1
|
Su K, Tang M, Wu J, Ye N, Jiang X, Zhao M, Zhang R, Cai X, Zhang X, Li N, Peng J, Lin L, Wu W, Ye H. Mechanisms and therapeutic strategies for NLRP3 degradation via post-translational modifications in ubiquitin-proteasome and autophagy lysosomal pathway. Eur J Med Chem 2025; 289:117476. [PMID: 40056798 DOI: 10.1016/j.ejmech.2025.117476] [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: 12/13/2024] [Revised: 02/20/2025] [Accepted: 03/03/2025] [Indexed: 03/10/2025]
Abstract
The NLRP3 inflammasome is crucial for immune responses. However, its overactivation can lead to severe inflammatory diseases, underscoring its importance as a target for therapeutic intervention. Although numerous inhibitors targeting NLRP3 exist, regulating its degradation offers an alternative and promising strategy to suppress its activation. The degradation of NLRP3 is primarily mediated by the proteasomal and autophagic pathways. The review not only elaborates on the traditional concepts of ubiquitination and NLRP3 degradation but also investigates the important roles of indirect regulatory modifications, such as phosphorylation, acetylation, ubiquitin-like modifications, and palmitoylation-key post-translational modifications (PTMs) that influence NLRP3 degradation. Additionally, we also discuss the potential targets that may affect NLRP3 degradation during the proteasomal and autophagic pathways. By unraveling these complex regulatory mechanisms, the review aims to enhance the understanding of NLRP3 regulation and its implications for developing therapeutic strategies to combat inflammatory diseases.
Collapse
Affiliation(s)
- Kaiyue Su
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Minghai Tang
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Jie Wu
- Key Laboratory of Hydrodynamics (Ministry of Education), School of Ocean and Civil Engineering, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Neng Ye
- Scaled Manufacturing Center of Biological Products, Management Office of National Facility for Translational Medicine, West China Hospital, Sichuan University Chengdu 610041, China
| | - Xueqin Jiang
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Min Zhao
- Laboratory of Metabolomics and Drug-induced Liver Injury, Department of Gastroenterology & Hepatology, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Ruijia Zhang
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Xiaoying Cai
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Xinlu Zhang
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Na Li
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Jing Peng
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Lei Lin
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Wenshuang Wu
- Division of Thyroid Surgery, Department of General Surgery and Laboratory of Thyroid and Parathyroid Disease, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, China.
| | - Haoyu Ye
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China.
| |
Collapse
|
2
|
Wang Y, Xu X, Zhang P, Hu S, Zhang L, Chen H. E3 Ubiquitin Ligase TRIM7 Alleviates Lipopolysaccharide-Induced Acute Lung Injury via Inhibiting NLRP3 Inflammasome Activation. THE AMERICAN JOURNAL OF PATHOLOGY 2025; 195:639-651. [PMID: 39864619 DOI: 10.1016/j.ajpath.2024.12.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/29/2024] [Revised: 11/25/2024] [Accepted: 12/27/2024] [Indexed: 01/28/2025]
Abstract
Acute lung injury (ALI) is a common clinical disease with high mortality, characterized by tissue damage caused by excessive activation of inflammation. TRIM7 is an E3 ligase that plays an important role in regulating viral infection, tumor progression, and innate immune response. However, its function in ALI is unclear. In this study, lipopolysaccharide (LPS) was used to stimulate C57BL/6j mice and HULEC-5a cells to establish ALI models in vivo and in vitro. TRIM7 expression was down-regulated during ALI. Furthermore, overexpressing TRIM7 in HULEC-5a cells relieved cell damage and inflammatory activation induced by LPS stimulation. TRIM7 knockdown had the opposite effect. Trim7-overexpressing mice were established by endotracheal injection of adeno-associated virus 6-Trim7 virus in vivo; the ALI model was then induced by LPS stimulation. Overexpression of TRIM7 could alleviate lung tissue injury, pulmonary interstitial hemorrhage, increased alveolar and vascular permeability, inflammatory cell infiltration, and secretion of inflammatory factors induced by LPS stimulation. Mechanistically, TRIM7 inhibited the expression of NOD-, LRR- and pyrin domain-containing 3 (NLRP3) and the activation of the NLRP3 inflammasome. The regulatory effect of TRIM7 on ALI depended on the NLRP3 inflammasome. This investigation, for the first time, showed the inhibitory effect of TRIM7 on ALI and activation of the NLRP3 inflammasome, providing new targets and ideas for the research on the mechanism and treatment of ALI.
Collapse
Affiliation(s)
- Youna Wang
- Department of Pulmonary and Critical Care Medicine, Renmin Hospital of Wuhan University, Wuhan, China
| | - Xiaohong Xu
- Department of Pulmonary and Critical Care Medicine, Hanchuan People's Hospital, Hanchuan, China
| | - Peng Zhang
- Taikang Medical School (School of Basic Medical Sciences), Wuhan University, Wuhan, China
| | - Sha Hu
- Taikang Medical School (School of Basic Medical Sciences), Wuhan University, Wuhan, China
| | - Li Zhang
- Center for Animal Experiment, Wuhan University, Wuhan, China.
| | - Hongbin Chen
- Department of Pulmonary and Critical Care Medicine, Renmin Hospital of Wuhan University, Wuhan, China.
| |
Collapse
|
3
|
Beesetti S. Ubiquitin Ligases in Control: Regulating NLRP3 Inflammasome Activation. FRONT BIOSCI-LANDMRK 2025; 30:25970. [PMID: 40152367 DOI: 10.31083/fbl25970] [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: 08/02/2024] [Revised: 09/04/2024] [Accepted: 09/11/2024] [Indexed: 03/29/2025]
Abstract
Ubiquitin ligases play pivotal roles in the regulation of NLR family pyrin domain containing 3 (NLRP3) inflammasome activation, a critical process in innate immunity and inflammatory responses. This review explores the intricate mechanisms by which various E3 ubiquitin ligases exert both positive and negative influences on NLRP3 inflammasome activity through diverse post-translational modifications. Negative regulation of NLRP3 inflammasome assembly is mediated by several E3 ligases, including F-box and leucine-rich repeat protein 2 (FBXL2), tripartite motif-containing protein 31 (TRIM31), and Casitas B-lineage lymphoma b (Cbl-b), which induce K48-linked ubiquitination of NLRP3, targeting it for proteasomal degradation. Membrane-associated RING-CH 7 (MARCH7) similarly promotes K48-linked ubiquitination leading to autophagic degradation, while RING finger protein (RNF125) induces K63-linked ubiquitination to modulate NLRP3 function. Ariadne homolog 2 (ARIH2) targets the nucleotide-binding domain (NBD) domain of NLRP3, inhibiting its activation, and tripartite motif-containing protein (TRIM65) employs dual K48 and K63-linked ubiquitination to suppress inflammasome assembly. Conversely, Pellino2 exemplifies a positive regulator, promoting NLRP3 inflammasome activation through K63-linked ubiquitination. Additionally, ubiquitin ligases influence other components critical for inflammasome function. TNF receptor-associated factor 3 (TRAF3) mediates K63 polyubiquitination of apoptosis-associated speck-like protein containing a CARD (ASC), facilitating its degradation, while E3 ligases regulate caspase-1 activation and DEAH-box helicase 33 (DHX33)-NLRP3 complex formation through specific ubiquitination events. Beyond direct inflammasome regulation, ubiquitin ligases impact broader innate immune signaling pathways, modulating pattern-recognition receptor responses and dendritic cell maturation. Furthermore, they intricately control NOD1/NOD2 signaling through K63-linked polyubiquitination of receptor-interacting protein 2 (RIP2), crucial for nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) and mitogen-activated protein kinase (MAPK) activation. Furthermore, we explore how various pathogens, including bacteria, viruses, and parasites, have evolved sophisticated strategies to hijack the host ubiquitination machinery, manipulating NLRP3 inflammasome activation to evade immune responses. This comprehensive analysis provides insights into the molecular mechanisms underlying inflammasome regulation and their implications for inflammatory diseases, offering potential avenues for therapeutic interventions targeting the NLRP3 inflammasome. In conclusion, ubiquitin ligases emerge as key regulators of NLRP3 inflammasome activation, exhibiting a complex array of functions that finely tune immune responses. Understanding these regulatory mechanisms not only sheds light on fundamental aspects of inflammation but also offers potential therapeutic avenues for inflammatory disorders and infectious diseases.
Collapse
Affiliation(s)
- Swarna Beesetti
- Department of Immunology, St Jude Children's Research Hospital, Memphis, TN 38105, USA
| |
Collapse
|
4
|
Li Y, Wang X, Ren Y, Han BZ, Xue Y. Exploring the health benefits of food bioactive compounds from a perspective of NLRP3 inflammasome activation: an insight review. Crit Rev Food Sci Nutr 2025:1-26. [PMID: 39757837 DOI: 10.1080/10408398.2024.2448768] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2025]
Abstract
The food industry has been focusing on food bioactive compounds with multiple physiological and immunological properties that benefit human health. These bioactive compounds, including polyphenols, flavonoids, and terpenoids, have great potential to limit inflammatory responses especially NLRP3 inflammasome activation, which is a key innate immune platform for inflammation. Current studies have revealed numerous food bioactive compounds with promising activities for unraveling immune metabolic disorders and excessive inflammatory responses by directly and indirectly regulating the NLRP3 inflammasome activation. This review explores the food hazards, including microbial and abiotic factors, that may trigger NLRP3-mediated illnesses and inflammation. It also highlights bioactive compounds in food that can suppress NLRP3 inflammasome activation through various mechanisms, linking its activation and inhibition to different pathways. Especially, this review provided further insight into NLRP3-related targets where food bioactive compounds can interact to block the NLRP3 inflammasome activation process, as well as mechanisms on how these compounds facilitate inactivation processes.
Collapse
Affiliation(s)
- Yabo Li
- Beijing Laboratory for Food Quality and Safety, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, China
| | - Xinyi Wang
- Beijing Laboratory for Food Quality and Safety, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, China
| | - Ying Ren
- Beijing Laboratory for Food Quality and Safety, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, China
| | - Bei-Zhong Han
- Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, China Agricultural University, Beijing, China
| | - Yansong Xue
- Beijing Laboratory for Food Quality and Safety, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, China
- Key Laboratory of Food Bioengineering (China National Light Industry), College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, China
| |
Collapse
|
5
|
Abolfathi F, Ranjbar R, Tabandeh MR, Habibi A. Cold water immersion regulates NLRP3 inflammasome pathway in the rat skeletal muscle after eccentric exercise by regulating the ubiquitin proteasome related proteins. Cytokine 2024; 184:156793. [PMID: 39467485 DOI: 10.1016/j.cyto.2024.156793] [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: 04/22/2024] [Revised: 09/28/2024] [Accepted: 10/21/2024] [Indexed: 10/30/2024]
Abstract
BACKGROUND Eccentric exercise (ECC) can induce NLRP3-related inflammation in skeletal muscle tissue. Limited available data have shown that Cold water immersion (CWI) after ECC can suppress skeletal muscle inflammation. This study aims to investigate the effect of CWI after ECC on the NLRP3 inflammasome pathway, and the expression of ubiquitin-proteasome-related proteins (UPPs) in the skeletal muscle of rats. METHODS Twenty-five male Wistar rats were randomly divided into control, ECC, ECC + CWI, ECC + NWI (normal water immersion), and ECC + AR (active recovery) groups. The Eccentric exercise consisted of 90 min of downhill running on a treadmill with a speed of 16 m/min and -16° incline. Animals in the NWI and CWI groups were immersed in water at 25 °C and 10 °C after ECC. Eventually, The soleus muscle was isolated and the expression of NLRP3, caspase-1, FBXL2, TRIM31, and PARKIN was evaluated by western blot. Tissue levels of IL-1β and IL-18 were measured by ELISA assay. RESULTS ECC significantly increased the expression of NLRP3, caspase-1, and the tissue levels of IL-1β and IL-18 compared to the control group. After ECC, FBXL2, and PARKIN were downregulated, whereas TRIM31 was up-regulated (P < 0.05). CWI after ECC suppressed the NLRP3 inflammasome components and increased the protein levels of FBXL2 and TRIM31 at higher levels than other recovery methods (P < 0.05). CWI and AR had the same increase in PARKIN expression and the same decrease in CK level compared to the ECC group (P < 0.05). CONCLUSION Our results indicated that CWI increased the expression of NLRP3-related UPPs in concomitant with suppression of NLRP3 in the soleus muscle of rats after ECC. As a result the beneficial effects of CWI on the attenuation of skeletal muscle inflammation may contribute to an alteration of UPPs expression.
Collapse
Affiliation(s)
- Farzaneh Abolfathi
- Department of Sport Physiology, Faculty of Sport Sciences, Shahid Chamran University of Ahvaz, Ahvaz, Iran.
| | - Rouhollah Ranjbar
- Department of Sport Physiology, Faculty of Sport Sciences, Shahid Chamran University of Ahvaz, Ahvaz, Iran.
| | - Mohammad Reza Tabandeh
- Department of Basic Sciences, Division of Biochemistry and Molecular Biology, Faculty of Veterinary Medicine, Shahid Chamran University of Ahvaz, Ahvaz, Iran; Stem Cells and Transgenic Technology Research Center, Shahid Chamran University of Ahvaz, Ahvaz, Iran.
| | - Abdolhamid Habibi
- Department of Sport Physiology, Faculty of Sport Sciences, Shahid Chamran University of Ahvaz, Ahvaz, Iran.
| |
Collapse
|
6
|
Chen H, Xie S, Zhou Y, Chen L, Xu J, Cai J. MEK1/2 promote ROS production and deubiquitinate NLRP3 independent of ERK1/2 during NLRP3 inflammasome activation. Biochem Pharmacol 2024; 230:116572. [PMID: 39396647 DOI: 10.1016/j.bcp.2024.116572] [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: 04/28/2024] [Revised: 08/22/2024] [Accepted: 10/10/2024] [Indexed: 10/15/2024]
Abstract
Inflammasomes are cytosolic supramolecular complexes that play a key role in the innate immune response. Overactivation of NLR family pyrin domain containing 3 (NLRP3) inflammasome leads to multiple diseases. Post-translational modifications (PTMs) are essential modulators of inflammasomes especially in activation phase. Here we found that MEK1/2 kinase activity was indispensable in NLRP3 inflammasome activation both in vitro and in vivo. Inhibition of MEK1/2 resulted in reactive oxygen species (ROS) scavenging and ubiquitination of NLRP3, which further blocked NLRP3 inflammasome activation. These effects were independent of ERK1/2, which were classic downstream of MEK1/2. These investigations proposed a mechanism that MEK1/2 regulated inflammation via non-transcriptional regulation of NLRP3 inflammasome and might help better understanding the effects and side-effects of MEK inhibitors in clinical use.
Collapse
Affiliation(s)
- Hanwen Chen
- Department of Gastroenterology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310009, PR China
| | - Shujun Xie
- Department of Translational Medicine Research Center, Key Laboratory of Clinical Cancer Pharmacology and Toxicology Research of Zhejiang Province, Affiliated Hangzhou First People's Hospital, School of Medicine, Westlake University, Hangzhou, Zhejiang, 310006, PR China
| | - Yichen Zhou
- MOA Key Laboratory of Animal Virology, Center for Veterinary Sciences, Department of Veterinary Medicine, College of Animal Sciences, Zhejiang University, Hangzhou, Zhejiang, 310058, PR China
| | - Lin Chen
- Department of General Practice, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, 310016, PR China
| | - Jian Xu
- Department of Central Laboratory, Affiliated Hangzhou First People's Hospital, School of Medicine, Westlake University, Hangzhou, 310006, PR China
| | - Jianting Cai
- Department of Gastroenterology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310009, PR China.
| |
Collapse
|
7
|
Zhou YR, Dang JJ, Yang QC, Sun ZJ. The regulation of pyroptosis by post-translational modifications: molecular mechanisms and therapeutic targets. EBioMedicine 2024; 109:105420. [PMID: 39476537 PMCID: PMC11564932 DOI: 10.1016/j.ebiom.2024.105420] [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: 05/21/2024] [Revised: 08/23/2024] [Accepted: 10/09/2024] [Indexed: 11/17/2024] Open
Abstract
Pyroptosis, a type of programmed cell death mediated by gasdermin family proteins, releases a large amount of immune stimulatory substances, which further contribute to inflammation and elicit an adaptive immune response against tumours and pathogens. And it occurs through multiple pathways that involve the activation of specific caspases and the cleavage of gasdermins. Post-translational modifications (PTMs) could influence the chemical properties of the modified residues and neighbouring regions, ultimately affecting the activity, stability, and functions of proteins to regulate pyroptosis. Many studies have been conducted to explore the influence of PTMs on the regulation of pyroptosis. In this review, we provide a comprehensive summary of different types of PTMs that influence pyroptosis, along with their corresponding modifying enzymes. Moreover, it elaborates on the specific contributions of different PTMs to pyroptosis and delves into how the regulation of these modifications can be leveraged for therapeutic interventions in cancer and inflammatory diseases.
