1
|
Moss CG, Dilworth MR, Harris LK, Freeman S, Heazell AEP. Understanding a Potential Role for the NLRP3 Inflammasome in Placenta-Mediated Pregnancy Complications. Am J Reprod Immunol 2025; 93:e70077. [PMID: 40260875 PMCID: PMC12013246 DOI: 10.1111/aji.70077] [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: 12/18/2024] [Revised: 03/20/2025] [Accepted: 04/03/2025] [Indexed: 04/24/2025] Open
Abstract
Stillbirth affects approximately 2 million pregnancies annually and is closely linked to placental dysfunction, which may also present clinically as foetal growth restriction (FGR) or pre-eclampsia (PE). Placental dysfunction can arise from a range of insults, including the inflammatory conditions villitis of unknown aetiology (VUE) and chronic histiocytic intervillositis (CHI). Despite ample research regarding the pathophysiology of placental dysfunction, the literature surrounding placental inflammation is more limited, with no currently established treatments. In the absence of infection, placental inflammation is hypothesised to be stimulated by damage-associated molecular patterns (DAMPs), known as sterile inflammation. The NLRP3 inflammasome, a protein scaffold that unites within the cytosol of cells, is a proposed contributor. The NLRP3 inflammasome is dysregulated in numerous diseases and has shown evidence of activation through the sterile inflammatory pathway via DAMPs. Studies have demonstrated the upregulation of the NLRP3 inflammasome and its components in placentally-mediated pregnancy pathologies. However, the link between placental dysfunction seen in these disorders and the NLRP3 inflammasome is not yet firmly established. This manuscript aims to review the evidence regarding placental inflammation seen with placental dysfunction, discuss its association with the NLRP3 inflammasome, and identify potential therapeutic interventions for this pathological inflammatory response.
Collapse
Affiliation(s)
- Chloe G. Moss
- Maternal and Fetal Health Research CentreDivision of Developmental Biology and MedicineUniversity of ManchesterManchesterUK
- Manchester Academic Health Science CentreManchester University NHS Foundation TrustManchesterUK
| | - Mark R. Dilworth
- Maternal and Fetal Health Research CentreDivision of Developmental Biology and MedicineUniversity of ManchesterManchesterUK
- Manchester Academic Health Science CentreManchester University NHS Foundation TrustManchesterUK
| | - Lynda K. Harris
- Department of Obstetrics and GynaecologyOlson Center for Women's HealthUniversity of Nebraska Medical CentreOmahaUSA
| | - Sally Freeman
- Division of Pharmacy and OptometryUniversity of ManchesterManchesterUK
| | - Alexander E. P. Heazell
- Maternal and Fetal Health Research CentreDivision of Developmental Biology and MedicineUniversity of ManchesterManchesterUK
- Manchester Academic Health Science CentreManchester University NHS Foundation TrustManchesterUK
| |
Collapse
|
2
|
Vithalkar MP, Pradhan S, Sandra KS, Bharath HB, Nayak Y. Modulating NLRP3 Inflammasomes in Idiopathic Pulmonary Fibrosis: A Comprehensive Review on Flavonoid-Based Interventions. Cell Biochem Biophys 2025:10.1007/s12013-025-01696-4. [PMID: 39966334 DOI: 10.1007/s12013-025-01696-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/06/2025] [Indexed: 02/20/2025]
Abstract
Idiopathic Pulmonary Fibrosis (IPF) is a severe, rapidly advancing disease that drastically diminishes life expectancy. Without treatment, it can progress to lung cancer. The precise etiology of IPF remains unknown, but inflammation and damage to the alveolar epithelium are widely thought to be pivotal in its development. Research has indicated that activating the NLRP3 inflammasome is a crucial mechanism in IPF pathogenesis, as it triggers the release of pro-inflammatory cytokines such as IL-1β, IL-18, and TGF-β. These cytokines contribute to the myofibroblast differentiation and extracellular matrix (ECM) accumulation. Currently, treatment options for IPF are limited. Only two FDA-approved medications, pirfenidone and nintedanib, are available. While these drugs can decelerate disease progression, they come with a range of side effects and do not cure the disease. Additional treatment strategies primarily involve supportive care and therapy. Emerging research has highlighted that numerous flavonoids derived from traditional medicines can inhibit the critical regulators responsible for activating the NLRP3 inflammasome. These flavonoids show promise as potential therapeutic agents for managing IPF, offering a new avenue for treatment that targets the core inflammatory processes of this debilitating condition.
Collapse
Affiliation(s)
- Megh Pravin Vithalkar
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, Pin 576104, India
| | - Shreya Pradhan
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, Pin 576104, India
| | - K S Sandra
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, Pin 576104, India
| | - H B Bharath
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, Pin 576104, India
| | - Yogendra Nayak
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, Pin 576104, India.
| |
Collapse
|
3
|
Whitehead DM, Fischer C, Briard E, Farady CJ, Graveleau N, Karrer J, Kaupmann K, Lapointe G, Mackay A, Reichert L, Wright M, Mu L, Auberson YP. [ 18F]NP3-627, a Candidate PET Imaging Agent Targeting the NLRP3 Inflammasome in the Central Nervous System. ChemMedChem 2025; 20:e202400816. [PMID: 39540320 DOI: 10.1002/cmdc.202400816] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2024] [Revised: 11/12/2024] [Accepted: 11/13/2024] [Indexed: 11/16/2024]
Abstract
We describe the identification of a candidate positron emission tomography (PET) imaging agent for the NLRP3 protein. NLRP3 plays a critical role in the immune system and has proven a difficult target for the development of imaging agents due to its low and cell-specific expression profile. A recently described series of pyridazine-based inhibitors, with improved permeability and brain-penetration properties, was used as a starting point for the development of a suitable PET imaging agent. Optimization of affinity, non-specific binding and pharmacokinetic properties led to the identification of aminopyridazine (R)-2-(6-((1-cyclopropylpiperidin-3-yl)amino)pyridazin-3-yl)-5-fluoro-3-methylphenol (17 b), which meets the preclinical profile of a successful imaging agent, and whose tritiated version demonstrated excellent specificity in a radioligand saturation binding assay, confirming its imaging potential.18F labeling led to [18F]NP3-627, the proposed PET imaging agent.
