicine, Chungnam National University, 266 Munhwa-ro, Daejeon, 35015, Republic of Korea
| | - InWha Park
- Natural Product Informatics Research Center, KIST Gangneung Institute of Natural Products, Gangneung, 25451, Republic of Korea
| | - MinKyun Na
- College of Pharmacy, Chungnam National University, 99 Daehak-ro, Daejeon, 34134, Republic of Korea
| | - Gang Min Hur
- Department of Pharmacology and Department of Medical Science, College of Medicine, Chungnam National University, 266 Munhwa-ro, Daejeon, 35015, Republic of Korea
| |
Collapse
|
47
|
Yang T, Xiang CG, Wang XH, Li QQ, Lei SY, Zhang KR, Ren J, Lu HM, Feng CL, Tang W. RIPK1 inhibitor ameliorates pulmonary injury by modulating the function of neutrophils and vascular endothelial cells. Cell Death Discov 2024; 10:152. [PMID: 38521771 PMCID: PMC10960796 DOI: 10.1038/s41420-024-01921-8] [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: 01/22/2024] [Revised: 03/15/2024] [Accepted: 03/18/2024] [Indexed: 03/25/2024] Open
Abstract
Acute lung injury (ALI) is an acute and progressive hypoxic respiratory failure that could progress to acute respiratory distress syndrome (ARDS) with a high mortality rate, thus immediate medical attention and supportive care are necessary. The pathophysiology of ALI is characterized by the disruption of the alveolar-capillary barrier and activation of neutrophils, leading to lung tissue damage. The receptor-interacting protein kinase 1 (RIPK1) has emerged as a promising target for the treatment of multiple inflammatory diseases, but the role of RIPK1 in the ALI remains poorly understood. In this study, we aimed to figure out the pathological role of RIPK1 in ALI, especially in the pulmonary immune microenvironment involving neutrophils and endothelial cells. In vivo experiments showed that RIPK1 inhibitor protected against lipopolysaccharide (LPS)-induced lung injury in mouse models, with reduced neutrophils and monocytes infiltration in the lungs. Further studies demonstrated that, besides the inhibitory action on necroptosis, RIPK1 inhibitor directly suppressed reactive oxygen species (ROS) generation and inflammatory cytokines secretion from neutrophils. Furthermore, RIPK1 inhibition maintains the barrier function in TNF-α-primed vascular endothelial cells and prevents their activation induced by the supernatant from LPS-stimulated neutrophils. Mechanistically, the aforementioned effects of RIPK1 inhibitor are associated with the NF-κB signaling pathway, which is partially independent of necroptosis inhibition. These results provide new evidence that RIPK1 inhibitor directly regulates the function of neutrophils and endothelial cells, as well as interferes with the interactions between these two cell types, therefore contributing to a better understanding of RIPK1 in ALI and providing a potential avenue for future therapeutic interventions.
Collapse
Affiliation(s)
- Tao Yang
- State Key Laboratory of Chemical Biology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
- School of Pharmacy, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Cai-Gui Xiang
- State Key Laboratory of Chemical Biology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
- School of Pharmacy, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Xiao-Han Wang
- State Key Laboratory of Chemical Biology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
- School of Pharmacy, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Qing-Qing Li
- State Key Laboratory of Chemical Biology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Shu-Yue Lei
- State Key Laboratory of Chemical Biology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
- School of Pharmacy, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Kai-Rong Zhang
- School of Pharmaceutical Science, Nanchang University, Nanchang, 330006, China
| | - Jing Ren
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, 210000, China
| | - Hui-Min Lu
- State Key Laboratory of Chemical Biology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
- School of Pharmacy, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Chun-Lan Feng
- State Key Laboratory of Chemical Biology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Wei Tang
- State Key Laboratory of Chemical Biology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China.
- School of Pharmacy, University of Chinese Academy of Sciences, Beijing, 100049, China.