Collapse
Affiliation(s)
- Yi-Rao Zhou
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Frontier Science Centre for Immunology and Metabolism, Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan, 430079, China
| | - Jun-Jie Dang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Frontier Science Centre for Immunology and Metabolism, Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan, 430079, China
| | - Qi-Chao Yang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Frontier Science Centre for Immunology and Metabolism, Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan, 430079, China.
| | - Zhi-Jun Sun
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Frontier Science Centre for Immunology and Metabolism, Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan, 430079, China.
| |
Collapse
|
8
|
Wang T, Sun G, Tao B. Updated insights into the NLRP3 inflammasome in postoperative cognitive dysfunction: emerging mechanisms and treatments. Front Aging Neurosci 2024; 16:1480502. [PMID: 39411285 PMCID: PMC11474915 DOI: 10.3389/fnagi.2024.1480502] [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: 08/14/2024] [Accepted: 09/17/2024] [Indexed: 10/19/2024] Open
Abstract
Postoperative cognitive dysfunction (POCD) poses a significant threat to patients undergoing anesthesia and surgery, particularly elderly patients. It is characterized by diminished cognitive functions post surgery, such as impaired memory and decreased concentration. The potential risk factors for POCD include age, surgical trauma, anesthetic type, and overall health condition; however, the precise mechanisms underlying POCD remain elusive. Recent studies suggest that neuroinflammation might be a primary pathogenic factor. NOD-, LRR- and pyrin domain-containing protein 3 (NLRP3) inflammasomes are implicated in exacerbating POCD by promoting the release of inflammatory factors and proteins that initiate pyroptosis, further influencing the disease process. The regulation of NLRP3 inflammasome activity, including its activation and degradation, is tightly controlled through multiple pathways and mechanisms. In addition, autophagy, a protective mechanism, regulates the NLRP3 inflammasome to control the progression of POCD. This review reviews recent findings on the role of the NLRP3 inflammasome in POCD pathogenesis and discusses therapeutic strategies aimed at reducing NLRP3 sources, inhibiting cellular pyroptosis, and enhancing autophagy.
Collapse
Affiliation(s)
| | | | - Bingdong Tao
- Department of Anesthesiology, Shengjing Hospital of China Medical University, Shenyang, China
| |
Collapse
|
9
|
Kim Y, Lee S, Park YH. NLRP3 Negative Regulation Mechanisms in the Resting State and Its Implications for Therapeutic Development. Int J Mol Sci 2024; 25:9018. [PMID: 39201704 PMCID: PMC11354250 DOI: 10.3390/ijms25169018] [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: 07/17/2024] [Revised: 08/16/2024] [Accepted: 08/16/2024] [Indexed: 09/03/2024] Open
Abstract
The NACHT-, leucine-rich-repeat-, and pyrin domain-containing protein 3 (NLRP3) is a critical intracellular sensor of the innate immune system that detects various pathogen- and danger-associated molecular patterns, leading to the assembly of the NLRP3 inflammasome and release of interleukin (IL) 1β and IL-18. However, the abnormal activation of the NLRP3 inflammasome has been implicated in the pathogenesis of autoinflammatory diseases such as cryopyrin-associated autoinflammatory syndromes (CAPS) and common diseases such as Alzheimer's disease and asthma. Recent studies have revealed that pyrin functions as an indirect sensor, similar to the plant guard system, and is regulated by binding to inhibitory 14-3-3 proteins. Upon activation, pyrin transitions to its active form. NLRP3 is predicted to follow a similar regulatory mechanism and maintain its inactive form in the cage model, as it also acts as an indirect sensor. Additionally, newly developed NLRP3 inhibitors have been found to inhibit NLRP3 activity by stabilizing its inactive form. Most studies and reviews on NLRP3 have focused on the activation of the NLRP3 inflammasome. This review highlights the molecular mechanisms that regulate NLRP3 in its resting state, and discusses how targeting this inhibitory mechanism can lead to novel therapeutic strategies for NLRP3-related diseases.
Collapse
Affiliation(s)
- YeJi Kim
- Department of Microbiology, Ajou University School of Medicine, Suwon 16499, Republic of Korea; (Y.K.); (S.L.)
- Department of Biomedical Sciences, Graduate School of Ajou University, Suwon 16499, Republic of Korea
| | - Sumin Lee
- Department of Microbiology, Ajou University School of Medicine, Suwon 16499, Republic of Korea; (Y.K.); (S.L.)
- Department of Biomedical Sciences, Graduate School of Ajou University, Suwon 16499, Republic of Korea
| | - Yong Hwan Park
- Department of Microbiology, Ajou University School of Medicine, Suwon 16499, Republic of Korea; (Y.K.); (S.L.)
- Department of Biomedical Sciences, Graduate School of Ajou University, Suwon 16499, Republic of Korea
| |
Collapse
|
10
|
Cescato M, Zhu YYJ, Le Corre L, Py BF, Georgin-Lavialle S, Rodero MP. Implication of the LRR Domain in the Regulation and Activation of the NLRP3 Inflammasome. Cells 2024; 13:1365. [PMID: 39195255 PMCID: PMC11352923 DOI: 10.3390/cells13161365] [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: 07/09/2024] [Revised: 08/02/2024] [Accepted: 08/04/2024] [Indexed: 08/29/2024] Open
Abstract
The NLRP3 inflammasome is a critical component of the innate immune response. NLRP3 activation is a tightly controlled process involving an initial priming to express NLRP3, pro-IL-1 β, and pro-IL-18, followed by an activation signal. The precise mechanism of activation is not fully understood due to the diverse range of activators, yet it effectively orchestrates the activation of caspase-1, which subsequently triggers the release of proinflammatory cytokines IL-1β and IL-18. NLRP3 dysregulation can lead to a variety of inflammatory diseases, highlighting its significant role in immune response and disease pathogenesis. NLRP3 is divided into three domains: the PYD, the NACHT, and the LRR domains. This review focuses on the LRR domain of NLRP3, detailing its structural characteristics, its function in pathogen sensing, its role in the degradation process, and its involvement in inflammasome auto-inhibition and activation. Additionally, we discuss the impact of mutations within the LRR domain found in atypical Cryopyrin-Associated Periodic Syndromes (CAPS), highlighting the clinical relevance of this domain.
Collapse
Affiliation(s)
- Margaux Cescato
- Laboratory of Pharmacological and Toxicological Chemistry and Biochemistry, CNRS, Paris Cité University, 75006 Paris, France; (M.C.); (Y.Y.J.Z.); (L.L.C.)
| | - Yixiang Y J Zhu
- Laboratory of Pharmacological and Toxicological Chemistry and Biochemistry, CNRS, Paris Cité University, 75006 Paris, France; (M.C.); (Y.Y.J.Z.); (L.L.C.)
- National Reference Center for Autoinflammatory Diseases and AA Amyloidosis, Department of Internal Medicine, Tenon Hospital, Sorbonne University, Assistance Publique—Hôpitaux de Paris (APHP), 75020 Paris, France;
| | - Laurent Le Corre
- Laboratory of Pharmacological and Toxicological Chemistry and Biochemistry, CNRS, Paris Cité University, 75006 Paris, France; (M.C.); (Y.Y.J.Z.); (L.L.C.)
| | - Bénédicte F Py
- CIRI, International Center for Research in Infectiology, Inserm, University Claude Bernard Lyon 1, 69007 Lyon, France;
| | - Sophie Georgin-Lavialle
- National Reference Center for Autoinflammatory Diseases and AA Amyloidosis, Department of Internal Medicine, Tenon Hospital, Sorbonne University, Assistance Publique—Hôpitaux de Paris (APHP), 75020 Paris, France;
| | - Mathieu P Rodero
- Laboratory of Pharmacological and Toxicological Chemistry and Biochemistry, CNRS, Paris Cité University, 75006 Paris, France; (M.C.); (Y.Y.J.Z.); (L.L.C.)
| |
Collapse
|
11
|
Henedak NT, El-Abhar HS, Soubh AA, Abdallah DM. NLRP3 Inflammasome: A central player in renal pathologies and nephropathy. Life Sci 2024; 351:122813. [PMID: 38857655 DOI: 10.1016/j.lfs.2024.122813] [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: 02/12/2024] [Revised: 05/16/2024] [Accepted: 06/04/2024] [Indexed: 06/12/2024]
Abstract
The cytoplasmic oligomer NLR Family Pyrin Domain Containing 3 (NLRP3) inflammasome has been implicated in most inflammatory and autoimmune diseases. Here, we highlight the significance of NLRP3 in diverse renal disorders, demonstrating its activation in macrophages and non-immune tubular epithelial and mesangial cells in response to various stimuli. This activation leads to the release of pro-inflammatory cytokines, contributing to the development of acute kidney injury (AKI), chronic renal injury, or fibrosis. In AKI, NLRP3 inflammasome activation and pyroptotic renal tubular cell death is driven by contrast and chemotherapeutic agents, sepsis, and rhabdomyolysis. Nevertheless, inflammasome is provoked in disorders such as crystal and diabetic nephropathy, obesity-related renal fibrosis, lupus nephritis, and hypertension-induced renal damage that induce chronic kidney injury and/or fibrosis. The mechanisms by which the inflammatory NLRP3/ Apoptosis-associated Speck-like protein containing a Caspase recruitment domain (ASC)/caspase-1/interleukin (IL)-1β & IL-18 pathway can turn on renal fibrosis is also comprehended. This review further outlines the involvement of dopamine and its associated G protein-coupled receptors (GPCRs), including D1-like (D1, D5) and D2-like (D2-D4) subtypes, in regulating this inflammation-linked renal dysfunction pathway. Hence, we identify D-related receptors as promising targets for renal disease management by inhibiting the functionality of the NLRP3 inflammasome.
Collapse
Affiliation(s)
- Nada T Henedak
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Ahram Canadian University, 6(th) of October City, Giza, Egypt
| | - Hanan S El-Abhar
- Department of Pharmacology, Toxicology, and Biochemistry, Faculty of Pharmacy, Future University in Egypt, Cairo 11835, Egypt
| | - Ayman A Soubh
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Ahram Canadian University, 6(th) of October City, Giza, Egypt
| | - Dalaal M Abdallah
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Cairo 11562, Egypt.
| |
Collapse
|
12
|
Gu W, Wu G, Chen G, Meng X, Xie Z, Cai S. Polyphenols alleviate metabolic disorders: the role of ubiquitin-proteasome system. Front Nutr 2024; 11:1445080. [PMID: 39188976 PMCID: PMC11345163 DOI: 10.3389/fnut.2024.1445080] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2024] [Accepted: 07/04/2024] [Indexed: 08/28/2024] Open
Abstract
Metabolic disorders include obesity, nonalcoholic fatty liver disease, insulin resistance and type 2 diabetes. It has become a major health issue around the world. Ubiquitin-proteasome system (UPS) is essential for nearly all cellular processes, functions as a primary pathway for intracellular protein degradation. Recent researches indicated that dysfunctions in the UPS may result in the accumulation of toxic proteins, lipotoxicity, oxidative stress, inflammation, and insulin resistance, all of which contribute to the development and progression of metabolic disorders. An increasing body of evidence indicates that specific dietary polyphenols ameliorate metabolic disorders by preventing lipid synthesis and transport, excessive inflammation, hyperglycemia and insulin resistance, and oxidative stress, through regulation of the UPS. This review summarized the latest research progress of natural polyphenols improving metabolic disorders by regulating lipid accumulation, inflammation, oxidative stress, and insulin resistance through the UPS. In addition, the possible mechanisms of UPS-mediated prevention of metabolic disorders are comprehensively proposed. We aim to provide new angle to the development and utilization of polyphenols in improving metabolic disorders.
Collapse
Affiliation(s)
- Wei Gu
- State Key Laboratory of Tea Plant Biology and Utilization, School of Tea and Food Sciences and Technology, Anhui Agricultural University, Hefei, Anhui, China
- Joint Research Center for Food Nutrition and Health of IHM, Anhui Agricultural University, Hefei, Anhui, China
| | - Guohuo Wu
- State Key Laboratory of Tea Plant Biology and Utilization, School of Tea and Food Sciences and Technology, Anhui Agricultural University, Hefei, Anhui, China
- Joint Research Center for Food Nutrition and Health of IHM, Anhui Agricultural University, Hefei, Anhui, China
| | - Guijie Chen
- State Key Laboratory of Tea Plant Biology and Utilization, School of Tea and Food Sciences and Technology, Anhui Agricultural University, Hefei, Anhui, China
- Joint Research Center for Food Nutrition and Health of IHM, Anhui Agricultural University, Hefei, Anhui, China
| | - Xianghui Meng
- The First Affiliated Hospital of University of Science and Technology of China, Hefei, Anhui, China
| | - Zhongwen Xie
- State Key Laboratory of Tea Plant Biology and Utilization, School of Tea and Food Sciences and Technology, Anhui Agricultural University, Hefei, Anhui, China
- Joint Research Center for Food Nutrition and Health of IHM, Anhui Agricultural University, Hefei, Anhui, China
| | - Shanbao Cai
- The First Affiliated Hospital of University of Science and Technology of China, Hefei, Anhui, China
| |
Collapse
|
13
|
Chen P, Li X. NLRP3 inflammasome in atherosclerosis: Mechanisms and targeted therapies. Front Pharmacol 2024; 15:1430236. [PMID: 39144618 PMCID: PMC11322363 DOI: 10.3389/fphar.2024.1430236] [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: 05/09/2024] [Accepted: 07/17/2024] [Indexed: 08/16/2024] Open
Abstract
Atherosclerosis (AS) is the primary pathology behind various cardiovascular diseases and the leading cause of death and disability globally. Recent evidence suggests that AS is a chronic vascular inflammatory disease caused by multiple factors. In this context, the NLRP3 inflammasome, acting as a signal transducer of the immune system, plays a critical role in the onset and progression of AS. The NLRP3 inflammasome is involved in endothelial injury, foam cell formation, and pyroptosis in AS. Therefore, targeting the NLRP3 inflammasome offers a new treatment strategy for AS. This review highlights the latest insights into AS pathogenesis and the pharmacological therapies targeting the NLRP3 inflammasome, focusing on optimal targets for small molecule inhibitors. These insights are valuable for rational drug design and the pharmacological assessment of new targeted NLRP3 inflammasome inhibitors in treating AS.
Collapse
Affiliation(s)
- Pengfei Chen
- Marine College, Shandong University, Weihai, China
| | - Xia Li
- Marine College, Shandong University, Weihai, China
- Shandong Kelun Pharmaceutical Co, Ltd., Binzhou, China
| |
Collapse
|
14
|
Liu F, Chen J, Li K, Li H, Zhu Y, Zhai Y, Lu B, Fan Y, Liu Z, Chen X, Jia X, Dong Z, Liu K. Ubiquitination and deubiquitination in cancer: from mechanisms to novel therapeutic approaches. Mol Cancer 2024; 23:148. [PMID: 39048965 PMCID: PMC11270804 DOI: 10.1186/s12943-024-02046-3] [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: 04/17/2024] [Accepted: 06/15/2024] [Indexed: 07/27/2024] Open
Abstract
Ubiquitination, a pivotal posttranslational modification of proteins, plays a fundamental role in regulating protein stability. The dysregulation of ubiquitinating and deubiquitinating enzymes is a common feature in various cancers, underscoring the imperative to investigate ubiquitin ligases and deubiquitinases (DUBs) for insights into oncogenic processes and the development of therapeutic interventions. In this review, we discuss the contributions of the ubiquitin-proteasome system (UPS) in all hallmarks of cancer and progress in drug discovery. We delve into the multiple functions of the UPS in oncology, including its regulation of multiple cancer-associated pathways, its role in metabolic reprogramming, its engagement with tumor immune responses, its function in phenotypic plasticity and polymorphic microbiomes, and other essential cellular functions. Furthermore, we provide a comprehensive overview of novel anticancer strategies that leverage the UPS, including the development and application of proteolysis targeting chimeras (PROTACs) and molecular glues.
Collapse
Affiliation(s)
- Fangfang Liu
- Tianjian Laboratory of Advanced Biomedical Sciences, Academy of Medical Sciences, College of Medicine, Zhengzhou University, Zhengzhou, Henan, 450001, China
- China-US (Henan) Hormel Cancer Institute, Zhengzhou, Henan, 450000, China
| | - Jingyu Chen
- Department of Pediatric Medicine, School of Third Clinical Medical Sciences, Zhengzhou University, Zhengzhou, 450000, China
| | - Kai Li
- Department of Clinical Medicine, School of First Clinical Medical Sciences, Zhengzhou University, Zhengzhou, 450000, China
| | - Haochen Li
- Department of Clinical Medicine, School of First Clinical Medical Sciences, Zhengzhou University, Zhengzhou, 450000, China
| | - Yiyi Zhu
- Department of Clinical Medicine, School of First Clinical Medical Sciences, Zhengzhou University, Zhengzhou, 450000, China
| | - Yubo Zhai
- Department of Pathophysiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450000, China
| | - Bingbing Lu
- Department of Pathophysiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450000, China
| | - Yanle Fan
- China-US (Henan) Hormel Cancer Institute, Zhengzhou, Henan, 450000, China
| | - Ziyue Liu
- Department of Pathophysiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450000, China
| | - Xiaojie Chen
- School of Basic Medicine, Henan University of Science and Technology, Luoyang, China
| | - Xuechao Jia
- Henan International Joint Laboratory of TCM Syndrome and Prescription in Signaling, Traditional Chinese Medicine (Zhong Jing) School, Henan University of Chinese Medicine, Zhengzhou, Henan, China.
| | - Zigang Dong
- Tianjian Laboratory of Advanced Biomedical Sciences, Academy of Medical Sciences, College of Medicine, Zhengzhou University, Zhengzhou, Henan, 450001, China.
- China-US (Henan) Hormel Cancer Institute, Zhengzhou, Henan, 450000, China.
- Department of Pathophysiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450000, China.
| | - Kangdong Liu
- Tianjian Laboratory of Advanced Biomedical Sciences, Academy of Medical Sciences, College of Medicine, Zhengzhou University, Zhengzhou, Henan, 450001, China.