Collapse
Affiliation(s)
- David M Whitehead
- Novartis Biomedical Research, Global Discovery Chemistry, Novartis Campus, WSJ-88.10.110, 4056, Basel, Switzerland
| | - Christian Fischer
- Novartis Biomedical Research, Global Discovery Chemistry, Novartis Campus, WSJ-88.10.110, 4056, Basel, Switzerland
| | - Emmanuelle Briard
- Novartis Biomedical Research, Global Discovery Chemistry, Novartis Campus, WSJ-88.10.110, 4056, Basel, Switzerland
| | - Christopher J Farady
- Novartis Biomedical Research, Immunology Research, Novartis Campus, WSJ-386, 4056, Basel, Switzerland
| | - Nadège Graveleau
- Novartis Biomedical Research, Global Discovery Chemistry, Novartis Campus, WSJ-88.10.110, 4056, Basel, Switzerland
| | - Joel Karrer
- Novartis Biomedical Research, Global Discovery Chemistry, Novartis Campus, WSJ-88.10.110, 4056, Basel, Switzerland
| | - Klemens Kaupmann
- Novartis Biomedical Research, Immunology Research, Novartis Campus, WSJ-386, 4056, Basel, Switzerland
| | - Guillaume Lapointe
- Novartis Biomedical Research, Global Discovery Chemistry, Novartis Campus, WSJ-88.10.110, 4056, Basel, Switzerland
| | - Angela Mackay
- Novartis Biomedical Research, Global Discovery Chemistry, Novartis Campus, WSJ-88.10.110, 4056, Basel, Switzerland
| | - Lisa Reichert
- ETH Zurich, Institute of Pharmaceutical Sciences, Vladimir-Prelog Weg 4, 8093, Zurich, Switzerland
| | - Michael Wright
- Novartis Biomedical Research, Global Discovery Chemistry, Novartis Campus, WSJ-88.10.110, 4056, Basel, Switzerland
| | - Linjing Mu
- ETH Zurich, Institute of Pharmaceutical Sciences, Vladimir-Prelog Weg 4, 8093, Zurich, Switzerland
| | - Yves P Auberson
- Novartis Biomedical Research, Global Discovery Chemistry, Novartis Campus, WSJ-88.10.110, 4056, Basel, Switzerland
| |
Collapse
|
4
|
Zhang X, Jin L, Wu Y, Huang B, Chen K, Huang W, Li J. Anti-inflammatory properties of biflavonoids derived from Selaginella moellendorffii Hieron: Targeting NLRP3 inflammasome-dependent pyroptosis. JOURNAL OF ETHNOPHARMACOLOGY 2025; 340:119172. [PMID: 39643022 DOI: 10.1016/j.jep.2024.119172] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/26/2024] [Accepted: 11/27/2024] [Indexed: 12/09/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Selaginella moellendorffii Hieron. has been used as ethnic drug for chronic inflammation treatment. Biflavonoids represent a crucial class of bioactive compounds recognized for their potent anti-inflammatory activity in S. moellendorffii (SM). However, the effective components, targets, and pathways that SM in anti-inflammasome remain unclear. AIM OF THE STUDY Therefore, this study initially evaluated the effective components of SM and explored the underlying mechanisms. MATERIALS AND METHODS Firstly, a series of biflavonoids were isolated from SM, and then all compounds were evaluated for their anti-inflammatory ability in the THP-macrophages co-stimulated with lipopolysaccharide (LPS) and NLRP3 inflammasome inducers. Secondly, transcriptomic analysis and metabolomics analysis revealed the differential genes and metabolites associated with effective components treatment. Finally, molecular docking of effective components with NLRP3 was performed and western blotting was performed in order to determine the expression of related proteins. RESULTS Overall, eleven biflavonoids were successfully isolated from SM. Particularly, F7 exhibited the most potent inhibitory effect against NLRP3 inflammasome-mediated cytokines levels, cell membrane integrity and Ca2+ influx. Transcriptomic studies demonstrated that the differential genes (DEGs) were mainly enriched in NF-κB signaling pathway and NOD-like receptor signaling pathway. Metabolomics studies that the metabolites were mainly involved the pyrimidine metabolites. Further validation analysis manifested that F7's significant downregulation of NLRP3 inflammasome-related genes and proteins expression (P < 0.05, P < 0.01), encompassing both priming (NLRP3, TNF-α, p-p65/p65) and activation stages (IL-1β, IL-18, Caspase-1, GSDMD-N/GSDMD). Moreover, NLRP3 knockdown attenuated F7-mediated inhibition of pyroptosis. Finally, in silico results showed that F7 exhibited promising predicted binding affinity towards NLRP3. CONCLUSIONS Collectively, these findings revealed an anti-inflammatory material basis for SM and confirmed F7 as a potent inhibitor of pyroptosis by suppressing NF-κB/NLRP3 Pathway.
Collapse
Affiliation(s)
- Xueyan Zhang
- Key Laboratory of Ministry of Education on Traditional Chinese Medicine Resource and Compound Prescription, Key Laboratory of Resources and Chemistry of Chinese Medicine, College of Pharmacy, Hubei University of Chinese Medicine, Huang-Jia-Hu West Road 16(#), Hongshan District, Wuhan, Hubei, 430065, China; Department of Pharmacy, Maternal and Child Health Hospital of Hubei Province, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430070, China
| | - Lu Jin
- Key Laboratory of Ministry of Education on Traditional Chinese Medicine Resource and Compound Prescription, Key Laboratory of Resources and Chemistry of Chinese Medicine, College of Pharmacy, Hubei University of Chinese Medicine, Huang-Jia-Hu West Road 16(#), Hongshan District, Wuhan, Hubei, 430065, China
| | - You Wu
- Key Laboratory of Ministry of Education on Traditional Chinese Medicine Resource and Compound Prescription, Key Laboratory of Resources and Chemistry of Chinese Medicine, College of Pharmacy, Hubei University of Chinese Medicine, Huang-Jia-Hu West Road 16(#), Hongshan District, Wuhan, Hubei, 430065, China
| | - Bisheng Huang
- Key Laboratory of Ministry of Education on Traditional Chinese Medicine Resource and Compound Prescription, Key Laboratory of Resources and Chemistry of Chinese Medicine, College of Pharmacy, Hubei University of Chinese Medicine, Huang-Jia-Hu West Road 16(#), Hongshan District, Wuhan, Hubei, 430065, China
| | - Keli Chen
- Key Laboratory of Ministry of Education on Traditional Chinese Medicine Resource and Compound Prescription, Key Laboratory of Resources and Chemistry of Chinese Medicine, College of Pharmacy, Hubei University of Chinese Medicine, Huang-Jia-Hu West Road 16(#), Hongshan District, Wuhan, Hubei, 430065, China
| | - Wei Huang
- Key Laboratory of Ministry of Education on Traditional Chinese Medicine Resource and Compound Prescription, Key Laboratory of Resources and Chemistry of Chinese Medicine, College of Pharmacy, Hubei University of Chinese Medicine, Huang-Jia-Hu West Road 16(#), Hongshan District, Wuhan, Hubei, 430065, China.
| | - Juan Li
- Key Laboratory of Ministry of Education on Traditional Chinese Medicine Resource and Compound Prescription, Key Laboratory of Resources and Chemistry of Chinese Medicine, College of Pharmacy, Hubei University of Chinese Medicine, Huang-Jia-Hu West Road 16(#), Hongshan District, Wuhan, Hubei, 430065, China.
| |
Collapse
|
5
|
Yang W, Zeng S, Shao R, Jin R, Huang J, Wang X, Liu E, Zhou T, Li F, Chen Y, Chen D. Sulforaphane regulation autophagy-mediated pyroptosis in autoimmune hepatitis via AMPK/mTOR pathway. Int Immunopharmacol 2025; 146:113826. [PMID: 39673998 DOI: 10.1016/j.intimp.2024.113826] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2024] [Revised: 11/28/2024] [Accepted: 12/07/2024] [Indexed: 12/16/2024]
Abstract
Autoimmune hepatitis (AIH) is a liver disease marked by inflammation of unknown origin. If untreated, it can progress to cirrhosis or liver failure, posing a significant health risk. Currently, effective drug therapies are lacking in clinical practice. Sulforaphane (SFN), a natural anti-inflammatory and antioxidant compound found in various cruciferous vegetables, alleviate pyroptosis and improve impaired autophagic flux, both of which contribute to AIH progression. However, whether SFN modulates autophagic flux and pyroptosis in S100-induced EAH through the AMPK/mTOR pathway remains unclear. Therefore, this study aims to investigate whether SFN can regulate AIH and elucidate its potential mechanisms of action. In this study, experimental AIH (EAH) was induced in male C57BL/6 J mice through intraperitoneal (i.p.) injection of S100. SFN was administered intraperitoneally every other day. After 28 days, the mice were euthanized, and their livers and serum were collected for histological and biochemical analyses. AML12 cells were used for the in vitro studies. The results showed that SFN mitigated pyroptosis by inhibiting the NLRP3 inflammasome and improving autophagic flux, which alleviates S100-induced EAH. Conversely, the autophagy inhibitor 3-MA negated the protective effects of SFN against inflammasome-mediated pyroptosis. Furthermore, SFN activated the AMPK/mTOR signaling pathway, offering protection against S100-induced EAH. Selective inhibition of AMPK suppressed the improvement in autophagic flux and protected against SFN-induced pyroptosis. Overall, SFN significantly ameliorates S100-induced EAH by enhancing autophagic flux and mitigating pyroptosis through activation of the AMPK/mTOR signaling pathway.