| |
Collapse
|
48
|
Matsuda R, Sorobetea D, Zhang J, Peterson ST, Grayczyk JP, Yost W, Apenes N, Kovalik ME, Herrmann B, O’Neill RJ, Bohrer AC, Lanza M, Assenmacher CA, Mayer-Barber KD, Shin S, Brodsky IE. A TNF-IL-1 circuit controls Yersinia within intestinal pyogranulomas. J Exp Med 2024; 221:e20230679. [PMID: 38363547 PMCID: PMC10873131 DOI: 10.1084/jem.20230679] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Revised: 11/22/2023] [Accepted: 01/19/2024] [Indexed: 02/17/2024] Open
Abstract
Tumor necrosis factor (TNF) is a pleiotropic inflammatory cytokine that mediates antimicrobial defense and granuloma formation in response to infection by numerous pathogens. We previously reported that Yersinia pseudotuberculosis colonizes the intestinal mucosa and induces the recruitment of neutrophils and inflammatory monocytes into organized immune structures termed pyogranulomas (PG) that control Yersinia infection. Inflammatory monocytes are essential for the control and clearance of Yersinia within intestinal PG, but how monocytes mediate Yersinia restriction is poorly understood. Here, we demonstrate that TNF signaling in monocytes is required for bacterial containment following enteric Yersinia infection. We further show that monocyte-intrinsic TNFR1 signaling drives the production of monocyte-derived interleukin-1 (IL-1), which signals through IL-1 receptors on non-hematopoietic cells to enable PG-mediated control of intestinal Yersinia infection. Altogether, our work reveals a monocyte-intrinsic TNF-IL-1 collaborative inflammatory circuit that restricts intestinal Yersinia infection.
Collapse
Affiliation(s)
- Rina Matsuda
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Daniel Sorobetea
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Jenna Zhang
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Stefan T. Peterson
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - James P. Grayczyk
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Winslow Yost
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Nicolai Apenes
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Maria E. Kovalik
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Beatrice Herrmann
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Rosemary J. O’Neill
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Andrea C. Bohrer
- Inflammation and Innate Immunity Unit, Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Matthew Lanza
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Charles-Antoine Assenmacher
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Katrin D. Mayer-Barber
- Inflammation and Innate Immunity Unit, Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Sunny Shin
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Igor E. Brodsky
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
49
|
Zhou X, Zhu Y, Gao L, Li Y, Li H, Huang C, Liu Y, Hu A, Ying C, Song Y. Binding of RAGE and RIPK1 induces cognitive deficits in chronic hyperglycemia-derived neuroinflammation. CNS Neurosci Ther 2024; 30:e14449. [PMID: 37665158 PMCID: PMC10916433 DOI: 10.1111/cns.14449] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Revised: 07/08/2023] [Accepted: 08/16/2023] [Indexed: 09/05/2023] Open
Abstract
AIMS Chronic hyperglycemia-induced inflammation of the hippocampus is an important cause of cognitive deficits in diabetic patients. The receptor for advanced glycation end products (RAGE), which is widely expressed in the hippocampus, is a crucial factor in this inflammation and the associated cognitive deficits. We aimed to reveal the underlying mechanism by which RAGE regulates neuroinflammation in the pathogenesis of diabetes-induced cognitive impairment. METHODS We used db/db mice as a model for type 2 diabetes to investigate whether receptor-interacting serine/threonine protein kinase 1 (RIPK1), which is expressed in microglia in the hippocampal region, is a key protein partner for RAGE. GST pull-down assays and AutoDock Vina simulations were performed to identify the key structural domain in RAGE that binds to RIPK1. Western blotting, co-immunoprecipitation (Co-IP), and immunofluorescence (IF) were used to detect the levels of key proteins or interaction between RAGE and RIPK1. Cognitive deficits in the mice were assessed with the Morris water maze (MWM) and new object recognition (NOR) and fear-conditioning tests. RESULTS RAGE binds directly to RIPK1 via the amino acid sequence (AAs) 362-367, thereby upregulating phosphorylation of RIPK1, which results in activation of the NLRP3 inflammasome in microglia and ultimately leads to cognitive impairments in db/db mice. We mutated RAGE AAs 362-367 to reverse neuroinflammation in the hippocampus and improve cognitive function, suggesting that RAGE AAs 362-367 is a key structural domain that binds directly to RIPK1. These results also indicate that hyperglycemia-induced inflammation in the hippocampus is dependent on direct binding of RAGE and RIPK1. CONCLUSION Direct interaction of RAGE and RIPK1 via AAs 362-367 is an important mechanism for enhanced neuroinflammation in the hyperglycemic environment and is a key node in the development of cognitive deficits in diabetes.