- Department of Pathophysiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450000, China.
| |
Collapse
|
15
|
Ramachandran R, Manan A, Kim J, Choi S. NLRP3 inflammasome: a key player in the pathogenesis of life-style disorders. Exp Mol Med 2024; 56:1488-1500. [PMID: 38945951 PMCID: PMC11297159 DOI: 10.1038/s12276-024-01261-8] [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: 11/10/2023] [Revised: 02/27/2024] [Accepted: 03/25/2024] [Indexed: 07/02/2024] Open
Abstract
Proinflammatory cytokines and chemokines play a crucial role in regulating the inflammatory response, which is essential for the proper functioning of our immune system. When infections or threats to the body's defense mechanisms are detected, the innate immune system takes the lead. However, an excessive inflammatory response can lead to the production of high concentrations of cytotoxic molecules, resulting in tissue damage. Inflammasomes are significant contributors to innate immunity, and one of the most extensively studied inflammasome complexes is NOD-like receptor 3 (NLRP3). NLRP3 has a wide range of recognition mechanisms that streamline immune activation and eliminate pathogens. These cytosolic multiprotein complexes are composed of effector, adaptor, and sensor proteins, which are crucial for identifying intracellular bacterial breakdown products and initiating an innate immune cascade. To understand the diverse behavior of NLRP3 activation and its significance in the development of lifestyle-related diseases, one must delve into the study of the immune response and apoptosis mediated by the release of proinflammatory cytokines. In this review, we briefly explore the immune response in the context of lifestyle associated disorders such as obesity, hyperlipidemia, diabetes, chronic respiratory disease, oral disease, and cardiovascular disease.
Collapse
Affiliation(s)
- Rajath Ramachandran
- Department of Molecular Science and Technology, Ajou University, Suwon, 16499, Korea.
| | - Abdul Manan
- Department of Molecular Science and Technology, Ajou University, Suwon, 16499, Korea
| | - Jei Kim
- Department of Molecular Science and Technology, Ajou University, Suwon, 16499, Korea
- S&K Therapeutics, Ajou University Campus Plaza 418, 199 Worldcup-ro, Yeongtong-gu, Suwon, 16502, Korea
| | - Sangdun Choi
- Department of Molecular Science and Technology, Ajou University, Suwon, 16499, Korea.
- S&K Therapeutics, Ajou University Campus Plaza 418, 199 Worldcup-ro, Yeongtong-gu, Suwon, 16502, Korea.
| |
Collapse
|
16
|
Kuang S, Sheng W, Meng J, Liu W, Xiao Y, Tang H, Fu X, Kuang M, He Q, Gao S. Pyroptosis-related crosstalk in osteoarthritis: Macrophages, fibroblast-like synoviocytes and chondrocytes. J Orthop Translat 2024; 47:223-234. [PMID: 39040491 PMCID: PMC11262125 DOI: 10.1016/j.jot.2024.06.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/21/2024] [Revised: 04/28/2024] [Accepted: 06/20/2024] [Indexed: 07/24/2024] Open
Abstract
The pathogenesis of osteoarthritis (OA) involves a multifaceted interplay of inflammatory processes. The initiation of pyroptosis involves the secretion of pro-inflammatory cytokines and has been identified as a critical factor in regulating the development of OA. Upon initiation of pyroptosis, a multitude of inflammatory mediators are released and can be disseminated throughout the synovial fluid within the joint cavity, thereby facilitating intercellular communication across the entire joint. The main cellular components of joints include chondrocytes (CC), fibroblast-like synoviocytes (FLS) and macrophages (MC). Investigating their interplay can enhance our understanding of OA pathogenesis. Therefore, we comprehensively examine the mechanisms underlying pyroptosis and specifically investigate the intercellular interactions associated with pyroptosis among these three cell types, thereby elucidating their collective contribution to the progression of OA. We propose the concept of ' CC-FLS-MC pyroptosis-related crosstalk', describe the various pathways of pyroptotic interactions among these three cell types, and focus on recent advances in intervening pyroptosis in these three cell types for treating OA. We hope this will provide a possible direction for diversification of treatment for OA. The Translational potential of this article. The present study introduces the concept of 'MC-FLS-CC pyroptosis-related crosstalk' and provides an overview of the mechanisms underlying pyroptosis, as well as the pathways through which it affects MC, FLS, and CC. In addition, the role of regulation of these three types of cellular pyroptosis in OA has also been concerned. This review offers novel insights into the interplay between these cell types, with the aim of providing a promising avenue for diversified management of OA.
Collapse
Affiliation(s)
- Shida Kuang
- College of Traditional Chinese Medicine, Hunan University of Chinese Medicine, Changsha, China
- Andrology Laboratory, Hunan University of Chinese Medicine, Changsha, China
- Hunan University of Medicine, Huaihua, Hunan, China
| | - Wen Sheng
- College of Traditional Chinese Medicine, Hunan University of Chinese Medicine, Changsha, China
- Andrology Laboratory, Hunan University of Chinese Medicine, Changsha, China
- Hunan University of Medicine, Huaihua, Hunan, China
| | - Jiahao Meng
- Department of Orthopaedics, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Hunan Key Laboratory of Joint Degeneration and Injury, Changsha, Hunan, China
- Hunan Engineering Research Center of Osteoarthritis, Changsha, Hunan, China
- National Clinical Research Center of Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Weijie Liu
- Department of Orthopaedics, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Hunan Key Laboratory of Joint Degeneration and Injury, Changsha, Hunan, China
- Hunan Engineering Research Center of Osteoarthritis, Changsha, Hunan, China
- National Clinical Research Center of Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Yifan Xiao
- Department of Orthopaedics, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Hunan Key Laboratory of Joint Degeneration and Injury, Changsha, Hunan, China
- Hunan Engineering Research Center of Osteoarthritis, Changsha, Hunan, China
- National Clinical Research Center of Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Hang Tang
- Department of Orthopaedics, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Hunan Key Laboratory of Joint Degeneration and Injury, Changsha, Hunan, China
- Hunan Engineering Research Center of Osteoarthritis, Changsha, Hunan, China
- National Clinical Research Center of Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Xinying Fu
- College of Traditional Chinese Medicine, Hunan University of Chinese Medicine, Changsha, China
- Andrology Laboratory, Hunan University of Chinese Medicine, Changsha, China
- Hunan University of Medicine, Huaihua, Hunan, China
| | - Min Kuang
- College of Traditional Chinese Medicine, Hunan University of Chinese Medicine, Changsha, China
- Andrology Laboratory, Hunan University of Chinese Medicine, Changsha, China
- Hunan University of Medicine, Huaihua, Hunan, China
| | - Qinghu He
- College of Traditional Chinese Medicine, Hunan University of Chinese Medicine, Changsha, China
- Andrology Laboratory, Hunan University of Chinese Medicine, Changsha, China
- Hunan University of Medicine, Huaihua, Hunan, China
| | - Shuguang Gao
- Department of Orthopaedics, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Hunan Key Laboratory of Joint Degeneration and Injury, Changsha, Hunan, China
- Hunan Engineering Research Center of Osteoarthritis, Changsha, Hunan, China
- National Clinical Research Center of Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
| |
Collapse
|
17
|
Xu D, Jiang J, Liu Y, Pang J, Suo J, Li Y, Peng Z. TIMP2 protects against sepsis-associated acute kidney injury by cAMP/NLRP3 axis-mediated pyroptosis. Am J Physiol Cell Physiol 2024; 326:C1353-C1366. [PMID: 38497110 DOI: 10.1152/ajpcell.00577.2023] [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: 11/01/2023] [Revised: 03/12/2024] [Accepted: 03/13/2024] [Indexed: 03/19/2024]
Abstract
The tissue inhibitor of metalloproteinases 2 (TIMP2) has emerged as a promising biomarker for predicting the risk of sepsis-associated acute kidney injury (SA-AKI). However, its exact role in SA-AKI and the underlying mechanism remains unclear. In this study, we investigated the impact of kidney tubule-specific Timp2 knockout mice on kidney injury and inflammation. Our findings demonstrated that Timp2-knockout mice exhibited more severe kidney injury than wild-type mice, along with elevated levels of pyroptosis markers NOD-like receptor protein 3 (NLRP3), Caspase1, and gasdermin D (GSDMD) in the early stage of SA-AKI. Conversely, the expression of exogenous TIMP2 in TIMP2-knockout mice still protected against kidney damage and inflammation. In in vitro experiments, using recombinant TIMP2 protein, TIMP2 knockdown demonstrated that exogenous TIMP2 inhibited pyroptosis of renal tubular cells stimulated by lipopolysaccharide (LPS). Mechanistically, TIMP2 promoted the ubiquitination and autophagy-dependent degradation of NLRP3 by increasing intracellular cyclic adenosine monophosphate (cAMP), which mediated NLRP3 degradation through recruiting the E3 ligase MARCH7, attenuating downstream pyroptosis, and thus alleviating primary tubular cell damage. These results revealed the renoprotective role of extracellular TIMP2 in SA-AKI by attenuating tubular pyroptosis, and suggested that exogenous administration of TIMP2 could be a promising therapeutic intervention for SA-AKI treatment.NEW & NOTEWORTHY Tissue inhibitor of metalloproteinase 2 (TIMP-2) has been found to be the best biomarker for predicting the risk of sepsis-associated acute kidney injury (SA-AKI). However, its role and the underlying mechanism in SA-AKI remain elusive. The authors demonstrated in this study using kidney tubule-specific knockout mice model of SA-AKI and primary renal tubule cells stimulated with lipopolysaccharide (LPS) that extracellular TIMP-2 promoted NOD-like receptor protein 3 (NLRP3) ubiquitination and autophagy-dependent degradation by increasing intracellular cyclic adenosine monophosphate (cAMP), thus attenuated pyroptosis and alleviated renal damage.
Collapse
Affiliation(s)
- Dongxue Xu
- Department of Critical Care Medicine, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Jun Jiang
- Department of Critical Care Medicine, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Ye Liu
- Department of Critical Care Medicine, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Jingjing Pang
- Department of Critical Care Medicine, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Jinmeng Suo
- Department of Critical Care Medicine, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Yiming Li
- Department of Critical Care Medicine, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Zhiyong Peng
- Department of Critical Care Medicine, Zhongnan Hospital of Wuhan University, Wuhan, China
- Clinical Research Center of Hubei Critical Care Medicine, Wuhan, China
- Department of Critical Care Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, United States
- Intensive Care Unit of the second affiliated Hospital of Hainan Medical College, Haikou, China
| |
Collapse
|
18
|
Xu Z, Kombe Kombe AJ, Deng S, Zhang H, Wu S, Ruan J, Zhou Y, Jin T. NLRP inflammasomes in health and disease. MOLECULAR BIOMEDICINE 2024; 5:14. [PMID: 38644450 PMCID: PMC11033252 DOI: 10.1186/s43556-024-00179-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: 01/01/2024] [Accepted: 03/20/2024] [Indexed: 04/23/2024] Open
Abstract
NLRP inflammasomes are a group of cytosolic multiprotein oligomer pattern recognition receptors (PRRs) involved in the recognition of pathogen-associated molecular patterns (PAMPs) and danger-associated molecular patterns (DAMPs) produced by infected cells. They regulate innate immunity by triggering a protective inflammatory response. However, despite their protective role, aberrant NLPR inflammasome activation and gain-of-function mutations in NLRP sensor proteins are involved in occurrence and enhancement of non-communicating autoimmune, auto-inflammatory, and neurodegenerative diseases. In the last few years, significant advances have been achieved in the understanding of the NLRP inflammasome physiological functions and their molecular mechanisms of activation, as well as therapeutics that target NLRP inflammasome activity in inflammatory diseases. Here, we provide the latest research progress on NLRP inflammasomes, including NLRP1, CARD8, NLRP3, NLRP6, NLRP7, NLRP2, NLRP9, NLRP10, and NLRP12 regarding their structural and assembling features, signaling transduction and molecular activation mechanisms. Importantly, we highlight the mechanisms associated with NLRP inflammasome dysregulation involved in numerous human auto-inflammatory, autoimmune, and neurodegenerative diseases. Overall, we summarize the latest discoveries in NLRP biology, their forming inflammasomes, and their role in health and diseases, and provide therapeutic strategies and perspectives for future studies about NLRP inflammasomes.
Collapse
Affiliation(s)
- Zhihao Xu
- Center of Disease Immunity and Intervention, College of Medicine, Lishui University, Lishui, 323000, China
| | - Arnaud John Kombe Kombe
- Laboratory of Structural Immunology, the CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230027, China
| | - Shasha Deng
- Laboratory of Structural Immunology, the CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230027, China
| | - Hongliang Zhang
- Center of Disease Immunity and Intervention, College of Medicine, Lishui University, Lishui, 323000, China
| | - Songquan Wu
- Center of Disease Immunity and Intervention, College of Medicine, Lishui University, Lishui, 323000, China
| | - Jianbin Ruan
- Department of Immunology, University of Connecticut Health Center, Farmington, 06030, USA.
| | - Ying Zhou
- Department of Obstetrics and Gynecology, Core Facility Center, Division of Life Sciences and Medicine, The First Affiliated Hospital of USTC, University of Science and Technology of China, Hefei, 230001, Anhui, China.
| | - Tengchuan Jin
- Center of Disease Immunity and Intervention, College of Medicine, Lishui University, Lishui, 323000, China.
- Laboratory of Structural Immunology, the CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230027, China.
- Department of Obstetrics and Gynecology, Core Facility Center, Division of Life Sciences and Medicine, The First Affiliated Hospital of USTC, University of Science and Technology of China, Hefei, 230001, Anhui, China.
- Institute of Health and Medicine, Hefei Comprehensive National Science Center, Hefei, Anhui, China.
- Biomedical Sciences and Health Laboratory of Anhui Province, University of Science & Technology of China, Hefei, 230027, China.
- Clinical Research Hospital of Chinese Academy of Sciences (Hefei), University of Science and Technology of China, Hefei, 230001, China.
| |
Collapse
|
19
|
Ha J, Kim M, Park JS, Lee Y, Lee JY, Shin JC, Seo D, Park SS, You J, Jung SM, Kim HY, Mizuno S, Takahashi S, Kim SJ, Park SH. SERTAD1 initiates NLRP3-mediated inflammasome activation through restricting NLRP3 polyubiquitination. Cell Rep 2024; 43:113752. [PMID: 38341852 DOI: 10.1016/j.celrep.2024.113752] [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: 08/07/2023] [Revised: 01/03/2024] [Accepted: 01/23/2024] [Indexed: 02/13/2024] Open
Abstract
We here demonstrate that SERTAD1 is an adaptor protein responsible for the regulation of lysine 63 (K63)-linked NLRP3 polyubiquitination by the Cullin1 E3 ubiquitin ligase upon inflammasome activation. SERTAD1 specifically binds to NLRP3 but not to other inflammasome sensors. This endogenous interaction increases after inflammasome activation, interfering with the interaction between NLRP3 and Cullin1. Interleukin (IL)-1β and IL-18 secretion, as well as the cleavage of gasdermin D, are decreased in SERTAD1 knockout bone-marrow-derived macrophages, together with reduced formation of the NLRP3 inflammasome complex. Additionally, SERTAD1-deficient mice show attenuated severity of monosodium-uric-acid-induced peritonitis and experimental autoimmune encephalomyelitis. Analysis of public datasets indicates that expression of SERTAD1 mRNA is significantly increased in the patients of autoimmune diseases. Thus, our findings uncover a function of SERTAD1 that specifically reduces Cullin1-mediated NLRP3 polyubiquitination via direct binding to NLRP3, eventually acting as a crucial factor to regulate the initiation of NLRP3-mediated inflammasome activation.
Collapse
Affiliation(s)
- Jihoon Ha
- Department of Biological Sciences, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Minbeom Kim
- Department of Biological Sciences, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Jin Seok Park
- Department of Biological Sciences, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Yerin Lee
- Department of Biological Sciences, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Jae Young Lee
- Department of Biological Sciences, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Jin-Cheol Shin
- Department of Biological Sciences, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Dongyeob Seo
- Department of Biological Sciences, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Seong Shil Park
- Department of Biological Sciences, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Jiyeon You
- Department of Biological Sciences, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Su Myung Jung
- Department of Biological Sciences, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Hye Young Kim
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul 03080, Republic of Korea; SRC Center for Immune Research on Non-lymphoid Organs, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Seiya Mizuno
- Laboratory Animal Resource Center, Transborder Medical Research Center, Institute of Medicine, University of Tsukuba, Tsukuba, Ibaraki 305-8578, Japan
| | - Satoru Takahashi
- Laboratory Animal Resource Center, Transborder Medical Research Center, Institute of Medicine, University of Tsukuba, Tsukuba, Ibaraki 305-8578, Japan
| | - Seong-Jin Kim
- GILO Institute, GILO Foundation, Seoul 06668, Republic of Korea
| | - Seok Hee Park
- Department of Biological Sciences, Sungkyunkwan University, Suwon 16419, Republic of Korea; SRC Center for Immune Research on Non-lymphoid Organs, Sungkyunkwan University, Suwon 16419, Republic of Korea.
| |
Collapse
|
20
|
Zhang Z, Li M, Tai Y, Xing Y, Zuo H, Jin X, Ma J. ZNF70 regulates IL-1β secretion of macrophages to promote the proliferation of HCT116 cells via activation of NLRP3 inflammasome and STAT3 pathway in colitis-associated colorectal cancer. Cell Signal 2024; 114:110979. [PMID: 38000525 DOI: 10.1016/j.cellsig.2023.110979] [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: 06/20/2023] [Revised: 10/02/2023] [Accepted: 11/13/2023] [Indexed: 11/26/2023]
Abstract
Chronic inflammation is a key driver for colitis-associated colorectal cancer (CAC). It has been reported that inflammatory cytokines, such as IL-1β, could promote CAC. Zinc finger protein 70 (ZNF70) is involved in multiple biological processes. Here, we identified a previously unknown role for ZNF70 regulates macrophages IL-1β secretion to promote HCT116 proliferation in CAC, and investigated its underlying mechanism. We showed ZNF70 is much higher expressed in CAC tumor tissues compared with adjacent normal tissues in clinical CAC samples. Further experiments showed ZNF70 promoted macrophages IL-1β secretion and HCT116 proliferation. In LPS/ATP-stimulated THP-1 cells, we found ZNF70 activated NLRP3 inflammasome, resulting in robust IL-1β secretion. Interestingly, we discovered the ZnF domain of ZNF70 could interact with NLRP3 and decrease the K48-linked ubiquitination of NLRP3. Moreover, ZNF70 could activate STAT3, thereby promoting IL-1β synthesis. Noteworthy, ZNF70 enhanced proliferation by upregulating STAT3 activation in HCT116 cells cultured in the conditioned medium of THP-1 macrophages treated with LPS/ATP. Finally, the vivo observations were confirmed using AAV-mediated ZNF70 knockdown, which improved colitis-associated colorectal cancer in the AOM/DSS model. The correlation between ZNF70 expression and overall survival/IL-1β expression in colorectal cancer was verified by TCGA database. Taken together, ZNF70 regulates macrophages IL-1β secretion to promote the HCT116 cells proliferation via activation of NLRP3 inflammasome and STAT3 pathway, suggesting that ZNF70 may be a promising preventive target for treating in CAC.