Collapse
Affiliation(s)
- Weijian Yang
- Hepatology Diagnosis and Treatment Center, The First Affiliated Hospital of Wenzhou Medical University and Zhejiang Provincial Key Laboratory for Accurate Diagnosis and Treatment of Chronic Liver Diseases, Wenzhou 325035, China
| | - Shiyi Zeng
- Hepatology Diagnosis and Treatment Center, The First Affiliated Hospital of Wenzhou Medical University and Zhejiang Provincial Key Laboratory for Accurate Diagnosis and Treatment of Chronic Liver Diseases, Wenzhou 325035, China
| | - Rongrong Shao
- Hepatology Diagnosis and Treatment Center, The First Affiliated Hospital of Wenzhou Medical University and Zhejiang Provincial Key Laboratory for Accurate Diagnosis and Treatment of Chronic Liver Diseases, Wenzhou 325035, China
| | - Ru Jin
- Hepatology Diagnosis and Treatment Center, The First Affiliated Hospital of Wenzhou Medical University and Zhejiang Provincial Key Laboratory for Accurate Diagnosis and Treatment of Chronic Liver Diseases, Wenzhou 325035, China
| | - Jiayin Huang
- Hepatology Diagnosis and Treatment Center, The First Affiliated Hospital of Wenzhou Medical University and Zhejiang Provincial Key Laboratory for Accurate Diagnosis and Treatment of Chronic Liver Diseases, Wenzhou 325035, China
| | - Xinyu Wang
- Hepatology Diagnosis and Treatment Center, The First Affiliated Hospital of Wenzhou Medical University and Zhejiang Provincial Key Laboratory for Accurate Diagnosis and Treatment of Chronic Liver Diseases, Wenzhou 325035, China
| | - Enqian Liu
- Hepatology Diagnosis and Treatment Center, The First Affiliated Hospital of Wenzhou Medical University and Zhejiang Provincial Key Laboratory for Accurate Diagnosis and Treatment of Chronic Liver Diseases, Wenzhou 325035, China
| | - Tenghui Zhou
- Hepatology Diagnosis and Treatment Center, The First Affiliated Hospital of Wenzhou Medical University and Zhejiang Provincial Key Laboratory for Accurate Diagnosis and Treatment of Chronic Liver Diseases, Wenzhou 325035, China
| | - Fengfan Li
- Hepatology Diagnosis and Treatment Center, The First Affiliated Hospital of Wenzhou Medical University and Zhejiang Provincial Key Laboratory for Accurate Diagnosis and Treatment of Chronic Liver Diseases, Wenzhou 325035, China
| | - Yongping Chen
- Hepatology Diagnosis and Treatment Center, The First Affiliated Hospital of Wenzhou Medical University and Zhejiang Provincial Key Laboratory for Accurate Diagnosis and Treatment of Chronic Liver Diseases, Wenzhou 325035, China.
| | - Dazhi Chen
- Hepatology Diagnosis and Treatment Center, The First Affiliated Hospital of Wenzhou Medical University and Zhejiang Provincial Key Laboratory for Accurate Diagnosis and Treatment of Chronic Liver Diseases, Wenzhou 325035, China; School of Clinical Medicine, The First People's Hospital of Lin'an District, Hangzhou, Lin'an People's Hospital Affiliated to Hangzhou Medical College, Hangzhou Medical College, Hangzhou 311300, China.
| |
Collapse
|
6
|
Urrutia‐Ortega IM, Valencia I, Ispanixtlahuatl‐Meraz O, Benítez‐Flores JC, Espinosa‐González AM, Estrella‐Parra EA, Flores‐Ortiz CM, Chirino YI, Avila‐Acevedo JG. Full-spectrum cannabidiol reduces UVB damage through the inhibition of TGF-β1 and the NLRP3 inflammasome. Photochem Photobiol 2025; 101:83-105. [PMID: 38958000 PMCID: PMC11737019 DOI: 10.1111/php.13993] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Revised: 06/11/2024] [Accepted: 06/13/2024] [Indexed: 07/04/2024]
Abstract
The thermodynamic characteristics, antioxidant potential, and photoprotective benefits of full-spectrum cannabidiol (FS-CBD) against UVB-induced cellular death were examined in this study. In silico analysis of CBD showed antioxidant capacity via proton donation and UV absorption at 209.09, 254.73, and 276.95 nm, according to the HAT and SPLET methodologies. FS-CBD protected against UVB-induced bacterial death for 30 min. FS-CBD protected against UVB-induced cell death by 42% (1.5 μg/mL) and 35% (3.5 μg/mL) in an in vitro keratinocyte cell model. An in vivo acute irradiated CD-1et/et mouse model (UVB-irradiated for 5 min) presented very low photoprotection when FS-CBD was applied cutaneously, as determined by histological analyses. In vivo skin samples showed that FS-CBD regulated inflammatory responses by inhibiting the inflammatory markers TGF-β1 and NLRP3. The docking analysis showed that the CBD molecule had a high affinity for TGF-β1 and NLRP3, indicating that protection against inflammation might be mediated by blocking these proinflammatory molecules. This result was corroborated by the docking interactions between CBD and TGF-β1 and NLRP3, which resulted in a high affinity and inhibition of both proteins The present work suggested a FS-CBD moderate photoprotective agent against UVB light-induced skin damage and that this effect is partially mediated by its anti-inflammatory activity.
Collapse
Affiliation(s)
- I. M. Urrutia‐Ortega
- Laboratorio de Carcinogénesis y Toxicología, Unidad de Investigación en Biomedicina, Facultad de Estudios Superiores IztacalaUniversidad Nacional Autónoma de MéxicoTlalnepantla de BazEstado de MéxicoMexico
- Laboratorio de Fitoquímica, Unidad de Biotecnología y Prototipos, Facultad de Estudios Superiores IztacalaUniversidad Nacional Autónoma de MéxicoTlalnepantla de BazEstado de MéxicoMexico
| | - I. Valencia
- Laboratorio de Fitoquímica, Unidad de Biotecnología y Prototipos, Facultad de Estudios Superiores IztacalaUniversidad Nacional Autónoma de MéxicoTlalnepantla de BazEstado de MéxicoMexico
| | - O. Ispanixtlahuatl‐Meraz
- Laboratorio de Carcinogénesis y Toxicología, Unidad de Investigación en Biomedicina, Facultad de Estudios Superiores IztacalaUniversidad Nacional Autónoma de MéxicoTlalnepantla de BazEstado de MéxicoMexico
| | - J. C. Benítez‐Flores
- Laboratorio de Histología, Unidad de Morfología y Función, Facultad de Estudios Superiores IztacalaUniversidad Nacional Autónoma de MéxicoTlalnepantla de BazEstado de MéxicoMexico
| | - A. M. Espinosa‐González
- Laboratorio de Fitoquímica, Unidad de Biotecnología y Prototipos, Facultad de Estudios Superiores IztacalaUniversidad Nacional Autónoma de MéxicoTlalnepantla de BazEstado de MéxicoMexico
| | - E. A. Estrella‐Parra
- Laboratorio de Fitoquímica, Unidad de Biotecnología y Prototipos, Facultad de Estudios Superiores IztacalaUniversidad Nacional Autónoma de MéxicoTlalnepantla de BazEstado de MéxicoMexico
| | - C. M. Flores‐Ortiz
- Laboratorio de Fisiología Vegetal, Facultad de Estudios Superiores IztacalaUniversidad Nacional Autónoma de MéxicoTlalnepantla de BazEstado de MéxicoMexico
- Laboratorio Nacional en Salud, Facultad de Estudios Superiores IztacalaUniversidad Nacional Autónoma de MéxicoTlalnepantla de BazEstado de MéxicoMexico
| | - Y. I. Chirino
- Laboratorio de Carcinogénesis y Toxicología, Unidad de Investigación en Biomedicina, Facultad de Estudios Superiores IztacalaUniversidad Nacional Autónoma de MéxicoTlalnepantla de BazEstado de MéxicoMexico
| | - J. G. Avila‐Acevedo
- Laboratorio de Fitoquímica, Unidad de Biotecnología y Prototipos, Facultad de Estudios Superiores IztacalaUniversidad Nacional Autónoma de MéxicoTlalnepantla de BazEstado de MéxicoMexico
| |
Collapse
|
7
|
Li XQ, Gu YQ, Ling YY, Wang M, Miao J, Xue L, Ji W, Liu J. Association between mitophagy and NLRP3 inflammasome in uric acid nephropathy. Ren Fail 2024; 46:2438847. [PMID: 39681479 DOI: 10.1080/0886022x.2024.2438847] [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: 02/05/2024] [Revised: 11/24/2024] [Accepted: 12/02/2024] [Indexed: 12/18/2024] Open
Abstract
OBJECTIVE This study was recruited to investigate the role of mitophagy in activating NLRP3 inflammasome in the kidney of uric acid (UA) nephropathy (UAN) rats. METHODS This study developed a uric acid nephropathy (UAN) rat model divided into five groups: Negative control (NC), UAN model (M), UAN + autophagy inhibitor (3-MA), UAN + lysosome inhibitor (CQ), and ROS scavenger (N-acetylcysteine, N). H&E staining assessed renal structure, ROS levels were measured with 2, 7-dichlorofluorescin diacetate, and ELISA measured serum markers (creatinine, UA, cystatin C, NGAL, IL-1β, IL-18). Western blot and qRT-PCR evaluated autophagy and inflammation-related protein (LC3 II/I, p62, Pink1, Parkin, NLRP3, Caspase1, IL-1β) expression. NRK-52E cells treated with uric acid and shRNA were analyzed by western blot. RESULTS Renal injury in UAN rats was aggravated by ROS accumulation, which promoted mitophagy and activated the NLRP3 inflammasome. Eliminating ROS reduced mitophagy, inhibited NLRP3 activation, lowered IL-1β and IL-18 levels, and alleviated renal injury. Notably, inhibiting mitophagy increased ROS accumulation, up-regulated NLRP3, Caspase1, and IL-1β expression, further worsening renal injury. In vitro, uric acid treatment of NRK-52E cells altered autophagy-related protein and pro-inflammatory cytokine levels, highlighting the interplay between mitophagy and inflammation in uric acid nephropathy. CONCLUSION Mitophagy influences renal injury in uric acid nephropathy (UAN) by regulating ROS accumulation and NLRP3 inflammasome activation, suggesting that mitophagy may serve as a potential therapeutic target for UAN.