Collapse
Affiliation(s)
- Xiaoyan Zhou
- Xuzhou Engineering Research Center of Medical Genetics and Transformation, Department of GeneticsXuzhou Medical UniversityXuzhouJiangsuChina
| | - Yandong Zhu
- The Graduate SchoolXuzhou Medical UniversityXuzhouJiangsuChina
| | - Lin Gao
- The Graduate SchoolXuzhou Medical UniversityXuzhouJiangsuChina
| | - Yan Li
- The Graduate SchoolXuzhou Medical UniversityXuzhouJiangsuChina
| | - Hui Li
- The Graduate SchoolXuzhou Medical UniversityXuzhouJiangsuChina
| | - Chengyu Huang
- The Graduate SchoolXuzhou Medical UniversityXuzhouJiangsuChina
| | - Yan Liu
- The Graduate SchoolXuzhou Medical UniversityXuzhouJiangsuChina
| | - Ankang Hu
- Lab Animal CenterXuzhou Medical UniversityXuzhouChina
| | - Changjiang Ying
- Department of EndocrinologyAffiliated Hospital of Xuzhou Medical UniversityXuzhouJiangsuChina
| | - Yuanjian Song
- Xuzhou Engineering Research Center of Medical Genetics and Transformation, Department of GeneticsXuzhou Medical UniversityXuzhouJiangsuChina
| |
Collapse
|
50
|
Li Y, Jie W, Qi Y, Mo M, Lian Y, Yin L, Huang H. Inhibition of RIPK1 alleviating vascular smooth muscle cells osteogenic transdifferentiation via Runx2. iScience 2024; 27:108766. [PMID: 38318355 PMCID: PMC10839642 DOI: 10.1016/j.isci.2023.108766] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Revised: 09/05/2023] [Accepted: 12/18/2023] [Indexed: 02/07/2024] Open
Abstract
Vascular calcification (VC) is recognized as a crucial risk factor for cardiovascular diseases. Our previous report revealed that the osteogenic transdifferentiation of vascular smooth muscle cells (VSMCs) plays a role in this process. However, the underlying molecular mechanisms remain elusive. Notably, receptor-interacting protein kinase 1 (RIPK1) has been implicated in the development of cardiovascular diseases, yet its role and mechanisms in VC remain unexplored. To address this gap, we established models using chronic kidney disease mice and calcifying VSMCs to investigate the impact of RIPK1 on VC. Subsequently, a RIPK1-specific inhibitor (NEC-1) was applied in both in vitro and in vivo models. Our findings indicate significant activation of RIPK1 in calcified human arterial tissue, as well as in animal and cellular models. RIPK1 activation promotes the osteogenic transdifferentiation of VSMCs. Treatment with the NEC-1 substantially reduced VC. These results demonstrate that RIPK1 is a target for preventing VC.
Collapse
Affiliation(s)
- Yue Li
- Cardiovascular Department, The Eighth Affiliated Hospital, Joint Laboratory of Guangdong-Hong Kong-Macao Universities for Nutritional Metabolism and Precise Prevention and Control of Major Chronic Diseases, Sun Yat-sen University, Shenzhen, China
| | - Wei Jie
- Cardiovascular Department, The Eighth Affiliated Hospital, Joint Laboratory of Guangdong-Hong Kong-Macao Universities for Nutritional Metabolism and Precise Prevention and Control of Major Chronic Diseases, Sun Yat-sen University, Shenzhen, China
| | - Yanli Qi
- Cardiovascular Department, The Eighth Affiliated Hospital, Joint Laboratory of Guangdong-Hong Kong-Macao Universities for Nutritional Metabolism and Precise Prevention and Control of Major Chronic Diseases, Sun Yat-sen University, Shenzhen, China
| | - Mingxing Mo
- Cardiovascular Department, The Eighth Affiliated Hospital, Joint Laboratory of Guangdong-Hong Kong-Macao Universities for Nutritional Metabolism and Precise Prevention and Control of Major Chronic Diseases, Sun Yat-sen University, Shenzhen, China
| | - Yaxin Lian
- Cardiovascular Department, The Eighth Affiliated Hospital, Joint Laboratory of Guangdong-Hong Kong-Macao Universities for Nutritional Metabolism and Precise Prevention and Control of Major Chronic Diseases, Sun Yat-sen University, Shenzhen, China
| | - Li Yin
- Cardiovascular Department, The Eighth Affiliated Hospital, Joint Laboratory of Guangdong-Hong Kong-Macao Universities for Nutritional Metabolism and Precise Prevention and Control of Major Chronic Diseases, Sun Yat-sen University, Shenzhen, China
| | - Hui Huang
- Cardiovascular Department, The Eighth Affiliated Hospital, Joint Laboratory of Guangdong-Hong Kong-Macao Universities for Nutritional Metabolism and Precise Prevention and Control of Major Chronic Diseases, Sun Yat-sen University, Shenzhen, China
| |
Collapse
|