Collapse
Affiliation(s)
- Zhihong Zhang
- Key Laboratory of Natural Medicines of the Changbai Mountain, Ministry of Education, Molecular Medicine Research Center, College of Pharmacy, Yanbian University, Yanji 133002, Jilin Province, China; Department of Pharmacology, College of Pharmacy, Beihua University, East Binjiang Road, 3999, Jilin, China
| | - Mingyue Li
- Key Laboratory of Natural Medicines of the Changbai Mountain, Ministry of Education, Molecular Medicine Research Center, College of Pharmacy, Yanbian University, Yanji 133002, Jilin Province, China.
| | - Yi Tai
- Key Laboratory of Natural Medicines of the Changbai Mountain, Ministry of Education, Molecular Medicine Research Center, College of Pharmacy, Yanbian University, Yanji 133002, Jilin Province, China
| | - Yue Xing
- Key Laboratory of Natural Medicines of the Changbai Mountain, Ministry of Education, Molecular Medicine Research Center, College of Pharmacy, Yanbian University, Yanji 133002, Jilin Province, China
| | - Hongxiang Zuo
- Key Laboratory of Natural Medicines of the Changbai Mountain, Ministry of Education, Molecular Medicine Research Center, College of Pharmacy, Yanbian University, Yanji 133002, Jilin Province, China
| | - Xuejun Jin
- Key Laboratory of Natural Medicines of the Changbai Mountain, Ministry of Education, Molecular Medicine Research Center, College of Pharmacy, Yanbian University, Yanji 133002, Jilin Province, China.
| | - Juan Ma
- Key Laboratory of Natural Medicines of the Changbai Mountain, Ministry of Education, Molecular Medicine Research Center, College of Pharmacy, Yanbian University, Yanji 133002, Jilin Province, China.
| |
Collapse
|
21
|
Liu F, Gao C. Regulation of the Inflammasome Activation by Ubiquitination Machinery. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2024; 1466:123-134. [PMID: 39546140 DOI: 10.1007/978-981-97-7288-9_9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/17/2024]
Abstract
Inflammasomes are multiprotein complexes that assemble in response to the detection of stress- or infection-associated stimuli and lead to the activation of caspase-1 and consequent maturation of caspase-1 target molecules such as interleukin (IL)-1β and IL-18. Although inflammasome is the essential component of the innate immunity system to defense against insults, inappropriate or prolonged activation of inflammasome may be harmful and is associated with various diseases, e.g., gout, atherosclerosis, diabetes, and Alzheimer's disease. Therefore, regulating inflammasome activation is crucial for maintaining immune homeostasis. Studies have found that post-translational modifications (PTMs), e.g., ubiquitination and phosphorylation, are critical for inflammasome activation. Ubiquitination is an important form of post-translational modification of proteins that plays a pivotal role in various cellular functions. In recent years, its function in regulating inflammasome assembly has been a hot topic of interest. This study discussed the function and mechanism of the ubiquitin system controlling inflammasome activation and highlighted the challenges of this research area.
Collapse
Affiliation(s)
- Feng Liu
- Key Laboratory of Infection and Immunity of Shandong Province, Shandong University, Jinan, Shandong, P.R. China
- Department of Immunology, School of Biomedical Sciences, Shandong University, Jinan, Shandong, P.R. China
| | - Chengjiang Gao
- Key Laboratory of Infection and Immunity of Shandong Province, Shandong University, Jinan, Shandong, P.R. China.
- Department of Immunology, School of Biomedical Sciences, Shandong University, Jinan, Shandong, P.R. China.
| |
Collapse
|
22
|
Wang P, Tang CT, Li J, Huang X, Jin R, Yin F, Liu Z, Chen Y, Zeng C. The E3 ubiquitin ligase RNF31 mediates the development of ulcerative colitis by regulating NLRP3 inflammasome activation. Int Immunopharmacol 2023; 125:111194. [PMID: 37951199 DOI: 10.1016/j.intimp.2023.111194] [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: 07/21/2023] [Revised: 11/04/2023] [Accepted: 11/05/2023] [Indexed: 11/13/2023]
Abstract
Ulcerative colitis (UC) is characterized by dysregulated inflammation and disruption of the intestinal barrier. The NLRP3 inflammasome, which is composed of NLRP3, ASC, and caspase-1, plays a crucial role in UC pathogenesis by triggering the production of proinflammatory cytokines. In this study, we investigated the regulatory role of RNF31 in NLRP3 inflammasome activation during UC development. Through comprehensive analysis of ulcerative colitis tissues using the GEO database and immunohistochemistry, we found that RNF31 expression was elevated in UC tissues, which prompted further investigation into its function. We constructed an RNF31 knockdown cell model and observed a significant reduction in NLRP3 inflammasome activation, indicating the involvement of RNF31 in regulating NLRP3. Mechanistically, RNF31 could interact with NLRP3 through the RBR structural domain, leading to increased K63-linked ubiquitination of NLRP3 and consequent stabilization. Coimmunoprecipitation experiments revealed a mutual interaction between RNF31 and NLRP3, substantiating their functional association. Finally, an in vivo mouse model with RNF31 knockdown showed a notable reduction in NLRP3 expression, which was accompanied by a decrease in the proinflammatory cytokines IL-18 and IL-1β. The successful attenuation of DSS-induced tissue inflammation by this treatment confirmed the physiological relevance of RNF31-mediated regulation of NLRP3. This study unveils a novel regulatory pathway by which RNF31 affects NLRP3 inflammasome activation, providing new insights into UC pathogenesis and potential therapeutic targets for UC treatment.
Collapse
Affiliation(s)
- Peng Wang
- Department of Gastroenterology, Digestive Disease Hospital, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Chao-Tao Tang
- Department of Gastroenterology, Digestive Disease Hospital, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China; Jiangxi Clinical Research Center for Gastroenterology, Nanchang, Jiangxi, China
| | - Jun Li
- Department of Gastroenterology, Digestive Disease Hospital, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Xia Huang
- The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Ruiri Jin
- Department of Gastroenterology, Digestive Disease Hospital, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Fang Yin
- Department of Gastroenterology, Digestive Disease Hospital, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Zide Liu
- Department of Gastroenterology, Digestive Disease Hospital, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Youxiang Chen
- Department of Gastroenterology, Digestive Disease Hospital, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China; Jiangxi Clinical Research Center for Gastroenterology, Nanchang, Jiangxi, China
| | - Chunyan Zeng
- Department of Gastroenterology, Digestive Disease Hospital, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China; Jiangxi Clinical Research Center for Gastroenterology, Nanchang, Jiangxi, China.
| |
Collapse
|
23
|
Xu J, Guo R, Wen N, Li L, Yi Y, Chen J, He Z, Yang J, Xiao ZXJ, Niu M. FBXO3 stabilizes USP4 and Twist1 to promote PI3K-mediated breast cancer metastasis. PLoS Biol 2023; 21:e3002446. [PMID: 38134227 PMCID: PMC10745200 DOI: 10.1371/journal.pbio.3002446] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2023] [Accepted: 11/24/2023] [Indexed: 12/24/2023] Open
Abstract
Tumor metastasis is the major cause of breast cancer morbidity and mortality. It has been reported that the F-box protein FBXO3 functions as an E3 ubiquitin ligase in regulating various biological processes, including host autoimmune, antiviral innate immunity, and inflammatory response. However, the role of FBXO3 in tumor metastasis remains elusive. We have previously shown that ΔNp63α is a common inhibitory target in oncogene-induced cell motility and tumor metastasis. In this study, we show that FBXO3 plays a vital role in PI3K-mediated breast cancer metastasis independent of its E3 ligase activity and ΔNp63α in breast cancer cells and in mouse. FBXO3 can bind to and stabilize USP4, leading to Twist1 protein stabilization and increased breast cancer cell migration and tumor metastasis. Mechanistically, FBXO3 disrupts the interaction between USP4 and aspartyl aminopeptidase (DNPEP), thereby protecting USP4 from DNPEP-mediated degradation. Furthermore, p110αH1047R facilitates the phosphorylation and stabilization of FBXO3 in an ERK1-dependent manner. Knockdown of either FBXO3 or USP4 leads to significant inhibition of PI3K-induced breast cancer metastasis. Clinically, elevated expression of p110α/FBXO3/USP4/Twist1 is associated with poor overall survival (OS) and recurrence-free survival (RFS) of breast cancer patients. Taken together, this study reveals that the FBXO3-USP4-Twist1 axis is pivotal in PI3K-mediated breast tumor metastasis and that FBXO3/USP4 may be potential therapeutic targets for breast cancer treatment.
Collapse
Affiliation(s)
- Jing Xu
- Center of Growth, Metabolism and Aging, Key Laboratory of Bio-Resource and Eco-Environment, Ministry of Education, College of Life Sciences, Sichuan University, Chengdu, China
| | - Rongtian Guo
- Center of Growth, Metabolism and Aging, Key Laboratory of Bio-Resource and Eco-Environment, Ministry of Education, College of Life Sciences, Sichuan University, Chengdu, China
| | - Nasi Wen
- Center of Growth, Metabolism and Aging, Key Laboratory of Bio-Resource and Eco-Environment, Ministry of Education, College of Life Sciences, Sichuan University, Chengdu, China
| | - Luping Li
- Center of Growth, Metabolism and Aging, Key Laboratory of Bio-Resource and Eco-Environment, Ministry of Education, College of Life Sciences, Sichuan University, Chengdu, China
| | - Yong Yi
- Center of Growth, Metabolism and Aging, Key Laboratory of Bio-Resource and Eco-Environment, Ministry of Education, College of Life Sciences, Sichuan University, Chengdu, China
| | - Jingzhen Chen
- Center of Growth, Metabolism and Aging, Key Laboratory of Bio-Resource and Eco-Environment, Ministry of Education, College of Life Sciences, Sichuan University, Chengdu, China
| | - Zongyu He
- Center of Growth, Metabolism and Aging, Key Laboratory of Bio-Resource and Eco-Environment, Ministry of Education, College of Life Sciences, Sichuan University, Chengdu, China
| | - Jian Yang
- Center of Growth, Metabolism and Aging, Key Laboratory of Bio-Resource and Eco-Environment, Ministry of Education, College of Life Sciences, Sichuan University, Chengdu, China
| | - Zhi-Xiong Jim Xiao
- Center of Growth, Metabolism and Aging, Key Laboratory of Bio-Resource and Eco-Environment, Ministry of Education, College of Life Sciences, Sichuan University, Chengdu, China
| | - Mengmeng Niu
- Center of Growth, Metabolism and Aging, Key Laboratory of Bio-Resource and Eco-Environment, Ministry of Education, College of Life Sciences, Sichuan University, Chengdu, China
| |
Collapse
|
24
|
Krantz M, Eklund D, Särndahl E, Hedbrant A. A detailed molecular network map and model of the NLRP3 inflammasome. Front Immunol 2023; 14:1233680. [PMID: 38077364 PMCID: PMC10699087 DOI: 10.3389/fimmu.2023.1233680] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2023] [Accepted: 10/16/2023] [Indexed: 12/18/2023] Open
Abstract
The NLRP3 inflammasome is a key regulator of inflammation that responds to a broad range of stimuli. The exact mechanism of activation has not been determined, but there is a consensus on cellular potassium efflux as a major common denominator. Once NLRP3 is activated, it forms high-order complexes together with NEK7 that trigger aggregation of ASC into specks. Typically, there is only one speck per cell, consistent with the proposal that specks form - or end up at - the centrosome. ASC polymerisation in turn triggers caspase-1 activation, leading to maturation and release of IL-1β and pyroptosis, i.e., highly inflammatory cell death. Several gain-of-function mutations in the NLRP3 inflammasome have been suggested to induce spontaneous activation of NLRP3 and hence contribute to development and disease severity in numerous autoinflammatory and autoimmune diseases. Consequently, the NLRP3 inflammasome is of significant clinical interest, and recent attention has drastically improved our insight in the range of involved triggers and mechanisms of signal transduction. However, despite recent progress in knowledge, a clear and comprehensive overview of how these mechanisms interplay to shape the system level function is missing from the literature. Here, we provide such an overview as a resource to researchers working in or entering the field, as well as a computational model that allows for evaluating and explaining the function of the NLRP3 inflammasome system from the current molecular knowledge. We present a detailed reconstruction of the molecular network surrounding the NLRP3 inflammasome, which account for each specific reaction and the known regulatory constraints on each event as well as the mechanisms of drug action and impact of genetics when known. Furthermore, an executable model from this network reconstruction is generated with the aim to be used to explain NLRP3 activation from priming and activation to the maturation and release of IL-1β and IL-18. Finally, we test this detailed mechanistic model against data on the effect of different modes of inhibition of NLRP3 assembly. While the exact mechanisms of NLRP3 activation remains elusive, the literature indicates that the different stimuli converge on a single activation mechanism that is additionally controlled by distinct (positive or negative) priming and licensing events through covalent modifications of the NLRP3 molecule. Taken together, we present a compilation of the literature knowledge on the molecular mechanisms on NLRP3 activation, a detailed mechanistic model of NLRP3 activation, and explore the convergence of diverse NLRP3 activation stimuli into a single input mechanism.
Collapse
Affiliation(s)
- Marcus Krantz
- School of Medical Sciences, Faculty of Medicine and Health, Örebro University, Örebro, Sweden
- Inflammatory Response and Infection Susceptibility Centre (iRiSC), Örebro University, Örebro, Sweden
| | - Daniel Eklund
- School of Medical Sciences, Faculty of Medicine and Health, Örebro University, Örebro, Sweden
- Inflammatory Response and Infection Susceptibility Centre (iRiSC), Örebro University, Örebro, Sweden
| | - Eva Särndahl
- School of Medical Sciences, Faculty of Medicine and Health, Örebro University, Örebro, Sweden
- Inflammatory Response and Infection Susceptibility Centre (iRiSC), Örebro University, Örebro, Sweden
| | - Alexander Hedbrant
- School of Medical Sciences, Faculty of Medicine and Health, Örebro University, Örebro, Sweden
- Inflammatory Response and Infection Susceptibility Centre (iRiSC), Örebro University, Örebro, Sweden
| |
Collapse
|
25
|
Qin Y, Meng X, Wang M, Liang W, Xu R, Chen J, Song H, Fu Y, Li J, Gao C, Jia M, Zhao C, Zhao W. Posttranslational ISGylation of NLRP3 by HERC enzymes facilitates inflammasome activation in models of inflammation. J Clin Invest 2023; 133:e161935. [PMID: 37651190 PMCID: PMC10575725 DOI: 10.1172/jci161935] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Accepted: 08/24/2023] [Indexed: 09/02/2023] Open
Abstract
The NOD-, LRR-, and pyrin domain-containing protein 3 (NLRP3) inflammasome is a crucial component of the innate immune system that initiates inflammatory responses. Posttranslational modifications (PTMs) of NLRP3, including ubiquitination and phosphorylation, control inflammasome activation and determine the intensity of inflammation. However, the role of other PTMs in controlling NLRP3 inflammasome activation remains unclear. This study found that TLR priming induced NLRP3 ISGylation (a type of PTM in which ISG15 covalently binds to the target protein) to stabilize the NLRP3 protein. Viral infection, represented by SARS-COV-2 infection, and type I IFNs induced expression of ISG15 and the predominant E3 ISGylation ligases HECT domain- and RCC1-like domain-containing proteins (HERCs; HERC5 in humans and HERC6 in mice). HERCs promoted NLRP3 ISGylation and inhibited K48-linked ubiquitination and proteasomal degradation, resulting in the enhancement of NLRP3 inflammasome activation. Concordantly, Herc6 deficiency ameliorated NLRP3-dependent inflammation as well as hyperinflammation caused by viral infection. The results illustrate the mechanism by which type I IFNs responses control inflammasome activation and viral infection-induced aberrant NLRP3 activation. This work identifies ISGylation as a PTM of NLRP3, revealing a priming target that modulates NLRP3-dependent immunopathology.