Collapse
Affiliation(s)
- Xiao-Qian Li
- Department of Nephrology, Nantong Hospital to Nanjing University of Chinese Medicine, Nantong Hospital of Traditional Chinese Medicine, Nantong, Jiangsu, China
| | - Yong-Qing Gu
- Department of Cardiology, Nantong Hospital to Nanjing University of Chinese Medicine, Nantong Hospital of Traditional Chinese Medicine, Nantong, Jiangsu, China
| | - Yuan-Yuan Ling
- Department of Geriatrics, Nantong Hospital to Nanjing University of Chinese Medicine, Nantong Hospital of Traditional Chinese Medicine, Nantong, Jiangsu, China
| | - Mei Wang
- Department of Pathology, Nantong Hospital to Nanjing University of Chinese Medicine, Nantong Hospital of Traditional Chinese Medicine, Nantong, Jiangsu, China
| | - Jin Miao
- Laboratory Animal Center, Nantong University, Nantong, Jiangsu, China
| | - Li Xue
- Department of Nephrology, Nantong Hospital to Nanjing University of Chinese Medicine, Nantong Hospital of Traditional Chinese Medicine, Nantong, Jiangsu, China
| | - Wei Ji
- Department of Rheumatology, Jiangsu Province Hospital of Chinese Medicine, Nanjing, Jiangsu, China
| | - Jun Liu
- Department of Nephrology, Nantong Hospital to Nanjing University of Chinese Medicine, Nantong Hospital of Traditional Chinese Medicine, Nantong, Jiangsu, China
| |
Collapse
|
8
|
Tantra T, Rahaman T A A, Nandini, Chaudhary S. Therapeutic role of NLRP3 inflammasome inhibitors against Alzheimer's disease. Bioorg Chem 2024; 153:107912. [PMID: 39504636 DOI: 10.1016/j.bioorg.2024.107912] [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: 07/27/2024] [Revised: 09/18/2024] [Accepted: 10/21/2024] [Indexed: 11/08/2024]
Abstract
The NLRP3 inflammasome is a multiprotein complex that plays a vital role in regulating inflammatory signaling and the innate immune system. Activation of NLRP3 by accumulation of Aβ leads to its oligomerization and the activation of caspase-1, resulting in the secretion of pro-cytokines such as IL-18 and IL-1β. These pro-cytokines can contribute to cognitive impairment and neurodegeneration. The activation of NLRP3 is associated with neuroinflammation in animal models of Alzheimer's disease (AD). Therefore, the NLRP3 inflammasome is considered a potential therapeutic target for AD. Various natural and synthetic molecules have gained attention as NLRP3 inhibitors against AD. In this review, we will summarize the sources, chemical structures, synthesis, and biological activity of NLRP3 inhibitors as anti-Alzheimer's agents. Additionally, we will critically analyze the structure-activity relationship (SAR) of NLRP3 inhibitors. This detailed examination of the SAR-based investigation of NLRP3 inhibitors and their derivatives offers insights into the design and development of novel NLRP3 inhibitors as anti-Alzheimer's agents. It is expected that this review will assist researchers in developing innovative and effective NLRP3 inhibitors for the treatment of AD.
Collapse
Affiliation(s)
- Tanmoy Tantra
- Laboratory of Bioactive Heterocycles and Catalysis (BHC Lab), Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research-Raebareli (Transit Campus), Bijnor-Sisendi Road, Near CRPF Base Camp, Sarojini Nagar, Lucknow 226002, India
| | - Abdul Rahaman T A
- Laboratory of Bioactive Heterocycles and Catalysis (BHC Lab), Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research-Raebareli (Transit Campus), Bijnor-Sisendi Road, Near CRPF Base Camp, Sarojini Nagar, Lucknow 226002, India
| | - Nandini
- Laboratory of Bioactive Heterocycles and Catalysis (BHC Lab), Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research-Raebareli (Transit Campus), Bijnor-Sisendi Road, Near CRPF Base Camp, Sarojini Nagar, Lucknow 226002, India
| | - Sandeep Chaudhary
- Laboratory of Bioactive Heterocycles and Catalysis (BHC Lab), Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research-Raebareli (Transit Campus), Bijnor-Sisendi Road, Near CRPF Base Camp, Sarojini Nagar, Lucknow 226002, India.
| |
Collapse
|
9
|
Cheng L, Hu Z, Gu J, Li Q, Liu J, Liu M, Li J, Bi X. Exploring COX-Independent Pathways: A Novel Approach for Meloxicam and Other NSAIDs in Cancer and Cardiovascular Disease Treatment. Pharmaceuticals (Basel) 2024; 17:1488. [PMID: 39598398 PMCID: PMC11597362 DOI: 10.3390/ph17111488] [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: 10/06/2024] [Revised: 11/03/2024] [Accepted: 11/04/2024] [Indexed: 11/29/2024] Open
Abstract
As a fundamental process of innate immunity, inflammation is associated with the pathologic process of various diseases and constitutes a prevalent risk factor for both cancer and cardiovascular disease (CVD). Studies have indicated that several non-steroidal anti-inflammatory drugs (NSAIDs), including Meloxicam, may prevent tumorigenesis, reduce the risk of carcinogenesis, improve the efficacy of anticancer therapies, and reduce the risk of CVD, in addition to controlling the body's inflammatory imbalances. Traditionally, most NSAIDs work by inhibiting cyclooxygenase (COX) activity, thereby blocking the synthesis of prostaglandins (PGs), which play a role in inflammation, cancer, and various cardiovascular conditions. However, long-term COX inhibition and reduced PGs synthesis can result in serious side effects. Recent studies have increasingly shown that some selective COX-2 inhibitors and NSAIDs, such as Meloxicam, may exert effects beyond COX inhibition. This emerging understanding prompts a re-evaluation of the mechanisms by which NSAIDs operate, suggesting that their benefits in cancer and CVD treatment may not solely depend on COX targeting. In this review, we will explore the potential COX-independent mechanisms of Meloxicam and other NSAIDs in addressing oncology and cardiovascular health.
Collapse
Affiliation(s)
- Lixia Cheng
- Department of Medical Genetics and Cell Biology, School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang 330006, China; (L.C.); (Z.H.); (Q.L.); (J.L.); (M.L.); (J.L.)
| | - Zhenghui Hu
- Department of Medical Genetics and Cell Biology, School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang 330006, China; (L.C.); (Z.H.); (Q.L.); (J.L.); (M.L.); (J.L.)
| | - Jiawei Gu
- Department of Precision Genomics, Intermountain Healthcare, 5121 Cottonwood St., Murray, UT 84107, USA;
| | - Qian Li
- Department of Medical Genetics and Cell Biology, School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang 330006, China; (L.C.); (Z.H.); (Q.L.); (J.L.); (M.L.); (J.L.)
| | - Jiahao Liu
- Department of Medical Genetics and Cell Biology, School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang 330006, China; (L.C.); (Z.H.); (Q.L.); (J.L.); (M.L.); (J.L.)
| | - Meiling Liu
- Department of Medical Genetics and Cell Biology, School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang 330006, China; (L.C.); (Z.H.); (Q.L.); (J.L.); (M.L.); (J.L.)
| | - Jie Li
- Department of Medical Genetics and Cell Biology, School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang 330006, China; (L.C.); (Z.H.); (Q.L.); (J.L.); (M.L.); (J.L.)
| | - Xiaowen Bi
- Department of Medical Genetics and Cell Biology, School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang 330006, China; (L.C.); (Z.H.); (Q.L.); (J.L.); (M.L.); (J.L.)
| |
Collapse
|
10
|
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
|
11
|
Sun C, Jiang Y, Li C, Sun S, Lin J, Wang W, Zhou L, Li L, Shah M, Che Q, Zhang G, Wang D, Zhu T, Li D. Discovery, Total Synthesis, and Anti-Inflammatory Evaluation of Naturally Occurring Naphthopyrone-Macrolide Hybrids as Potent NLRP3 Inflammasome Inhibitors. Angew Chem Int Ed Engl 2024; 63:e202405860. [PMID: 38837604 DOI: 10.1002/anie.202405860] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Revised: 06/02/2024] [Accepted: 06/04/2024] [Indexed: 06/07/2024]
Abstract
Numerous clinical disorders have been linked to the etiology of dysregulated NLRP3 (NACHT, LRR, and PYD domain-containing protein 3) inflammasome activation. Despite its potential as a pharmacological target, modulation of NLRP3 activity remains challenging. Only a sparse number of compounds have been reported that can modulate NLRP3 and none of them have been developed into a commercially available drug. In this research, we identified three potent NLRP3 inflammasome inhibitors, gymnoasins A-C (1-3), with unprecedented pentacyclic scaffolds, from an Antarctic fungus Pseudogymnoascus sp. HDN17-895, which represent the first naturally occurring naphthopyrone-macrolide hybrids. Additionally, biomimetic synthesis of gymnoasin A (1) was also achieved validating the chemical structure and affording ample amounts of material for exhaustive bioactivity assessments. Biological assays indicated that 1 could significantly inhibited in vitro NLRP3 inflammasome activation and in vivo pro-inflammatory cytokine IL-1β release, representing a valuable new lead compound for the development of novel therapeutics with the potential to inhibit the NLRP3 inflammasome.
Collapse
Affiliation(s)
- Chunxiao Sun
- Key Laboratory of Marine Drugs, Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao, 266100, China
| | - Yuqi Jiang
- Key Laboratory of Marine Drugs, Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao, 266100, China
- Laboratory for Marine Drugs and Bioproducts, Qingdao National Laboratory for Marine Science and Technology, Qingdao, 266237, China
- Marine Biomedical Research Institute of Qingdao, Qingdao, Shandong, 266071, China
| | - Changlong Li
- Key Laboratory of Marine Drugs, Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao, 266100, China
| | - Simin Sun
- Key Laboratory of Marine Drugs, Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao, 266100, China
| | - Jiaqi Lin
- Key Laboratory of Marine Drugs, Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao, 266100, China
| | - Wenxue Wang
- Key Laboratory of Marine Drugs, Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao, 266100, China
| | - Luning Zhou
- Key Laboratory of Marine Drugs, Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao, 266100, China
| | - Liping Li
- Key Laboratory of Marine Drugs, Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao, 266100, China
| | - Mudassir Shah
- Department of Pharmacy, Abbottabad University of Science and Technology, Havellian, District, Abbottabad, 22010 KPK, Pakistan
| | - Qian Che
- Key Laboratory of Marine Drugs, Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao, 266100, China
| | - Guojian Zhang
- Key Laboratory of Marine Drugs, Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao, 266100, China
- Laboratory for Marine Drugs and Bioproducts, Qingdao National Laboratory for Marine Science and Technology, Qingdao, 266237, China
- Marine Biomedical Research Institute of Qingdao, Qingdao, Shandong, 266071, China
| | - De Wang
- Key Laboratory of Marine Drugs, Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao, 266100, China
- Laboratory for Marine Drugs and Bioproducts, Qingdao National Laboratory for Marine Science and Technology, Qingdao, 266237, China
- Marine Biomedical Research Institute of Qingdao, Qingdao, Shandong, 266071, China
| | - Tianjiao Zhu
- Key Laboratory of Marine Drugs, Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao, 266100, China
| | - Dehai Li
- Key Laboratory of Marine Drugs, Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao, 266100, China
- Laboratory for Marine Drugs and Bioproducts, Qingdao National Laboratory for Marine Science and Technology, Qingdao, 266237, China
- Marine Biomedical Research Institute of Qingdao, Qingdao, Shandong, 266071, China
| |
Collapse
|
12
|
Lin TH, Chiu YJ, Lin CH, Chen YR, Lin W, Wu YR, Chang KH, Chen CM, Lee-Chen GJ. Coumarin-chalcone derivatives as dual NLRP1 and NLRP3 inflammasome inhibitors targeting oxidative stress and inflammation in neurotoxin-induced HMC3 and BE(2)-M17 cell models of Parkinson's disease. Front Aging Neurosci 2024; 16:1437138. [PMID: 39411284 PMCID: PMC11473416 DOI: 10.3389/fnagi.2024.1437138] [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: 05/23/2024] [Accepted: 09/16/2024] [Indexed: 10/19/2024] Open
Abstract
Background In Parkinson's disease (PD) brains, microglia are activated to release inflammatory factors to induce the production of reactive oxygen species (ROS) in neuron, and vice versa. Moreover, neuroinflammation and its synergistic interaction with oxidative stress contribute to the pathogenesis of PD. Methods In this study, we investigated whether in-house synthetic coumarin-chalcone derivatives protect human microglia HMC3 and neuroblastoma BE(2)-M17 cells against 1-methyl-4-phenyl pyridinium (MPP+)-induced neuroinflammation and associated neuronal damage. Results Treatment with MPP+ decreased cell viability as well as increased the release of inflammatory mediators including cytokines and nitric oxide in culture medium, and enhanced expression of microglial activation markers CD68 and MHCII in HMC3 cells. The protein levels of NLRP3, CASP1, iNOS, IL-1β, IL-6, and TNF-α were also increased in MPP+-stimulated HMC3 cells. Among the four tested compounds, LM-016, LM-021, and LM-036 at 10 μM counteracted the inflammatory action of MPP+ in HMC3 cells. In addition, LM-021 and LM-036 increased cell viability, reduced lactate dehydrogenase release, ameliorated cellular ROS production, decreased caspase-1, caspase-3 and caspase-6 activities, and promoted neurite outgrowth in MPP+-treated BE(2)-M17 cells. These protective effects were mediated by down-regulating inflammatory NLRP1, IL-1β, IL-6, and TNF-α, as well as up-regulating antioxidative NRF2, NQO1, GCLC, and PGC-1α, and neuroprotective CREB, BDNF, and BCL2. Conclusion The study results strengthen the involvement of neuroinflammation and oxidative stress in PD pathogenic mechanisms, and indicate the potential use of LM-021 and LM-036 as dual inflammasome inhibitors in treating both NLRP1- and NLRP3-associated PD.