Collapse
|
26
|
Qin Y, Zhao W. Posttranslational modifications of NLRP3 and their regulatory roles in inflammasome activation. Eur J Immunol 2023; 53:e2350382. [PMID: 37382218 DOI: 10.1002/eji.202350382] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2023] [Revised: 05/09/2023] [Accepted: 06/19/2023] [Indexed: 06/30/2023]
Abstract
The NACHT, LRR, and PYD domains-containing protein 3 (NLRP3) inflammasome is a multimolecular complex that plays a fundamental role in inflammation. Optimal activation of NLRP3 inflammasome is crucial for host defense against pathogens and the maintenance of immune homeostasis. Aberrant NLRP3 inflammasome activity has been implicated in various inflammatory diseases. Posttranslational modifications (PTMs) of NLRP3, a key inflammasome sensor, play critical roles in directing inflammasome activation and controlling the severity of inflammation and inflammatory diseases, such as arthritis, peritonitis, inflammatory bowel disease, atherosclerosis, and Parkinson's disease. Various NLRP3 PTMs, including phosphorylation, ubiquitination, and SUMOylation, could direct inflammasome activation and control inflammation severity by affecting the protein stability, ATPase activity, subcellular localization, and oligomerization of NLRP3 as well as the association between NLRP3 and other inflammasome components. Here, we provide an overview of the PTMs of NLRP3 and their roles in controlling inflammation and summarize potential anti-inflammatory drugs targeting NLRP3 PTMs.
Collapse
Affiliation(s)
- Ying Qin
- Department of Pathogenic Biology and Key Laboratory of Infection and Immunity of Shandong Province, School of Basic Medical Science, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Wei Zhao
- Department of Pathogenic Biology and Key Laboratory of Infection and Immunity of Shandong Province, School of Basic Medical Science, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| |
Collapse
|
27
|
Chen Y, Miao C, Zhao Y, Yang L, Wang R, Shen D, Ren N, Zhang Q. Inflammasomes in human reproductive diseases. Mol Hum Reprod 2023; 29:gaad035. [PMID: 37788097 DOI: 10.1093/molehr/gaad035] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2023] [Revised: 09/20/2023] [Indexed: 10/05/2023] Open
Abstract
Inflammasomes are multi-protein complexes localized within immune and non-immune cells that induce caspase activation, proinflammatory cytokine secretion, and ultimately pyroptosis-a type of cell death. Inflammasomes are involved in a variety of human diseases, especially acute or chronic inflammatory diseases. In this review, we focused on the strong correlation between the NLRP3 inflammasome and various reproductive diseases, including ovarian aging or premature ovarian insufficiency, PCOS, endometriosis, recurrent spontaneous abortion, preterm labor, pre-eclampsia, and male subfertility, as well as the multifaceted role of NLRP3 in the pathogenesis and treatment of these diseases. In addition, we provide an overview of the structure and amplification of inflammasomes. This comprehensive review demonstrates the vital role of NLRP3 inflammasome activation in human reproductive diseases together with the underlying mechanisms, offers new insights for mechanistic studies of reproduction, and provides promising possibilities for the development of drugs targeting the NLRP3 inflammasome for the treatment of reproductive disorders in the future.
Collapse
Affiliation(s)
- Yun Chen
- Department of TCM Gynecology, Hangzhou TCM Hospital affiliated to Zhejiang Chinese Medical University, Hangzhou, China
| | - Chenyun Miao
- Department of TCM Gynecology, Hangzhou TCM Hospital affiliated to Zhejiang Chinese Medical University, Hangzhou, China
| | - Ying Zhao
- Department of TCM Gynecology, Hangzhou TCM Hospital affiliated to Zhejiang Chinese Medical University, Hangzhou, China
| | - Liuqing Yang
- Department of TCM Gynecology, Hangzhou TCM Hospital affiliated to Zhejiang Chinese Medical University, Hangzhou, China
| | - Ruye Wang
- Department of TCM Gynecology, Hangzhou TCM Hospital affiliated to Zhejiang Chinese Medical University, Hangzhou, China
| | - Dan Shen
- Department of TCM Gynecology, Hangzhou TCM Hospital affiliated to Zhejiang Chinese Medical University, Hangzhou, China
| | - Ning Ren
- Department of TCM Gynecology, Hangzhou TCM Hospital affiliated to Zhejiang Chinese Medical University, Hangzhou, China
| | - Qin Zhang
- Department of TCM Gynecology, Hangzhou TCM Hospital affiliated to Zhejiang Chinese Medical University, Hangzhou, China
| |
Collapse
|
28
|
Hadian K, Stockwell BR. The therapeutic potential of targeting regulated non-apoptotic cell death. Nat Rev Drug Discov 2023; 22:723-742. [PMID: 37550363 DOI: 10.1038/s41573-023-00749-8] [Citation(s) in RCA: 134] [Impact Index Per Article: 67.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/16/2023] [Indexed: 08/09/2023]
Abstract
Cell death is critical for the development and homeostasis of almost all multicellular organisms. Moreover, its dysregulation leads to diverse disease states. Historically, apoptosis was thought to be the major regulated cell death pathway, whereas necrosis was considered to be an unregulated form of cell death. However, research in recent decades has uncovered several forms of regulated necrosis that are implicated in degenerative diseases, inflammatory conditions and cancer. The growing insight into these regulated, non-apoptotic cell death pathways has opened new avenues for therapeutic targeting. Here, we describe the regulatory pathways of necroptosis, pyroptosis, parthanatos, ferroptosis, cuproptosis, lysozincrosis and disulfidptosis. We discuss small-molecule inhibitors of the pathways and prospects for future drug discovery. Together, the complex mechanisms governing these pathways offer strategies to develop therapeutics that control non-apoptotic cell death.
Collapse
Affiliation(s)
- Kamyar Hadian
- Research Unit Signaling and Translation, Helmholtz Zentrum München, Neuherberg, Germany.
| | - Brent R Stockwell
- Department of Biological Sciences and Department of Chemistry, Columbia University, New York, NY, USA.
| |
Collapse
|
29
|
Han YH, Liu XD, Jin MH, Sun HN, Kwon T. Role of NLRP3 inflammasome-mediated neuronal pyroptosis and neuroinflammation in neurodegenerative diseases. Inflamm Res 2023; 72:1839-1859. [PMID: 37725102 DOI: 10.1007/s00011-023-01790-4] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Revised: 08/14/2023] [Accepted: 08/17/2023] [Indexed: 09/21/2023] Open
Abstract
BACKGROUND Neurodegenerative diseases are a common group of neurological disorders characterized by progressive loss of neuronal structure and function leading to cognitive impairment. Recent studies have shown that neuronal pyroptosis mediated by the NLRP3 inflammasome plays a crucial role in the pathogenesis of neurodegenerative diseases. OBJECTIVE AND METHOD The NLRP3 inflammasome is a multiprotein complex that, when activated within cells, triggers an inflammatory response, ultimately leading to pyroptotic cell death of neurons. Pyroptosis is a typical pro-inflammatory programmed cell death process occurring downstream of NLRP3 inflammasome activation, characterized by the formation of pores on the cell membrane by the GSDMD protein, leading to cell lysis and the release of inflammatory factors. It has been found that NLRP3 inflammasome-mediated neuronal pyroptosis is closely associated with the development of various neurodegenerative diseases, such as Alzheimer's disease, traumatic brain injury, and Parkinson's disease. Therefore, inhibiting NLRP3 inflammasome activation and attenuating neuronal pyroptosis could potentially serve as novel strategies for the treatment of neurodegenerative diseases. RESULTS The aim of this review is to explore the role of NLRP3 activation-mediated neuronal pyroptosis and neuroinflammation in neurodegenerative diseases. Firstly, we extensively discuss the relationship between NLRP3 inflammasome-mediated neuronal pyroptosis and neuroinflammation in various neurodegenerative diseases. Subsequently, we further explore the mechanisms driving NLRP3 activation and assembly, as well as the post-translational modifications regulating NLRP3 inflammasome activation. CONCLUSION Understanding these mechanisms will contribute to a deeper understanding of the link between neuronal pyroptosis and neurodegenerative diseases, and hold significant implications for the treatment and prevention of neurodegenerative diseases.
Collapse
Affiliation(s)
- Ying-Hao Han
- College of Life Science & Biotechnology, Heilongjiang Bayi Agricultural University, Daqing, 163319, China.
| | - Xiao-Dong Liu
- College of Life Science & Biotechnology, Heilongjiang Bayi Agricultural University, Daqing, 163319, China
| | - Mei-Hua Jin
- College of Life Science & Biotechnology, Heilongjiang Bayi Agricultural University, Daqing, 163319, China
| | - Hu-Nan Sun
- College of Life Science & Biotechnology, Heilongjiang Bayi Agricultural University, Daqing, 163319, China.
| | - Taeho Kwon
- Primate Resources Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Jeongeup-si, Jeonbuk, 56216, Republic of Korea.
- Department of Functional Genomics, KRIBB School of Bioscience, University of Science and Technology, Daejeon, 34113, Republic of Korea.
| |
Collapse
|
30
|
Shao S, Zhou D, Feng J, Liu Y, Baturuhu, Yin H, Zhan D. Regulation of inflammation and immunity in sepsis by E3 ligases. Front Endocrinol (Lausanne) 2023; 14:1124334. [PMID: 37465127 PMCID: PMC10351979 DOI: 10.3389/fendo.2023.1124334] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Accepted: 06/16/2023] [Indexed: 07/20/2023] Open
Abstract
Sepsis is a life-threatening organ dysfunction caused by an abnormal infection-induced immune response. Despite significant advances in supportive care, sepsis remains a considerable therapeutic challenge and is the leading cause of death in the intensive care unit (ICU). Sepsis is characterized by initial hyper-inflammation and late immunosuppression. Therefore, immune-modulatory therapies have great potential for novel sepsis therapies. Ubiquitination is an essential post-translational protein modification, which has been known to be intimately involved in innate and adaptive immune responses. Several E3 ubiquitin ligases have been implicated in innate immune signaling and T-cell activation and differentiation. In this article, we review the current literature and discuss the role of E3 ligases in the regulation of immune response and their effects on the course of sepsis to provide insights into the prevention and therapy for sepsis.
Collapse
Affiliation(s)
- Shasha Shao
- Department of Emergency Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Department of Critical Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Daixing Zhou
- Department of Emergency Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Department of Critical Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jun Feng
- Department of Emergency Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Department of Critical Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yanyan Liu
- Obstetrics and Gynecology Department, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Baturuhu
- Department of Neurosurgery Intensive Care Unit (ICU), People’s Hospital of Bortala Mongol Autonomous Prefecture, Bole, China
| | - Huimei Yin
- Department of Emergency Medicine, People’s Hospital of Bortala Mongol Autonomous Prefecture, Bole, China
| | - Daqian Zhan
- Department of Emergency Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Department of Critical Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
31
|
Pan J, Zhan C, Yuan T, Gu W, Wang W, Sun Y, Chen L. Long noncoding RNA signatures in intrauterine infection/inflammation-induced lung injury: an integrative bioinformatics study. BMC Pulm Med 2023; 23:194. [PMID: 37280583 DOI: 10.1186/s12890-023-02505-5] [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: 08/08/2022] [Accepted: 05/31/2023] [Indexed: 06/08/2023] Open
Abstract
BACKGROUND Intrauterine infection/inflammation can result in fetal and neonatal lung injury. However, the biological mechanisms of intrauterine infection/inflammation on fetal and neonatal lung injury and development are poorly known. To date, there are no reliable biomarkers for improving intrauterine infection/inflammation-induced lung injury. METHODS An animal model of intrauterine infection/inflammation-induced lung injury was established with pregnant Sprague-Dawley rats inoculated with Escherichia coli suspension. The intrauterine inflammatory status was assessed through the histological examination of the placenta and uterus. A serial of histological examinations of the fetal and neonatal rats lung tissues were performed. The fetal and neonatal rat lung tissues were harvested for next generation sequencing at embryonic day 17 and postnatal day 3, respectively. Differentially expressed mRNAs and lncRNAs were identified by conducting high-throughput sequencing technique. The target genes of identified differentially expressed lncRNAs were analyzed. Homology analyses for important differentially expressed lncRNAs were performed. RESULTS The histopathological results showed inflammatory infiltration, impaired alveolar vesicular structure, less alveolar numbers, and thickened alveolar septa in fetal and neonatal rat lung tissues. Transmission electron micrographs revealed inflammatory cellular swelling associated with diffuse alveolar damage and less surfactant-storing lamellar bodies in alveolar epithelial type II cells. As compared with the control group, there were 432 differentially expressed lncRNAs at embryonic day 17 and 125 differentially expressed lncRNAs at postnatal day 3 in the intrauterine infection group. The distribution, expression level, and function of these lncRNAs were shown in the rat genome. LncRNA TCONS_00009865, lncRNA TCONS_00030049, lncRNA TCONS_00081686, lncRNA TCONS_00091647, lncRNA TCONS_00175309, lncRNA TCONS_00255085, lncRNA TCONS_00277162, and lncRNA TCONS_00157962 may play an important role in intrauterine infection/inflammation-induced lung injury. Fifty homologous sequences in Homo sapiens were also identified. CONCLUSIONS This study provides genome-wide identification of novel lncRNAs which may serve as potential diagnostic biomarkers and therapeutic targets for intrauterine infection/inflammation-induced lung injury.
Collapse
Affiliation(s)
- Jiarong Pan
- Department of Neonatology, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, 310052, Zhejiang, China
| | - Canyang Zhan
- Department of Neonatology, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, 310052, Zhejiang, China
| | - Tianming Yuan
- Department of Neonatology, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, 310052, Zhejiang, China.
| | - Weizhong Gu
- Zhejiang Key Laboratory for Diagnosis and Therapy of Neonatal Diseases, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, 310052, Zhejiang, China
| | - Weiyan Wang
- Department of Neonatology, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, 310052, Zhejiang, China
| | - Yi Sun
- Department of Neonatology, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, 310052, Zhejiang, China
| | - Lihua Chen
- Department of Neonatology, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, 310052, Zhejiang, China
| |
Collapse
|
32
|
Jeon S, Kang J, Lee SB. BC-1215 inhibits ATP-induced IL-1β secretion via the FBXL2-mediated ubiquitination and degradation of not only NLRP3, but also pro-IL-1β in LPS-primed THP-1 cells. Biochem Biophys Res Commun 2023; 657:128-135. [PMID: 37004285 DOI: 10.1016/j.bbrc.2023.03.055] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 03/15/2023] [Accepted: 03/21/2023] [Indexed: 04/03/2023]
Abstract
BC-1215, bis-pyridinyl benzyl ethanediamine, is an inhibitor of F-box only protein 3 (FBXO3) and exerts anti-inflammatory effects. BC-1215 inhibits interactions between FBXO3-F-box and the leucine rich repeat protein 2 (FBXL2), leading to the upregulation of FBXL2 expression, FBXL2-mediated ubiquitination and the degradation of tumor necrosis factor receptor (TNFR)-associated factor 6 (TRAF6) or NOD-, LRR- and the pyrin domain-containing protein 3 (NLRP3), which subsequently results in the down-regulation of inflammatory cytokine production. In the current study, we investigated the issue of whether or how BC-1215 suppresses the ATP-induced secretion of IL-1β in LPS-primed human macrophage-like cells, THP-1 cells. Our result show that pre-treatment with BC-1215 attenuated the ATP-induced secretion of IL-1β in LPS-primed THP-1 cells. Treatment of the LPS-primed THP-1 cells with BC-1215 resulted in a decrease in the level of NLRP3 and pro-IL-1β at the protein level, but not at the mRNA level. In addition, treatment with MG-132, but not leupeptin, inhibited the BC-1215-induced degradation of NLRP3 and pro-IL-1β proteins, and restored their levels, suggesting that BC-1215 decreases the stability of NLRP3 and pro-IL-1β at the protein level via proteasome-dependent degradation. Our results also show that FBXL2, which is increased by BC-1215, bound to and ubiquitinated NLRP3 and pro-IL-1β, but not pro-caspase-1. These collective results indicate that treatment with BC-1215, an inhibitor of FBXO3, inhibits ATP-induced IL-1β secretion via the FBXL2-mediated ubiquitination and degradation of pro-IL-1β as well as NLRP3 in LPS-primed THP-1 cells, suggesting that FBXO3 is a potential therapeutic target for developing agents against inflammatory diseases.
Collapse
|
33
|
Bednash JS, Johns F, Farkas D, Elhance A, Adair J, Cress K, Yount JS, Kenney AD, Londino JD, Mallampalli RK. Inhibiting the Deubiquitinase UCHL1 Reduces SARS-CoV-2 Viral Uptake by ACE2. Am J Respir Cell Mol Biol 2023; 68:566-576. [PMID: 36730646 PMCID: PMC10174169 DOI: 10.1165/rcmb.2022-0331oc] [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: 08/24/2022] [Accepted: 02/02/2023] [Indexed: 02/04/2023] Open
Abstract
Coronavirus disease (COVID-19), caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), remains a significant public health burden with limited treatment options. Many β-coronaviruses, including SARS-CoV-2, gain entry to host cells through the interaction of SARS-CoV-2 spike protein with membrane-bound ACE2 (angiotensin-converting enzyme 2). Given its necessity for SARS-CoV-2 infection, ACE2 represents a potential therapeutic target in COVID-19. However, early attempts focusing on ACE2 in COVID-19 have not validated it as a druggable target nor identified other ACE2-related novel proteins for therapeutic intervention. Here, we identify a mechanism for ACE2 protein modulation by the deubiquitinase (DUB) enzyme UCHL1 (ubiquitin carboxyl-terminal hydrolase isozyme L1). ACE2 is constitutively ubiquitinated and degraded by the proteasome in lung epithelia. SARS-CoV-2 spike protein cellular internalization increased ACE2 protein abundance by decreasing its degradation. Using an siRNA library targeting 96 human DUBs, we identified UCHL1 as a putative regulator of ACE2 function as a viral receptor. Overexpressed UCHL1 preserved ACE2 protein abundance, whereas silencing of the DUB in cells destabilized ACE2 through increased polyubiquitination. A commercially available small molecule inhibitor of UCHL1 DUB activity decreased ACE2 protein concentrations coupled with inhibition of SARS-CoV-2 infection in epithelial cells. These findings describe a unique pathway of ACE2 regulation uncovering UCHL1 as a potential therapeutic target to modulate COVID-19 viral entry as a platform for future small molecule design and testing.
Collapse
Affiliation(s)
- Joseph S. Bednash
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Internal Medicine, and
| | - Finny Johns
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Internal Medicine, and
| | - Daniela Farkas
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Internal Medicine, and
| | - Ajit Elhance
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Internal Medicine, and
| | - Jessica Adair
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Internal Medicine, and
| | - Kirstin Cress
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Internal Medicine, and
| | - Jacob S. Yount
- Department of Microbial Infection and Immunity, The Ohio State University, Columbus, Ohio
| | - Adam D. Kenney
- Department of Microbial Infection and Immunity, The Ohio State University, Columbus, Ohio
| | - James D. Londino
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Internal Medicine, and
| | - Rama K. Mallampalli
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Internal Medicine, and
| |
Collapse
|
34
|
Ma Q. Pharmacological Inhibition of the NLRP3 Inflammasome: Structure, Molecular Activation, and Inhibitor-NLRP3 Interaction. Pharmacol Rev 2023; 75:487-520. [PMID: 36669831 PMCID: PMC10121800 DOI: 10.1124/pharmrev.122.000629] [Citation(s) in RCA: 70] [Impact Index Per Article: 35.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Revised: 12/20/2022] [Accepted: 12/21/2022] [Indexed: 01/22/2023] Open
Abstract
The nucleotide-binding, oligomerization domain-like receptor family pyrin domain containing 3 (NLRP3) inflammasome is a multiprotein complex that combines sensing, regulation, and effector functions to regulate inflammation in health and disease. NLRP3 is activated by a diverse range of inflammation-instigating signals known as pathogen associated molecular patterns and danger associated molecular patterns. Upon activation, NLRP3 oligomerizes and recruits partner proteins to form a supramolecular platform to process the maturation and release of interleukin (IL)-1β, IL-18, and gasdermin D, major mediators of inflammation and inflammatory cell death termed pyroptosis. The NLRP3 inflammasome has been implicated in the pathogenesis of a wide range of disease conditions, including chronic inflammatory disease that are associated with lifestyle and dietary changes, aging, and environmental exposures, and have become the leading cause of death worldwide. Pharmacological targeting of NLRP3 and signaling demonstrated promising efficacy in ameliorating a list of disease conditions in animal models. These findings underscore the potential and importance of NLRP3 as a druggable target for treating a range of diseases. In this review, recent progress in understanding the structure and mechanism of action of the NLRP3 inflammasome is discussed with focus on pharmacological inhibition of NLRP3 by small molecule inhibitors. New structural and mechanistic insights into NLRP3 activation and inhibitor-NLRP3 interactions would aid in the rational design and pharmacological evaluation of NLRP3 inhibitors for treatment of human disease. SIGNIFICANCE STATEMENT: The NLRP3 inflammasome plays central role in innate immune sensing and control of inflammation. Pharmacological inhibition of NLRP3 demonstrated promising efficacy in a range of diseases in animal models. Recent elucidation of the structure and inhibitor binding of NLRP3 generated new insights into its mode of action and inhibitor-NLRP3 interaction at molecular levels, providing new framework for developing small chemical inhibitors of NLRP3 with improved efficacy and specificity against chronic disease that has become major health concerns worldwide.
Collapse
Affiliation(s)
- Qiang Ma
- Receptor Biology Laboratory, Toxicology and Molecular Biology Branch, Health Effects Laboratory Division, National Institute for Occupational Safety and Health, Centers for Disease Control and Prevention, Morgantown, West Virginia
| |
Collapse
|
35
|
Xu J, Núñez G. The NLRP3 inflammasome: activation and regulation. Trends Biochem Sci 2023; 48:331-344. [PMID: 36336552 PMCID: PMC10023278 DOI: 10.1016/j.tibs.2022.10.002] [Citation(s) in RCA: 225] [Impact Index Per Article: 112.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Revised: 10/10/2022] [Accepted: 10/10/2022] [Indexed: 11/06/2022]
Abstract
The NOD-, LRR- and pyrin domain-containing protein 3 (NLRP3) inflammasome is a cytoplasmic supramolecular complex that is activated in response to cellular perturbations triggered by infection and sterile injury. Assembly of the NLRP3 inflammasome leads to activation of caspase-1, which induces the maturation and release of interleukin-1β (IL-1β) and IL-18, as well as cleavage of gasdermin D (GSDMD), which promotes a lytic form of cell death. Production of IL-1β via NLRP3 can contribute to the pathogenesis of inflammatory disease, whereas aberrant IL-1β secretion through inherited NLRP3 mutations causes autoinflammatory disorders. In this review, we discuss recent developments in the structure of the NLRP3 inflammasome, and the cellular processes and signaling events controlling its assembly and activation.
Collapse
Affiliation(s)
- Jie Xu
- Department of Pathology and Rogel Cancer Center, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Gabriel Núñez
- Department of Pathology and Rogel Cancer Center, University of Michigan Medical School, Ann Arbor, MI, USA.
| |
Collapse
|
36
|
Xia J, Jiang S, Dong S, Liao Y, Zhou Y. The Role of Post-Translational Modifications in Regulation of NLRP3 Inflammasome Activation. Int J Mol Sci 2023; 24:ijms24076126. [PMID: 37047097 PMCID: PMC10093848 DOI: 10.3390/ijms24076126] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Revised: 03/16/2023] [Accepted: 03/20/2023] [Indexed: 04/14/2023] Open
Abstract
Pathogen-associated molecular patterns (PAMPs) and danger-associated molecular patterns (DAMPs) induce NLRP3 inflammasome activation, and subsequent formation of active caspase-1 as well as the maturation of interleukin-1β (IL-1β) and gasdermin D (GSDMD), mediating the occurrence of pyroptosis and inflammation. Aberrant NLRP3 inflammasome activation causes a variety of diseases. Therefore, the NLRP3 inflammasome pathway is a target for prevention and treatment of relative diseases. Recent studies have suggested that NLRP3 inflammasome activity is closely associated with its post-translational modifications (PTMs). This review focuses on PTMs of the components of the NLRP3 inflammasome and the resultant effects on regulation of its activity to provide references for the exploration of the mechanisms by which the NLRP3 inflammasome is activated and controlled.
Collapse
Affiliation(s)
- Jing Xia
- College of Veterinary Medicine, Southwest University, Chongqing 402460, China
| | - Songhong Jiang
- College of Veterinary Medicine, Southwest University, Chongqing 402460, China
| | - Shiqi Dong
- College of Veterinary Medicine, Southwest University, Chongqing 402460, China
| | - Yonghong Liao
- College of Veterinary Medicine, Southwest University, Chongqing 402460, China
- National Center of Technology Innovation for Pigs, Chongqing 402460, China
| | - Yang Zhou
- College of Veterinary Medicine, Southwest University, Chongqing 402460, China
- National Center of Technology Innovation for Pigs, Chongqing 402460, China
| |
Collapse
|
37
|
Zhou Z, Shang L, Zhang Q, Hu X, Huang JF, Xiong K. DTX3L induced NLRP3 ubiquitination inhibit R28 cell pyroptosis in OGD/R injury. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2023; 1870:119433. [PMID: 36706922 DOI: 10.1016/j.bbamcr.2023.119433] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Revised: 01/10/2023] [Accepted: 01/16/2023] [Indexed: 01/26/2023]
Abstract
Ischemia/reperfusion (I/R) injury is one of the most common etiologies in many diseases. Retinal I/R leads to cytokine storm, resulting in tissue damage and cell death. Pyroptosis, a novel type of regulated cell death, occurs after cellular I/R injury. In this study, we established an oxygen glucose deprivation (OGD/R) cellular model (R28) to simulate retinal I/R injury. We conducted an LDH assay, and EthD-III and PI staining procedures to confirm pyroptosis. Mass spectrometry and bioinformatics analysis were used to identify the possible proteins interacting with NLRP3. Co-IP and various molecular biology techniques were used to investigate the possible modes regulating NLRP3 by DTX3L. EthD-III, PI staining and LDH assays demonstrated pyroptosis induced by OGD/R injury, mediated via NLRP3 pathway. Mass spectrometry and bioinformatics analysis screened out three candidate proteins interacting with NLRP3, and further Co-IP experiment indicated that DTX-3L may interact with NLRP3 to regulate its protein levels after injury. Co-IP experiments and various molecular biology methods demonstrated that DTX3L ubiquitinates NLRP3 resulting in pyroptosis after R28 OGD/R injury. Further, NLRP3 LRR and DTX3L RING domains interact with each other. Our study demonstrated that DTX3L may ubiquitinate NLRP3 to regulate OGD/R-induced pyroptosis globally in R28 cells.
Collapse
Affiliation(s)
- Ziyu Zhou
- Department of Anatomy and Neurobiology, School of Basic Medical Science, Central South University, Changsha 410008, China; The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou 310022, China
| | - Lei Shang
- Jiangxi Research Institute of Ophthalmology and Visual Sciences, Affiliated Eye Hospital of Nanchang University, Nanchang 330006, China.
| | - Qi Zhang
- Department of Anatomy and Neurobiology, School of Basic Medical Science, Central South University, Changsha 410008, China.
| | - Ximin Hu
- Department of Anatomy and Neurobiology, School of Basic Medical Science, Central South University, Changsha 410008, China
| | - Ju-Fang Huang
- Department of Anatomy and Neurobiology, School of Basic Medical Science, Central South University, Changsha 410008, China.
| | - Kun Xiong
- Department of Anatomy and Neurobiology, School of Basic Medical Science, Central South University, Changsha 410008, China; Hunan Key Laboratory of Ophthalmology, Changsha 410008, China; Key Laboratory of Emergency and Trauma, Ministry of Education, College of Emergency and Trauma, Hainan Medical University, Haikou 571199, China.
| |
Collapse
|
38
|
Liang D, Jiang L, Bhat SA, Missiroli S, Perrone M, Lauriola A, Adhikari R, Gudur A, Vasi Z, Ahearn I, Guardavaccaro D, Giorgi C, Philips M, Kuchay S. Palmitoylation and PDE6δ regulate membrane-compartment-specific substrate ubiquitylation and degradation. Cell Rep 2023; 42:111999. [PMID: 36662618 PMCID: PMC9988375 DOI: 10.1016/j.celrep.2023.111999] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Revised: 11/11/2022] [Accepted: 01/04/2023] [Indexed: 01/20/2023] Open
Abstract
Substrate degradation by the ubiquitin proteasome system (UPS) in specific membrane compartments remains elusive. Here, we show that the interplay of two lipid modifications and PDE6δ regulates compartmental substrate targeting via the SCFFBXL2. FBXL2 is palmitoylated in a prenylation-dependent manner on cysteines 417 and 419 juxtaposed to the CaaX motif. Palmitoylation/depalmitoylation regulates its subcellular trafficking for substrate engagement and degradation. To control its subcellular distribution, lipid-modified FBXL2 interacts with PDE6δ. Perturbing the equilibrium between FBXL2 and PDE6δ disrupts the delivery of FBXL2 to all membrane compartments, whereas depalmitoylated FBXL2 is enriched on the endoplasmic reticulum (ER). Depalmitoylated FBXL2(C417S/C419S) promotes the degradation of IP3R3 at the ER, inhibits IP3R3-dependent mitochondrial calcium overload, and counteracts calcium-dependent cell death upon oxidative stress. In contrast, disrupting the PDE6δ-FBXL2 equilibrium has the opposite effect. These findings describe a mechanism underlying spatially-restricted substrate degradation and suggest that inhibition of FBXL2 palmitoylation and/or binding to PDE6δ may offer therapeutic benefits.
Collapse
Affiliation(s)
- David Liang
- Department of Biochemistry and Molecular Genetics, University of Illinois at Chicago, MBRB #1157, Chicago, IL 60607, USA
| | - Liping Jiang
- Department of Biochemistry and Molecular Genetics, University of Illinois at Chicago, MBRB #1157, Chicago, IL 60607, USA
| | - Sameer Ahmed Bhat
- Department of Biochemistry and Molecular Genetics, University of Illinois at Chicago, MBRB #1157, Chicago, IL 60607, USA
| | - Sonia Missiroli
- Department of Medical Sciences, Section of Experimental Medicine, University of Ferrara, 44121 Ferrara, Italy
| | - Mariasole Perrone
- Department of Medical Sciences, Section of Experimental Medicine, University of Ferrara, 44121 Ferrara, Italy
| | - Angela Lauriola
- Department of Biotechnology, University of Verona, 37134 Verona, Italy
| | - Ritika Adhikari
- Department of Biochemistry and Molecular Genetics, University of Illinois at Chicago, MBRB #1157, Chicago, IL 60607, USA
| | - Anish Gudur
- Department of Biochemistry and Molecular Genetics, University of Illinois at Chicago, MBRB #1157, Chicago, IL 60607, USA
| | - Zahra Vasi
- Department of Biochemistry and Molecular Genetics, University of Illinois at Chicago, MBRB #1157, Chicago, IL 60607, USA
| | - Ian Ahearn
- Department of Dermatology and Perlmutter Cancer Center, New York University Grossman School of Medicine, New York, NY 10016, USA
| | | | - Carlotta Giorgi
- Department of Medical Sciences, Section of Experimental Medicine, University of Ferrara, 44121 Ferrara, Italy
| | - Mark Philips
- Department of Medicine and Perlmutter Cancer Center, New York University Grossman School of Medicine, New York, NY 10016, USA
| | - Shafi Kuchay
- Department of Biochemistry and Molecular Genetics, University of Illinois at Chicago, MBRB #1157, Chicago, IL 60607, USA.
| |
Collapse
|
39
|
Wang L, Cai J, Zhao X, Ma L, Zeng P, Zhou L, Liu Y, Yang S, Cai Z, Zhang S, Zhou L, Yang J, Liu T, Jin S, Cui J. Palmitoylation prevents sustained inflammation by limiting NLRP3 inflammasome activation through chaperone-mediated autophagy. Mol Cell 2023; 83:281-297.e10. [PMID: 36586411 DOI: 10.1016/j.molcel.2022.12.002] [Citation(s) in RCA: 107] [Impact Index Per Article: 53.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Revised: 10/27/2022] [Accepted: 11/30/2022] [Indexed: 12/31/2022]
Abstract
As a key component of the inflammasome, NLRP3 is a critical intracellular danger sensor emerging as an important clinical target in inflammatory diseases. However, little is known about the mechanisms that determine the kinetics of NLRP3 inflammasome stability and activity to ensure effective and controllable inflammatory responses. Here, we show that S-palmitoylation acts as a brake to turn NLRP3 inflammasome off. zDHHC12 is identified as the S-acyltransferase for NLRP3 palmitoylation, which promotes its degradation through the chaperone-mediated autophagy pathway. Zdhhc12 deficiency in mice enhances inflammatory symptoms and lethality following alum-induced peritonitis and LPS-induced endotoxic shock. Notably, several disease-associated mutations in NLRP3 are associated with defective palmitoylation, resulting in overt NLRP3 inflammasome activation. Thus, our findings identify zDHHC12 as a repressor of NLRP3 inflammasome activation and uncover a previously unknown regulatory mechanism by which the inflammasome pathway is tightly controlled by the dynamic palmitoylation of NLRP3.