Collapse
Affiliation(s)
- Te-Hsien Lin
- Department of Neurology, Chang Gung Memorial Hospital, Chang Gung University School of Medicine, Taoyuan, Taiwan
| | - Ya-Jen Chiu
- Department of Life Science, National Taiwan Normal University, Taipei, Taiwan
| | - Chih-Hsin Lin
- Department of Neurology, Chang Gung Memorial Hospital, Chang Gung University School of Medicine, Taoyuan, Taiwan
| | - Yi-Ru Chen
- Department of Chemistry, National Taiwan Normal University, Taipei, Taiwan
| | - Wenwei Lin
- Department of Chemistry, National Taiwan Normal University, Taipei, Taiwan
| | - Yih-Ru Wu
- Department of Neurology, Chang Gung Memorial Hospital, Chang Gung University School of Medicine, Taoyuan, Taiwan
| | - Kuo-Hsuan Chang
- Department of Neurology, Chang Gung Memorial Hospital, Chang Gung University School of Medicine, Taoyuan, Taiwan
| | - Chiung-Mei Chen
- Department of Neurology, Chang Gung Memorial Hospital, Chang Gung University School of Medicine, Taoyuan, Taiwan
| | - Guey-Jen Lee-Chen
- Department of Life Science, National Taiwan Normal University, Taipei, Taiwan
| |
Collapse
|
13
|
Tian Y, He X, Li R, Wu Y, Ren Q, Hou Y. Recent advances in the treatment of gout with NLRP3 inflammasome inhibitors. Bioorg Med Chem 2024; 112:117874. [PMID: 39167977 DOI: 10.1016/j.bmc.2024.117874] [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: 05/27/2024] [Revised: 08/04/2024] [Accepted: 08/12/2024] [Indexed: 08/23/2024]
Abstract
Gout is an autoinflammatory disorder characterized by the accumulation of monosodium urate crystals in joints and other tissues, representing the predominant type of inflammatory arthritis with a notable prevalence and propensity for severe outcomes. The NLRP3 inflammasome, a member of the pyrin domain-containing NOD-like receptor family, exerts a substantial impact on both innate and adaptive immune responses and serves as a pivotal factor in the pathogenesis of gout. In recent years, there has been significant academic and industrial interest in the development of NLRP3-targeted small molecule inhibitors as a promising therapeutic approach for gout. To assess the advancements in NLRP3 inflammasome inhibitors for gout treatment, this review offers a comprehensive analysis and evaluation of current clinical candidates and other inhibitors targeting NLRP3 inflammasome from a chemical structure standpoint, with the goal of identifying more efficacious options for clinical management of gout.
Collapse
Affiliation(s)
- Ye Tian
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 610072, China
| | - Xiaofang He
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 610072, China
| | - Ruping Li
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 610072, China
| | - Yanxin Wu
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 610072, China
| | - Qiang Ren
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu 610072, China.
| | - Yusen Hou
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 610072, China; State Key Laboratory of Southwestern Chinese Medicine Resources, College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 610072, China.
| |
Collapse
|
14
|
Al-Madhagi H, Muhammed MT. Targeting COVID-19 and varicocele by blocking inflammasome: Ligand-based virtual screening. Arch Biochem Biophys 2024; 759:110107. [PMID: 39074718 DOI: 10.1016/j.abb.2024.110107] [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: 05/12/2024] [Revised: 07/22/2024] [Accepted: 07/26/2024] [Indexed: 07/31/2024]
Abstract
COVID-19 is a new generation of outbreaks that invade not only local emerging region, continental but also the whole globe. Varicocele on the other hand, is a testicular vascular disease that underlies 40 % of male infertility cases. Fortunately, the two diseases can be blocked through targeting one common target, NLRP3 inflammasome. Upon searching for similar drugs that gained FDA-approval in ChEMBL library along with examining their potential blockade of the receptor through docking using CB-DOCK-2, three potential approved drugs can be repurposed, ChEMBL 4297185, ChEMBL 1201749, ChEMBL 1200545 which had binding energy of -9.8 and -9.7 kcal/mol (stronger than the reference inhibitor, -9.3 kcal/mol). Also, ADME profile of the top 3 drugs showed better attributes. Also, the simulated proteins exhibited stable pattern with strong free binding energies. Among the potential inhibitor drugs ChEMBL 4297185 was found to remain inside the binding site of the protein during the 200 ns simulation time. Hence, it is anticipated to have the highest binding and thus inhibition potential against the protein. The suggested drugs, especially ChEMBL 4297185, are potentially repurposable toward treating COVID-19 and varicocele which deserve further experimental validation.
Collapse
Affiliation(s)
| | - Muhammed Tilahun Muhammed
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Suleyman Demirel University, Isparta, Turkiye.
| |
Collapse
|
15
|
Gu J, Xu X, Li X, Yue L, Zhu X, Chen Q, Gao J, Takashi M, Zhao W, Zhao B, Zhang Y, Lin M, Zhou J, Liang Y, Dai S, Pan Y, Shao Q, Li Y, Wang Y, Xu Z, Qian Q, Huang T, Qian X, Lu L. Tumor-resident microbiota contributes to colorectal cancer liver metastasis by lactylation and immune modulation. Oncogene 2024; 43:2389-2404. [PMID: 38890429 PMCID: PMC11281901 DOI: 10.1038/s41388-024-03080-7] [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: 02/28/2024] [Revised: 05/31/2024] [Accepted: 06/05/2024] [Indexed: 06/20/2024]
Abstract
The role of tumor-resident microbiota in modulating tumor immunity remains unclear. Here, we discovered an abundance of intra-tumoral bacteria, such us E.coli, residing and resulting in Colorectal cancer liver metastasis (CRLM). E.coli enhanced lactate production, which mediated M2 macrophage polarization by suppressing nuclear factor-κB -gene binding (NF-κB) signaling through retinoic acid-inducible gene 1 (RIG-I) lactylation. Lactylation of RIG-I suppressed recruitment of NF-κB to the Nlrp3 promoter in macrophages, thereby reducing its transcription. This loss of Nlrp3 affected the immunosuppressive activities of regulatory T cells (Tregs) and the antitumor activities of and CD8+ T cells. Small-molecule compound screening identified a RIG-I lactylation inhibitor that suppressed M2 polarization and sensitized CRLM to 5-fluorouracil (5-FU). Our findings suggest that tumor-resident microbiota may be a potential target for preventing and treating CRLM.
Collapse
Affiliation(s)
- Jian Gu
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University and Research Unit of Liver Transplantation and Transplant Immunology, Chinese Academy of Medical Sciences, Nanjing, China.
- Jiangsu Key Laboratory of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, China.
| | - Xiaozhang Xu
- Department of General Surgery, The Affiliated BenQ Hospital of Nanjing Medical University, Nanjing, China
| | - Xiangyu Li
- Department of General Surgery, The Affiliated BenQ Hospital of Nanjing Medical University, Nanjing, China
| | - Lei Yue
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, China
| | - Xiaowen Zhu
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University and Research Unit of Liver Transplantation and Transplant Immunology, Chinese Academy of Medical Sciences, Nanjing, China
- Jiangsu Key Laboratory of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, China
| | - Qiuyang Chen
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University and Research Unit of Liver Transplantation and Transplant Immunology, Chinese Academy of Medical Sciences, Nanjing, China
- Jiangsu Key Laboratory of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, China
| | - Ji Gao
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University and Research Unit of Liver Transplantation and Transplant Immunology, Chinese Academy of Medical Sciences, Nanjing, China
- Jiangsu Key Laboratory of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, China
| | | | - Wenhu Zhao
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University and Research Unit of Liver Transplantation and Transplant Immunology, Chinese Academy of Medical Sciences, Nanjing, China
- Jiangsu Key Laboratory of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, China
| | - Bo Zhao
- National and Local Joint Engineering Research Center of Biomedical Functional Materials, School of Chemistry and Materials Science, Nanjing Normal University, Nanjing, China
| | - Yue Zhang
- National and Local Joint Engineering Research Center of Biomedical Functional Materials, School of Chemistry and Materials Science, Nanjing Normal University, Nanjing, China
| | - Minjie Lin
- The Clinical Skills Training Center, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Jinren Zhou
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University and Research Unit of Liver Transplantation and Transplant Immunology, Chinese Academy of Medical Sciences, Nanjing, China
- Jiangsu Key Laboratory of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, China
| | - Yuan Liang
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University and Research Unit of Liver Transplantation and Transplant Immunology, Chinese Academy of Medical Sciences, Nanjing, China
- Jiangsu Key Laboratory of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, China
- School of Biological Science & Medical Engineering, Southeast University, Nanjing, China
| | - Shipeng Dai
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University and Research Unit of Liver Transplantation and Transplant Immunology, Chinese Academy of Medical Sciences, Nanjing, China
- Jiangsu Key Laboratory of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, China
| | - Yufeng Pan
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University and Research Unit of Liver Transplantation and Transplant Immunology, Chinese Academy of Medical Sciences, Nanjing, China
- Jiangsu Key Laboratory of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, China
- School of Medicine, Southeast University, Nanjing, China
| | - Qing Shao
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University and Research Unit of Liver Transplantation and Transplant Immunology, Chinese Academy of Medical Sciences, Nanjing, China
- Jiangsu Key Laboratory of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, China
| | - Yu Li
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University and Research Unit of Liver Transplantation and Transplant Immunology, Chinese Academy of Medical Sciences, Nanjing, China
- Jiangsu Key Laboratory of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, China
| | - Yiming Wang
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University and Research Unit of Liver Transplantation and Transplant Immunology, Chinese Academy of Medical Sciences, Nanjing, China
- Jiangsu Key Laboratory of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, China
| | - Zibo Xu
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University and Research Unit of Liver Transplantation and Transplant Immunology, Chinese Academy of Medical Sciences, Nanjing, China
- Jiangsu Key Laboratory of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, China
| | - Qufei Qian
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University and Research Unit of Liver Transplantation and Transplant Immunology, Chinese Academy of Medical Sciences, Nanjing, China
- Jiangsu Key Laboratory of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, China
| | - Tianning Huang
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University and Research Unit of Liver Transplantation and Transplant Immunology, Chinese Academy of Medical Sciences, Nanjing, China
- Jiangsu Key Laboratory of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, China
| | - Xiaofeng Qian
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University and Research Unit of Liver Transplantation and Transplant Immunology, Chinese Academy of Medical Sciences, Nanjing, China.
- Jiangsu Key Laboratory of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, China.
| | - Ling Lu
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University and Research Unit of Liver Transplantation and Transplant Immunology, Chinese Academy of Medical Sciences, Nanjing, China.
- Jiangsu Key Laboratory of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, China.
- Department of General Surgery, The Affiliated BenQ Hospital of Nanjing Medical University, Nanjing, China.
| |
Collapse
|
16
|
Maran JJ, Mugisho OO. NLRP3 inflammasome plays a vital role in the pathogenesis of age-related diseases in the eye and brain. Neural Regen Res 2024; 19:1425-1426. [PMID: 38051879 PMCID: PMC10883518 DOI: 10.4103/1673-5374.387991] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Accepted: 10/02/2023] [Indexed: 12/07/2023] Open
Affiliation(s)
- Jack Jonathan Maran
- Buchanan Ocular Therapeutics Unit, Department of Ophthalmology, New Zealand National Eye Centre, University of Auckland, Auckland, New Zealand
| | | |
Collapse
|
17
|
Haque I, Thapa P, Burns DM, Zhou J, Sharma M, Sharma R, Singh V. NLRP3 Inflammasome Inhibitors for Antiepileptogenic Drug Discovery and Development. Int J Mol Sci 2024; 25:6078. [PMID: 38892264 PMCID: PMC11172514 DOI: 10.3390/ijms25116078] [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: 05/07/2024] [Revised: 05/28/2024] [Accepted: 05/29/2024] [Indexed: 06/21/2024] Open
Abstract
Epilepsy is one of the most prevalent and serious brain disorders and affects over 70 million people globally. Antiseizure medications (ASMs) relieve symptoms and prevent the occurrence of future seizures in epileptic patients but have a limited effect on epileptogenesis. Addressing the multifaceted nature of epileptogenesis and its association with the Nod-like receptor family pyrin domain containing 3 (NLRP3) inflammasome-mediated neuroinflammation requires a comprehensive understanding of the underlying mechanisms of these medications for the development of targeted therapeutic strategies beyond conventional antiseizure treatments. Several types of NLRP3 inhibitors have been developed and their effect has been validated both in in vitro and in vivo models of epileptogenesis. In this review, we discuss the advances in understanding the regulatory mechanisms of NLRP3 activation as well as progress made, and challenges faced in the development of NLRP3 inhibitors for the treatment of epilepsy.
Collapse
Affiliation(s)
- Inamul Haque
- Research and Development Service, Kansas City Veterans Affairs Medical Center, Kansas City, MO 64128, USA; (P.T.); (D.M.B.); (M.S.); (R.S.)
- Department of Math, Science and Business Technology, Kansas City Kansas Community College, Kansas City, KS 66112, USA
- Department of Cancer Biology, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Pritam Thapa
- Research and Development Service, Kansas City Veterans Affairs Medical Center, Kansas City, MO 64128, USA; (P.T.); (D.M.B.); (M.S.); (R.S.)
- Drug Discovery Program, Midwest Veterans’ Biomedical Research Foundation, KCVA Medical Center, Kansas City, MO 64128, USA
| | - Douglas M. Burns
- Research and Development Service, Kansas City Veterans Affairs Medical Center, Kansas City, MO 64128, USA; (P.T.); (D.M.B.); (M.S.); (R.S.)
| | - Jianping Zhou
- Renal Research Laboratory, Kansas City VA Medical Center, Kansas City, MO 64128, USA;
| | - Mukut Sharma
- Research and Development Service, Kansas City Veterans Affairs Medical Center, Kansas City, MO 64128, USA; (P.T.); (D.M.B.); (M.S.); (R.S.)
- Drug Discovery Program, Midwest Veterans’ Biomedical Research Foundation, KCVA Medical Center, Kansas City, MO 64128, USA
- Renal Research Laboratory, Kansas City VA Medical Center, Kansas City, MO 64128, USA;
| | - Ram Sharma
- Research and Development Service, Kansas City Veterans Affairs Medical Center, Kansas City, MO 64128, USA; (P.T.); (D.M.B.); (M.S.); (R.S.)
| | - Vikas Singh
- Research and Development Service, Kansas City Veterans Affairs Medical Center, Kansas City, MO 64128, USA; (P.T.); (D.M.B.); (M.S.); (R.S.)
- Drug Discovery Program, Midwest Veterans’ Biomedical Research Foundation, KCVA Medical Center, Kansas City, MO 64128, USA
- Division of Neurology, Kansas City VA Medical Center, Kansas City, MO 64128, USA
| |
Collapse
|
18
|
Ye T, Wang C, Yan J, Qin Z, Qin W, Ma Y, Wan Q, Lu W, Zhang M, Tay FR, Jiao K, Niu L. Lysosomal destabilization: A missing link between pathological calcification and osteoarthritis. Bioact Mater 2024; 34:37-50. [PMID: 38173842 PMCID: PMC10761323 DOI: 10.1016/j.bioactmat.2023.12.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Revised: 11/10/2023] [Accepted: 12/01/2023] [Indexed: 01/05/2024] Open
Abstract
Calcification of cartilage by hydroxyapatite is a hallmark of osteoarthritis and its deposition strongly correlates with the severity of osteoarthritis. However, no effective strategies are available to date on the prevention of hydroxyapatite deposition within the osteoarthritic cartilage and its role in the pathogenesis of this degenerative condition is still controversial. Therefore, the present work aims at uncovering the pathogenic mechanism of intra-cartilaginous hydroxyapatite in osteoarthritis and developing feasible strategies to counter its detrimental effects. With the use of in vitro and in vivo models of osteoarthritis, hydroxyapatite crystallites deposited in the cartilage are found to be phagocytized by resident chondrocytes and processed by the lysosomes of those cells. This results in lysosomal membrane permeabilization (LMP) and release of cathepsin B (CTSB) into the cytosol. The cytosolic CTSB, in turn, activates NOD-like receptor protein-3 (NLRP3) inflammasomes and subsequently instigates chondrocyte pyroptosis. Inhibition of LMP and CTSB in vivo are effective in managing the progression of osteoarthritis. The present work provides a conceptual therapeutic solution for the prevention of osteoarthritis via alleviation of lysosomal destabilization.