Collapse
Affiliation(s)
- Liqiu Wang
- MOE Key Laboratory of Gene Function and Regulation, State Key Laboratory of Biocontrol, School of Life Sciences of Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Jing Cai
- MOE Key Laboratory of Gene Function and Regulation, State Key Laboratory of Biocontrol, School of Life Sciences of Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Xin Zhao
- MOE Key Laboratory of Gene Function and Regulation, State Key Laboratory of Biocontrol, School of Life Sciences of Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Ling Ma
- MOE Key Laboratory of Gene Function and Regulation, State Key Laboratory of Biocontrol, School of Life Sciences of Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Ping Zeng
- The Department of Rheumatology, Guangzhou Women and Children's Medical Centre, Guangzhou, Guangdong, China
| | - Lingli Zhou
- MOE Key Laboratory of Gene Function and Regulation, State Key Laboratory of Biocontrol, School of Life Sciences of Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Yukun Liu
- Key Laboratory of Stem Cells and Tissue Engineering, Zhongshan School of Medicine, Sun Yat-sen University, Ministry of Education, Guangzhou, Guangdong, China
| | - Shuai Yang
- MOE Key Laboratory of Gene Function and Regulation, State Key Laboratory of Biocontrol, School of Life Sciences of Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Zhe Cai
- The Department of Rheumatology, Guangzhou Women and Children's Medical Centre, Guangzhou, Guangdong, China
| | - Song Zhang
- The Department of Rheumatology, Guangzhou Women and Children's Medical Centre, Guangzhou, Guangdong, China
| | - Liang Zhou
- MOE Key Laboratory of Gene Function and Regulation, State Key Laboratory of Biocontrol, School of Life Sciences of Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Jiahui Yang
- Huizhou Municipal Central Hospital, Huizhou, Guangdong, China
| | - Tao Liu
- MOE Key Laboratory of Gene Function and Regulation, State Key Laboratory of Biocontrol, School of Life Sciences of Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Shouheng Jin
- MOE Key Laboratory of Gene Function and Regulation, State Key Laboratory of Biocontrol, School of Life Sciences of Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Jun Cui
- MOE Key Laboratory of Gene Function and Regulation, State Key Laboratory of Biocontrol, School of Life Sciences of Sun Yat-sen University, Guangzhou, Guangdong, China.
| |
Collapse
|
40
|
Zhan X, Li Q, Xu G, Xiao X, Bai Z. The mechanism of NLRP3 inflammasome activation and its pharmacological inhibitors. Front Immunol 2023; 13:1109938. [PMID: 36741414 PMCID: PMC9889537 DOI: 10.3389/fimmu.2022.1109938] [Citation(s) in RCA: 58] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Accepted: 12/29/2022] [Indexed: 01/20/2023] Open
Abstract
NLRP3 (NOD-, LRR-, and pyrin domain-containing protein 3) is a cytosolic pattern recognition receptor (PRR) that recognizes multiple pathogen-associated molecular patterns (PAMPs) and damage-associated molecular patterns (DAMPs). Once activated, NLRP3 initiates the inflammasome assembly together with the adaptor ASC and the effector caspase-1, leading to caspase-1 activation and subsequent cleavage of IL-1β and IL-18. Aberrant NLRP3 inflammasome activation is linked with the pathogenesis of multiple inflammatory diseases, such as cryopyrin-associated periodic syndromes, type 2 diabetes, non-alcoholic steatohepatitis, gout, and neurodegenerative diseases. Thus, NLRP3 is an important therapeutic target, and researchers are putting a lot of effort into developing its inhibitors. The review summarizes the latest advances in the mechanism of NLRP3 inflammasome activation and its pharmacological inhibitors.
Collapse
Affiliation(s)
- Xiaoyan Zhan
- Department of Hepatology, Fifth Medical Center of Chinese PLA General Hospital, Beijing, China,China Military Institute of Chinese Materia, Fifth Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Qiang Li
- Department of Hepatology, Fifth Medical Center of Chinese PLA General Hospital, Beijing, China,China Military Institute of Chinese Materia, Fifth Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Guang Xu
- Department of Hepatology, Fifth Medical Center of Chinese PLA General Hospital, Beijing, China,China Military Institute of Chinese Materia, Fifth Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Xiaohe Xiao
- Department of Hepatology, Fifth Medical Center of Chinese PLA General Hospital, Beijing, China,China Military Institute of Chinese Materia, Fifth Medical Center of Chinese PLA General Hospital, Beijing, China,*Correspondence: Xiaohe Xiao, ; Zhaofang Bai,
| | - Zhaofang Bai
- Department of Hepatology, Fifth Medical Center of Chinese PLA General Hospital, Beijing, China,China Military Institute of Chinese Materia, Fifth Medical Center of Chinese PLA General Hospital, Beijing, China,*Correspondence: Xiaohe Xiao, ; Zhaofang Bai,
| |
Collapse
|
41
|
Role of NLRP3 Inflammasome and Its Inhibitors as Emerging Therapeutic Drug Candidate for Alzheimer's Disease: a Review of Mechanism of Activation, Regulation, and Inhibition. Inflammation 2023; 46:56-87. [PMID: 36006570 PMCID: PMC9403980 DOI: 10.1007/s10753-022-01730-0] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Accepted: 05/26/2022] [Indexed: 02/06/2023]
Abstract
Alzheimer's disease (AD) is one of the most prevalent neurodegenerative disorders. The etiology and pathology of AD are complicated, variable, and yet to be completely discovered. However, the involvement of inflammasomes, particularly the NLRP3 inflammasome, has been emphasized recently. NLRP3 is a critical pattern recognition receptor involved in the expression of immune responses and has been found to play a significant role in the development of various immunological and neurological disorders such as multiple sclerosis, ulcerative colitis, gout, diabetes, and AD. It is a multimeric protein which releases various cytokines and causes caspase-1 activation through the process known as pyroptosis. Increased levels of cytokines (IL-1β and IL-18), caspase-1 activation, and neuropathogenic stimulus lead to the formation of proinflammatory microglial M1. Progressive researches have also shown that besides loss of neurons, the pathophysiology of AD primarily includes amyloid beta (Aβ) accumulation, generation of oxidative stress, and microglial damage leading to activation of NLRP3 inflammasome that eventually leads to neuroinflammation and dementia. It has been suggested in the literature that suppressing the activity of the NLRP3 inflammasome has substantial potential to prevent, manage, and treat Alzheimer's disease. The present review discusses the functional composition, various models, signaling molecules, pathways, and evidence of NLRP3 activation in AD. The manuscript also discusses the synthetic drugs, their clinical status, and projected natural products as a potential therapeutic approach to manage and treat NLRP3 mediated AD.
Collapse
|
42
|
Hartmann A, Vila-Verde C, Guimarães FS, Joca SR, Lisboa SF. The NLRP3 Inflammasome in Stress Response: Another Target for the Promiscuous Cannabidiol. Curr Neuropharmacol 2023; 21:284-308. [PMID: 35410608 PMCID: PMC10190150 DOI: 10.2174/1570159x20666220411101217] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Revised: 03/14/2022] [Accepted: 03/27/2022] [Indexed: 11/22/2022] Open
Abstract
Many psychiatric patients do not respond to conventional therapy. There is a vast effort to investigate possible mechanisms involved in treatment resistance, trying to provide better treatment options, and several data points toward a possible involvement of inflammatory mechanisms. Microglia, glial, and resident immune cells are involved in complex responses in the brain, orchestrating homeostatic functions, such as synaptic pruning and maintaining neuronal activity. In contrast, microglia play a major role in neuroinflammation, neurodegeneration, and cell death. Increasing evidence implicate microglia dysfunction in neuropsychiatric disorders. The mechanisms are still unclear, but one pathway in microglia has received increased attention in the last 8 years, i.e., the NLRP3 inflammasome pathway. Stress response and inflammation, including microglia activation, can be attenuated by Cannabidiol (CBD). CBD has antidepressant, anti-stress, antipsychotic, anti-inflammatory, and other properties. CBD effects are mediated by direct or indirect modulation of many receptors, enzymes, and other targets. This review will highlight some findings for neuroinflammation and microglia involvement in stress-related psychiatric disorders, particularly addressing the NLRP3 inflammasome pathway. Moreover, we will discuss evidence and mechanisms for CBD effects in psychiatric disorders and animal models and address its potential effects on stress response via neuroinflammation and NLRP3 inflammasome modulation.
Collapse
Affiliation(s)
- Alice Hartmann
- Department of Pharmacology, School of Medicine of Ribeirão Preto (FMRP), University of São Paulo (USP), Ribeirão Preto, Brazil
| | - Carla Vila-Verde
- Department of Pharmacology, School of Medicine of Ribeirão Preto (FMRP), University of São Paulo (USP), Ribeirão Preto, Brazil
| | - Francisco S. Guimarães
- Department of Pharmacology, School of Medicine of Ribeirão Preto (FMRP), University of São Paulo (USP), Ribeirão Preto, Brazil
- Center for Interdisciplinary Research on Applied Neurosciences (NAPNA), University of São Paulo, São Paulo, Brazil
| | - Sâmia R. Joca
- Center for Interdisciplinary Research on Applied Neurosciences (NAPNA), University of São Paulo, São Paulo, Brazil
- BioMolecular Sciences Department, School of Pharmaceutical Sciences of Ribeirão Preto (FCFRP), University of São Paulo (USP);
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | - Sabrina F. Lisboa
- Center for Interdisciplinary Research on Applied Neurosciences (NAPNA), University of São Paulo, São Paulo, Brazil
- BioMolecular Sciences Department, School of Pharmaceutical Sciences of Ribeirão Preto (FCFRP), University of São Paulo (USP);
| |
Collapse
|
43
|
Gao Y, Xiao X, Luo J, Wang J, Peng Q, Zhao J, Jiang N, Zhao Y. E3 Ubiquitin Ligase FBXO3 Drives Neuroinflammation to Aggravate Cerebral Ischemia/Reperfusion Injury. Int J Mol Sci 2022; 23:13648. [PMID: 36362432 PMCID: PMC9658360 DOI: 10.3390/ijms232113648] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2022] [Revised: 10/27/2022] [Accepted: 11/02/2022] [Indexed: 08/04/2023] Open
Abstract
Ischemic stroke, one of the most universal causes of human mortality and morbidity, is pathologically characterized by inflammatory cascade, especially during the progression of ischemia/reperfusion (I/R) injury. F-Box Protein 3 (FBXO3), a substrate-recognition subunit of SKP1-cullin 1-F-box protein (SCF) E3 ligase complexes, has recently been proven to be severed as an underlying pro-inflammatory factor in pathological processes of diverse diseases. Given these considerations, the current study aims at investigating whether FBXO3 exerts impacts on inflammation in cerebral I/R injury. In this study, first, it was verified that FBXO3 protein expression increased after a middle cerebral artery occlusion/reperfusion (MCAO/R) model in Sprague-Dawley (SD) rats and was specifically expressed in neurons other than microglia or astrocytes. Meanwhile, in mouse hippocampal neuronal cell line HT22 cells, the elevation of FBXO3 protein was observed after oxygen and glucose deprivation/reoxygenation (OGD/R) treatment. It was also found that interference of FBXO3 with siRNA significantly alleviated neuronal damage via inhibiting the inflammatory response in I/R injury both in vivo and in vitro. The FBXO3 inhibitor BC-1215 was used to confirm the pro-inflammatory effect of FBXO3 in the OGD/R model as well. Furthermore, by administration of FBXO3 siRNA and BC-1215, FBXO3 was verified to reduce the protein level of Homeodomain-Interacting Protein Kinase 2 (HIPK2), likely through the ubiquitin-proteasome system (UPS), to aggravate cerebral I/R injury. Collectively, our results underline the detrimental effect FBXO3 has on cerebral I/R injury by accelerating inflammatory response, possibly through ubiquitylating and degrading HIPK2. Despite the specific interaction between FBXO3 and HIPK2 requiring further study, we believe that our data suggest the therapeutic relevance of FBXO3 to ischemic stroke and provide a new perspective on the mechanism of I/R injury.
Collapse
Affiliation(s)
- Yu Gao
- Department of Pathology, Chongqing Medical University, Chongqing 400016, China
- Molecular Medicine Diagnostic and Testing Center, Chongqing Medical University, Chongqing 400016, China
| | - Xinyu Xiao
- School of Basic Medical Science, Chongqing Medical University, Chongqing 400016, China
| | - Jing Luo
- Department of Pathology, Chongqing Medical University, Chongqing 400016, China
- Molecular Medicine Diagnostic and Testing Center, Chongqing Medical University, Chongqing 400016, China
| | - Jianwei Wang
- School of Basic Medical Science, Chongqing Medical University, Chongqing 400016, China
| | - Qiling Peng
- School of Basic Medical Science, Chongqing Medical University, Chongqing 400016, China
| | - Jing Zhao
- Institute of Neuroscience, Chongqing Medical University, Chongqing 400016, China
- Department of Pathophysiology, Chongqing Medical University, Chongqing 400016, China
| | - Ning Jiang
- Department of Pathology, Chongqing Medical University, Chongqing 400016, China
- Molecular Medicine Diagnostic and Testing Center, Chongqing Medical University, Chongqing 400016, China
| | - Yong Zhao
- Department of Pathology, Chongqing Medical University, Chongqing 400016, China
- Molecular Medicine Diagnostic and Testing Center, Chongqing Medical University, Chongqing 400016, China
| |
Collapse
|
44
|
Wang Q, Huang PY, Wu JG, Zhang TQ, Li LF, Huang LD, Yu YM, Wang MH, He J. miR-219a-5p inhibits the pyroptosis in knee osteoarthritis by inactivating the NLRP3 signaling via targeting FBXO3. ENVIRONMENTAL TOXICOLOGY 2022; 37:2673-2682. [PMID: 35962723 DOI: 10.1002/tox.23627] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Revised: 06/26/2022] [Accepted: 07/13/2022] [Indexed: 06/15/2023]
Abstract
PURPOSE This work was to identify the function and mechanism of miR-219a-5p in regulating knee osteoarthritis (KOA). METHODS Rat fibroblast-like synoviocytes (FLSs) were isolated to construct KOA cell model by lipopolysaccharide and adenosine triphosphate treatment. miR-219a-5p and FBXO3 expression in FLSs was modulated by transfection. Flow cytometry was executed to research FLSs apoptosis. Caspase-1 and IL-1β expression in FLSs was researched by immunofluorescence. The binding between miR-219a-5p and FBXO3 was identified by dual luciferase reporter gene assay. KOA rat model and miR-219a-5p up-modulation KOA rat model were constructed. Step size of rats was analyzed. Knee joints of rats were experienced Safranin O-fast green staining to evaluate the knee joint injury. FBXO3, pyroptosis-associated proteins, and IL-1β and IL-18 expression in FLSs and articular cartilage tissues of rats were assessed by Western blot, qRT-PCR and Enzyme-linked immunosorbent assay. RESULTS KOA cell model had higher apoptosis percentage, expression of pyroptosis-associated proteins, and IL-1β and IL-18 level. miR-219a-5p up-modulation decreased the above indicators, whereas miR-219a-5p down-modulation increased the above indicators. FBXO3 expression was directly repressed by miR-219a-5p. Loss of FBXO3 suppressed the above indicators. FBXO3 counteracted the suppression of miR-219a-5p on the above indicators. miR-219a-5p agomir attenuated knee joint injury, increased step size of KOA rats, and reduced FBXO3, pyroptosis-associated proteins and level of IL-1β and IL-18 in the articular cartilage tissues of KOA rats. CONCLUSION miR-219a-5p suppressed the pyroptosis in KOA by inactivating the NLRP3 signaling via targeting FBXO3, which might be a promising target for ameliorating KOA in the clinic.
Collapse
Affiliation(s)
- Qiang Wang
- Department of Orthopaedic Surgery, Shanghai Fifth People's Hospital Affiliated to Fudan University, Shanghai, China
| | - Pei-Yan Huang
- Department of Orthopaedic Surgery, Shanghai Fifth People's Hospital Affiliated to Fudan University, Shanghai, China
| | - Jun-Guo Wu
- Department of Orthopaedic Surgery, Shanghai Fifth People's Hospital Affiliated to Fudan University, Shanghai, China
| | - Tie-Qi Zhang
- Department of Orthopaedic Surgery, Shanghai Fifth People's Hospital Affiliated to Fudan University, Shanghai, China
| | - Ling-Feng Li
- Department of Orthopaedic Surgery, Shanghai Fifth People's Hospital Affiliated to Fudan University, Shanghai, China
| | - Liang-Da Huang
- Department of Orthopaedic Surgery, Shanghai Fifth People's Hospital Affiliated to Fudan University, Shanghai, China
| | - Yue-Ming Yu
- Department of Orthopaedic Surgery, Shanghai Fifth People's Hospital Affiliated to Fudan University, Shanghai, China
| | - Ming-Hai Wang
- Department of Orthopaedic Surgery, Shanghai Fifth People's Hospital Affiliated to Fudan University, Shanghai, China
| | - Jun He
- Department of Orthopaedic Surgery, Shanghai Fifth People's Hospital Affiliated to Fudan University, Shanghai, China
| |
Collapse
|
45
|
Akbal A, Dernst A, Lovotti M, Mangan MSJ, McManus RM, Latz E. How location and cellular signaling combine to activate the NLRP3 inflammasome. Cell Mol Immunol 2022; 19:1201-1214. [PMID: 36127465 PMCID: PMC9622870 DOI: 10.1038/s41423-022-00922-w] [Citation(s) in RCA: 100] [Impact Index Per Article: 33.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2022] [Accepted: 08/15/2022] [Indexed: 01/27/2023] Open
Abstract
NOD-, LRR-, and pyrin domain-containing 3 (NLRP3) is a cytosolic innate immune sensor of cellular stress signals, triggered by infection and sterile inflammation. Upon detection of an activating stimulus, NLRP3 transitions from an inactive homo-oligomeric multimer into an active multimeric inflammasome, which promotes the helical oligomeric assembly of the adaptor molecule ASC. ASC oligomers provide a platform for caspase-1 activation, leading to the proteolytic cleavage and activation of proinflammatory cytokines in the IL-1 family and gasdermin D, which can induce a lytic form of cell death. Recent studies investigating both the cellular requirement for NLRP3 activation and the structure of NLRP3 have revealed the complex regulation of NLRP3 and the multiple steps involved in its activation. This review presents a perspective on the biochemical and cellular processes controlling the assembly of the NLRP3 inflammasome with particular emphasis on structural regulation and the role of organelles. We also highlight the latest research on metabolic control of this inflammatory pathway and discuss promising clinical targets for intervention.
Collapse
Affiliation(s)
- Anil Akbal
- Institute of Innate Immunity, University Hospital Bonn, University of Bonn, 53127, Bonn, Germany
| | - Alesja Dernst
- Institute of Innate Immunity, University Hospital Bonn, University of Bonn, 53127, Bonn, Germany
| | - Marta Lovotti
- Institute of Innate Immunity, University Hospital Bonn, University of Bonn, 53127, Bonn, Germany
| | - Matthew S J Mangan
- Institute of Innate Immunity, University Hospital Bonn, University of Bonn, 53127, Bonn, Germany
| | - Róisín M McManus
- Institute of Innate Immunity, University Hospital Bonn, University of Bonn, 53127, Bonn, Germany
- German Center for Neurodegenerative Diseases, 53127, Bonn, Germany
| | - Eicke Latz
- Institute of Innate Immunity, University Hospital Bonn, University of Bonn, 53127, Bonn, Germany.
- German Center for Neurodegenerative Diseases, 53127, Bonn, Germany.
- Department of Infectious Diseases & Immunology, UMass Medical School, Worcester, MA, 01605, USA.
- Centre of Molecular Inflammation Research, Norwegian University of Science and Technology, 7491, Trondheim, Norway.
| |
Collapse
|
46
|
Guan Y, Gu Y, Li H, Liang B, Han C, Zhang Y, Liu Q, Wei W, Ma Y. NLRP3 inflammasome activation mechanism and its role in autoimmune liver disease. Acta Biochim Biophys Sin (Shanghai) 2022; 54:1577-1586. [PMID: 36148948 PMCID: PMC9828325 DOI: 10.3724/abbs.2022137] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
The NLRP3 inflammasome is a multiprotein binding compound comprising NLRP3, connector protein ASC, and effector protein pro-caspase-1. When the NLRP3 inflammasome senses a danger signal from the host or pathogen, activated caspase-1 cleaves the precursors of interleukin (IL)-1β and IL-18 into mature proinflammatory cytokines, simultaneously causing lysis via the pore-forming protein gasdermin D. This induction of cell inflammatory pyroptosis suggests that it is a key process in the innate immune response to pathogens or cellular stress. Recent studies have shown that NLRP3 inflammasome also plays an important role in regulating autoimmune liver diseases, including autoimmune hepatitis, primary biliary cholangitis, and primary sclerosclerotic cholangitis. In this review, we summarize the structure, activation and modulation of the NLRP3 inflammasome, highlight the progress in research on the role of NLRP3 inflammasome in the occurrence and development of autoimmune liver diseases, and discuss potential strategies for targeting the NLRP3 inflammasome in the treatment of autoimmune liver diseases.
Collapse
Affiliation(s)
- Yanling Guan
- Institute of Clinical PharmacologyAnhui Medical UniversityKey Laboratory of Anti-inflammatory and Immune MedicineMinistry of EducationAnhui Collaborative Innovation Center of Anti-inflammatory and Immune MedicineCenter of Rheumatoid Arthritis of Anhui Medical UniversityHefei230032China
| | - Yiyue Gu
- Department of Cardiologythe First People’s Hospital of XuzhouXuzhou221000China
| | - Hao Li
- Institute of Clinical PharmacologyAnhui Medical UniversityKey Laboratory of Anti-inflammatory and Immune MedicineMinistry of EducationAnhui Collaborative Innovation Center of Anti-inflammatory and Immune MedicineCenter of Rheumatoid Arthritis of Anhui Medical UniversityHefei230032China
| | - Bo Liang
- Institute of Dermatology and Department of Dermatologythe First Affiliated HospitalAnhui Medical UniversityHefei230032China
| | - Chenchen Han
- Institute of Clinical PharmacologyAnhui Medical UniversityKey Laboratory of Anti-inflammatory and Immune MedicineMinistry of EducationAnhui Collaborative Innovation Center of Anti-inflammatory and Immune MedicineCenter of Rheumatoid Arthritis of Anhui Medical UniversityHefei230032China
| | - Yu Zhang
- Institute of Clinical PharmacologyAnhui Medical UniversityKey Laboratory of Anti-inflammatory and Immune MedicineMinistry of EducationAnhui Collaborative Innovation Center of Anti-inflammatory and Immune MedicineCenter of Rheumatoid Arthritis of Anhui Medical UniversityHefei230032China
| | - Qian Liu
- Institute of Clinical PharmacologyAnhui Medical UniversityKey Laboratory of Anti-inflammatory and Immune MedicineMinistry of EducationAnhui Collaborative Innovation Center of Anti-inflammatory and Immune MedicineCenter of Rheumatoid Arthritis of Anhui Medical UniversityHefei230032China
| | - Wei Wei
- Institute of Clinical PharmacologyAnhui Medical UniversityKey Laboratory of Anti-inflammatory and Immune MedicineMinistry of EducationAnhui Collaborative Innovation Center of Anti-inflammatory and Immune MedicineCenter of Rheumatoid Arthritis of Anhui Medical UniversityHefei230032China,Correspondence address. Tel: +86-551-65161209; E-mail: (Y.M.) / E-mail: (W.W.) @ahmu.edu.cn
| | - Yang Ma
- Institute of Clinical PharmacologyAnhui Medical UniversityKey Laboratory of Anti-inflammatory and Immune MedicineMinistry of EducationAnhui Collaborative Innovation Center of Anti-inflammatory and Immune MedicineCenter of Rheumatoid Arthritis of Anhui Medical UniversityHefei230032China,Correspondence address. Tel: +86-551-65161209; E-mail: (Y.M.) / E-mail: (W.W.) @ahmu.edu.cn
| |
Collapse
|
47
|
Masuda T, Haji S, Nakashima Y, Tsuda M, Kimura D, Takamatsu A, Iwahashi N, Umakoshi H, Shiratsuchi M, Kikutake C, Suyama M, Ohkawa Y, Ogawa Y. Identification of a drug-response gene in multiple myeloma through longitudinal single-cell transcriptome sequencing. iScience 2022; 25:104781. [PMID: 35992084 PMCID: PMC9386061 DOI: 10.1016/j.isci.2022.104781] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2022] [Revised: 06/10/2022] [Accepted: 06/13/2022] [Indexed: 11/06/2022] Open
Abstract
Despite recent therapeutic advances for multiple myeloma (MM), relapse is very common. Here, we conducted longitudinal single-cell transcriptome sequencing (scRNA-seq) of MM cells from a patient with relapsed MM, treated with multiple anti-myeloma drugs. We observed five subclusters of MM cells, which appeared and/or disappeared in response to the therapeutic pressure, and identified cluster 3 which emerged during lenalidomide treatment and disappeared after proteasome inhibitor (PI) treatment. Among the differentially expressed genes in cluster 3, we found a candidate drug-response gene; pellino E3 ubiquitin-protein ligase family member 2 (PELI2), which is responsible for PI-induced cell death in in vitro assay. Kaplan-Meier survival analysis of database revealed that higher expression of PELI2 is associated with a better prognosis. Our integrated strategy combining longitudinal scRNA-seq analysis, in vitro functional assay, and database analysis would facilitate the understanding of clonal dynamics of MM in response to anti-myeloma drugs and identification of drug-response genes.
Collapse
Affiliation(s)
- Toru Masuda
- Department of Medicine and Bioregulatory Science, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan
| | - Shojiro Haji
- Department of Medicine and Bioregulatory Science, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan
| | - Yasuhiro Nakashima
- Department of Medicine and Bioregulatory Science, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan
| | - Mariko Tsuda
- Department of Medicine and Bioregulatory Science, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan
| | - Daisaku Kimura
- Department of Medicine and Bioregulatory Science, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan
| | - Akiko Takamatsu
- Department of Medicine and Bioregulatory Science, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan
| | - Norifusa Iwahashi
- Department of Medicine and Bioregulatory Science, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan
| | - Hironobu Umakoshi
- Department of Medicine and Bioregulatory Science, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan
| | - Motoaki Shiratsuchi
- Department of Medicine and Bioregulatory Science, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan
- Department of Hematology, Iizuka Hospital, Iizuka 820-8505, Japan
| | - Chie Kikutake
- Division of Bioinformatics, Medical Institute of Bioregulation, Kyushu University, Fukuoka 812-8582, Japan
| | - Mikita Suyama
- Division of Bioinformatics, Medical Institute of Bioregulation, Kyushu University, Fukuoka 812-8582, Japan
| | - Yasuyuki Ohkawa
- Division of Transcriptomics, Medical Institute of Bioregulation, Kyushu University, Fukuoka 812-8582, Japan
| | - Yoshihiro Ogawa
- Department of Medicine and Bioregulatory Science, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan
| |
Collapse
|
48
|
Activation and Pharmacological Regulation of Inflammasomes. Biomolecules 2022; 12:biom12071005. [PMID: 35883561 PMCID: PMC9313256 DOI: 10.3390/biom12071005] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Revised: 07/16/2022] [Accepted: 07/18/2022] [Indexed: 01/27/2023] Open
Abstract
Inflammasomes are intracellular signaling complexes of the innate immune system, which is part of the response to exogenous pathogens or physiological aberration. The multiprotein complexes mainly consist of sensor proteins, adaptors, and pro-caspase-1. The assembly of the inflammasome upon extracellular and intracellular cues drives the activation of caspase-1, which processes pro-inflammatory cytokines IL-1β and IL-18 to maturation and gasdermin-D for pore formation, leading to pyroptosis and cytokine release. Inflammasome signaling functions in numerous infectious or sterile inflammatory diseases, including inherited autoinflammatory diseases, metabolic disorders, cardiovascular diseases, cancers, neurodegenerative disorders, and COVID-19. In this review, we summarized current ideas on the organization and activation of inflammasomes, with details on the molecular mechanisms, regulations, and interventions. The recent developments of pharmacological strategies targeting inflammasomes as disease therapeutics were also covered.
Collapse
|
49
|
Ma X, Di Q, Li X, Zhao X, Zhang R, Xiao Y, Li X, Wu H, Tang H, Quan J, Wu Z, Xiao W, Chen W. Munronoid I Ameliorates DSS-Induced Mouse Colitis by Inhibiting NLRP3 Inflammasome Activation and Pyroptosis Via Modulation of NLRP3. Front Immunol 2022; 13:853194. [PMID: 35865528 PMCID: PMC9296101 DOI: 10.3389/fimmu.2022.853194] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Accepted: 06/07/2022] [Indexed: 12/31/2022] Open
Abstract
Inflammatory bowel diseases (IBDs) are increasingly common diseases characterized by chronic and relapsing inflammation of the gastrointestinal tract. NLRP3 might be a crucial regulator of the homeostatic balance of the intestine, but its upregulation leads to pyroptosis. Munronoid I is extracted and purified from Munronia sinica, which has shown an anti-inflammatory effect, but the efficacy of Munronoid I in IBD remains unproven. In this study, we attempted to determine the effect of Munronoid I on NLRP3 to regulate the inflammasome activation and pyroptosis in IBD. Our data demonstrated that Munronoid I treatment attenuated DSS-induced body weight loss, pathological injury of the colon, the production of IL-1β and IL-18, and the expression of pyroptosis-associated proteins in colon tissue in mice. Moreover, Munronoid I inhibited LPS/ATP-induced pyroptosis in mouse peritoneal macrophages, MODE-K cells, and DSS-induced pyroptosis in mouse colonic epithelial cells, and decreased the release of inflammatory cytokines IL-1β and IL-18 in mouse peritoneal macrophages. Mechanically, Munronoid I could suppress the NLRP3 inflammasome activation and pyroptosis by promoting the K48-linked ubiquitination and NLRP3 degradation. It is suggested that Munronoid I might be a potential therapeutic candidate for IBD.
Collapse
Affiliation(s)
- Xingyu Ma
- Marshall Laboratory of Biomedical Engineering, Department of Immunology, Shenzhen University School of Medicine, Shenzhen, China
| | - Qianqian Di
- Marshall Laboratory of Biomedical Engineering, Department of Immunology, Shenzhen University School of Medicine, Shenzhen, China
| | - Xiaoli Li
- Key Laboratory of Medicinal Chemistry for Natural Resource, Ministry of Education, Yunnan Research & Development Center for Natural Products, School of Chemical Science and Technology, Yunnan University, Kunming, China
| | - Xibao Zhao
- Marshall Laboratory of Biomedical Engineering, Department of Immunology, Shenzhen University School of Medicine, Shenzhen, China
| | - Ruihan Zhang
- Key Laboratory of Medicinal Chemistry for Natural Resource, Ministry of Education, Yunnan Research & Development Center for Natural Products, School of Chemical Science and Technology, Yunnan University, Kunming, China
| | - Yue Xiao
- Marshall Laboratory of Biomedical Engineering, Department of Immunology, Shenzhen University School of Medicine, Shenzhen, China
| | - Xunwei Li
- Marshall Laboratory of Biomedical Engineering, Department of Immunology, Shenzhen University School of Medicine, Shenzhen, China
| | - Han Wu
- Marshall Laboratory of Biomedical Engineering, Department of Immunology, Shenzhen University School of Medicine, Shenzhen, China
| | - Haimei Tang
- Marshall Laboratory of Biomedical Engineering, Department of Immunology, Shenzhen University School of Medicine, Shenzhen, China
| | - Jiazheng Quan
- Marshall Laboratory of Biomedical Engineering, Department of Immunology, Shenzhen University School of Medicine, Shenzhen, China
| | - Zherui Wu
- Marshall Laboratory of Biomedical Engineering, Department of Immunology, Shenzhen University School of Medicine, Shenzhen, China
| | - Weilie Xiao
- Key Laboratory of Medicinal Chemistry for Natural Resource, Ministry of Education, Yunnan Research & Development Center for Natural Products, School of Chemical Science and Technology, Yunnan University, Kunming, China
- *Correspondence: Weilie Xiao, ; Weilin Chen,
| | - Weilin Chen
- Marshall Laboratory of Biomedical Engineering, Department of Immunology, Shenzhen University School of Medicine, Shenzhen, China
- *Correspondence: Weilie Xiao, ; Weilin Chen,
| |
Collapse
|
50
|
Shi X, Li T, Liu Y, Yin L, Xiao L, Fu L, Zhu Y, Chen H, Wang K, Xiao X, Zhang H, Tan S, Tan S. HSF1 Protects Sepsis-Induced Acute Lung Injury by Inhibiting NLRP3 Inflammasome Activation. Front Immunol 2022; 13:781003. [PMID: 35720321 PMCID: PMC9199371 DOI: 10.3389/fimmu.2022.781003] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Accepted: 04/29/2022] [Indexed: 11/13/2022] Open
Abstract
As an important transcription factor, heat shock factor 1 (HSF1) plays an endogenous anti-inflammation role in the body and can alleviate multiple organ dysfunction caused by sepsis, which contributes to an uncontrolled inflammatory response. The NLRP3 inflammasome is a supramolecular complex that plays key roles in immune surveillance. Inflammation is accomplished by NLRP3 inflammasome activation, which leads to the proteolytic maturation of IL-1β and pyroptosis. However, whether HSF1 is involved in the activation of the NLRP3 inflammasome in septic acute lung injury (ALI) has not been reported. Here, we show that HSF1 suppresses NLRP3 inflammasome activation in transcriptional and post-translational modification levels. HSF1 can repress NLRP3 expression via inhibiting NF-κB phosphorylation. HSF1 can inhibit caspase-1 activation and IL-1β maturation via promoting NLRP3 ubiquitination. Our finding not only elucidates a novel mechanism for HSF1-mediated protection of septic ALI but also identifies new therapeutic targets for septic ALI and related diseases.
Collapse
Affiliation(s)
- Xueyan Shi
- Sepsis Translational Medicine Key Laboratory of Hunan Province, Central South University, Changsha, China.,State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research, Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union, Medical College, Beijing, China
| | - Tao Li
- Sepsis Translational Medicine Key Laboratory of Hunan Province, Central South University, Changsha, China.,Department of Pathophysiology, Medical College of Jiaying University, Meizhou, China
| | - Yanting Liu
- Sepsis Translational Medicine Key Laboratory of Hunan Province, Central South University, Changsha, China.,Department of Pathophysiology, Xiangya School of Medicine, Central South University, Changsha, China
| | - Leijin Yin
- Sepsis Translational Medicine Key Laboratory of Hunan Province, Central South University, Changsha, China.,Department of Pathophysiology, Xiangya School of Medicine, Central South University, Changsha, China
| | - Lan Xiao
- Department of Traditional Chinese Medicine, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Liyao Fu
- Sepsis Translational Medicine Key Laboratory of Hunan Province, Central South University, Changsha, China.,The Second Xiangya Hospital, Central South University, Changsha, China
| | - Yaxi Zhu
- Sepsis Translational Medicine Key Laboratory of Hunan Province, Central South University, Changsha, China.,Department of Pathophysiology, Xiangya School of Medicine, Central South University, Changsha, China
| | - Huan Chen
- Sepsis Translational Medicine Key Laboratory of Hunan Province, Central South University, Changsha, China.,Department of Pathophysiology, Xiangya School of Medicine, Central South University, Changsha, China
| | - Kangkai Wang
- Sepsis Translational Medicine Key Laboratory of Hunan Province, Central South University, Changsha, China.,Department of Pathophysiology, Xiangya School of Medicine, Central South University, Changsha, China
| | - Xianzhong Xiao
- Sepsis Translational Medicine Key Laboratory of Hunan Province, Central South University, Changsha, China.,Department of Pathophysiology, Xiangya School of Medicine, Central South University, Changsha, China
| | - Huali Zhang
- Sepsis Translational Medicine Key Laboratory of Hunan Province, Central South University, Changsha, China.,Department of Pathophysiology, Xiangya School of Medicine, Central South University, Changsha, China
| | - Sichuang Tan
- The Second Xiangya Hospital, Central South University, Changsha, China
| | - Sipin Tan
- Sepsis Translational Medicine Key Laboratory of Hunan Province, Central South University, Changsha, China.,Department of Pathophysiology, Xiangya School of Medicine, Central South University, Changsha, China
| |
Collapse
|