Collapse
Affiliation(s)
- Tao Ye
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration & National Clinical Research Center for Oral Diseases & Shaanxi Key Laboratory of Stomatology, Department of Prosthodontics, School of Stomatology, The Fourth Military Medical University, Xi'an, Shaanxi, 710032, PR China
| | - Chenyu Wang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration & National Clinical Research Center for Oral Diseases & Shaanxi Key Laboratory of Stomatology, Department of Prosthodontics, School of Stomatology, The Fourth Military Medical University, Xi'an, Shaanxi, 710032, PR China
| | - Jianfei Yan
- Department of Stomatology, Tangdu Hospital, State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration & National Clinical Research Center for Oral Diseases & Shaanxi Key Laboratory of Stomatology, School of Stomatology, The Fourth Military Medical University, Xi'an, Shaanxi, 710032, PR China
| | - Zixuan Qin
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration & National Clinical Research Center for Oral Diseases & Shaanxi Key Laboratory of Stomatology, Department of Prosthodontics, School of Stomatology, The Fourth Military Medical University, Xi'an, Shaanxi, 710032, PR China
| | - Wenpin Qin
- Department of Stomatology, Tangdu Hospital, State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration & National Clinical Research Center for Oral Diseases & Shaanxi Key Laboratory of Stomatology, School of Stomatology, The Fourth Military Medical University, Xi'an, Shaanxi, 710032, PR China
| | - Yuxuan Ma
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration & National Clinical Research Center for Oral Diseases & Shaanxi Key Laboratory of Stomatology, Department of Prosthodontics, School of Stomatology, The Fourth Military Medical University, Xi'an, Shaanxi, 710032, PR China
| | - Qianqian Wan
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration & National Clinical Research Center for Oral Diseases & Shaanxi Key Laboratory of Stomatology, Department of Prosthodontics, School of Stomatology, The Fourth Military Medical University, Xi'an, Shaanxi, 710032, PR China
| | - Weicheng Lu
- Department of Stomatology, Tangdu Hospital, State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration & National Clinical Research Center for Oral Diseases & Shaanxi Key Laboratory of Stomatology, School of Stomatology, The Fourth Military Medical University, Xi'an, Shaanxi, 710032, PR China
| | - Mian Zhang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration & National Clinical Research Center for Oral Diseases & Shaanxi Key Laboratory of Stomatology, Department of Prosthodontics, School of Stomatology, The Fourth Military Medical University, Xi'an, Shaanxi, 710032, PR China
| | - Franklin R. Tay
- The Dental College of Georgia, Augusta University, Augusta, GA, 30912, USA
| | - Kai Jiao
- Department of Stomatology, Tangdu Hospital, State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration & National Clinical Research Center for Oral Diseases & Shaanxi Key Laboratory of Stomatology, School of Stomatology, The Fourth Military Medical University, Xi'an, Shaanxi, 710032, PR China
| | - Lina Niu
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration & National Clinical Research Center for Oral Diseases & Shaanxi Key Laboratory of Stomatology, Department of Prosthodontics, School of Stomatology, The Fourth Military Medical University, Xi'an, Shaanxi, 710032, PR China
| |
Collapse
|
19
|
Chen IC, Chen WL, Chang KH, Lee JW, Lin TH, Lin W, Chen CM, Lee-Chen GJ. Investigating the therapeutic effects of novel compounds targeting inflammatory IL-1β and IL-6 signaling pathways in spinocerebellar ataxia type 3. Eur J Pharmacol 2024; 967:176370. [PMID: 38320719 DOI: 10.1016/j.ejphar.2024.176370] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Revised: 01/17/2024] [Accepted: 01/29/2024] [Indexed: 02/13/2024]
Abstract
At least seven dominantly inherited spinocerebellar ataxias (SCA) are caused by expansions of polyglutamine (polyQ)-encoding CAG repeat. The misfolded and aggregated polyQ-expanded proteins increase reactive oxygen species (ROS), cellular toxicity, and neuroinflammation in the disease pathogenesis. In this study, we evaluated the anti-inflammatory potentials of coumarin derivatives LM-021, LMDS-1, LMDS-2, and pharmacological chaperone tafamidis using mouse BV-2 microglia and SCA3 ataxin-3 (ATXN3)/Q75-GFP SH-SY5Y cells. The four tested compounds displayed anti-inflammatory activity by suppressing nitric oxide (NO), interleukin (IL)-1β, IL-6, and tumor necrosis factor (TNF)-α production, and CD68 antigen (CD68) and histocompatibility-2 (MHCII) expression in lipopolysaccharides (LPS)/interferon (IFN)-γ-stimulated BV-2 microglia. In retinoic acid-differentiated ATXN3/Q75-GFP-expressing SH-SY5Y cells inflamed with LPS/IFN-γ-primed BV-2 conditioned medium, treatment with test compounds mitigated the increased caspase 1 activity and lactate dehydrogenase release, reduced ROS and ATXN3/Q75 aggregation, and promoted neurite outgrowth. Examination of IL-1β and IL-6-mediated signaling pathways revealed that LM-021, LMDS-1, LMDS-2, and tafamidis decreased NLR family pyrin domain containing 1 (NLRP1), c-Jun N-terminal kinase/c-Jun proto-oncogene (JNK/JUN), inhibitor of kappa B (IκBα)/P65, mitogen-activated protein kinase 14/signal transducer and activator of transcription 1 (P38/STAT1), and/or Janus kinase 2/signal transducer and activator of transcription 3 (JAK2/STAT3) signaling. The study results suggest the potential of LM-021, LMDS-1, LMDS-2, and tafamidis in treating SCA3 and probable other polyQ diseases.
Collapse
Affiliation(s)
- I-Cheng Chen
- High-value Biomaterials Research and Commercialization Center, National Taipei University of Technology, Taipei, 10608, Taiwan; Department of Chemical Engineering and Biotechnology, National Taipei University of Technology, Taipei, 10608, Taiwan
| | - Wan-Ling Chen
- Department of Neurology, Chang-Gung Memorial Hospital, Chang-Gung University College of Medicine, Taoyuan, 33302, Taiwan
| | - Kuo-Hsuan Chang
- Department of Neurology, Chang-Gung Memorial Hospital, Chang-Gung University College of Medicine, Taoyuan, 33302, Taiwan
| | - Jun-Wei Lee
- Department of Neurology, Chang-Gung Memorial Hospital, Chang-Gung University College of Medicine, Taoyuan, 33302, Taiwan
| | - Te-Hsien Lin
- Department of Neurology, Chang-Gung Memorial Hospital, Chang-Gung University College of Medicine, Taoyuan, 33302, Taiwan
| | - Wenwei Lin
- Department of Chemistry, National Taiwan Normal University, Taipei, 11677, Taiwan
| | - Chiung-Mei Chen
- Department of Neurology, Chang-Gung Memorial Hospital, Chang-Gung University College of Medicine, Taoyuan, 33302, Taiwan.
| | - Guey-Jen Lee-Chen
- Department of Life Science, National Taiwan Normal University, Taipei, 11677, Taiwan.
| |
Collapse
|
20
|
Drew PD. Targeting the Nlrp3 inflammasome as potential treatment for ethanol-induced non-communicable diseases. THE AMERICAN JOURNAL OF DRUG AND ALCOHOL ABUSE 2024; 50:125-127. [PMID: 38563505 DOI: 10.1080/00952990.2024.2332951] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Accepted: 03/16/2024] [Indexed: 04/04/2024]
Affiliation(s)
- Paul D Drew
- Departments of Neurobiology and Developmental Sciences and Neurology, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| |
Collapse
|
21
|
Kim MY, Jeong B, Lee GS, Jeon H, Yang YM, Yang H, Han YH. Panaxydol extracted from Panax ginseng inhibits NLRP3 inflammasome activation to ameliorate NASH-induced liver injury. Int Immunopharmacol 2024; 128:111565. [PMID: 38262161 DOI: 10.1016/j.intimp.2024.111565] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2023] [Revised: 01/15/2024] [Accepted: 01/16/2024] [Indexed: 01/25/2024]
Abstract
Activation of NOD-like receptor protein 3 (NLRP3) inflammasome exacerbates liver inflammation and fibrosis in nonalcoholic steatohepatitis (NASH), suggesting that development of inflammasome inhibitor can become leading candidate to ameliorate NASH. Panax ginseng (P. ginseng) contains numerous bioactive natural components to reduce inflammation. This study aims to identify inhibitory components of P. ginseng for NLRP3 inflammasome activation. We separated polar and non-polar fractions of P. ginseng and tested modulation of NLRP3 inflammasome, and then identified pure component for inflammasome inhibitor which ameliorates diet-induced NASH. Non-polar P. ginseng fractions obtained from ethyl acetate solvent attenuated IL-1β secretion and expression of active caspase-1. We revealed that panaxydol (PND) is pure component to inhibit NLRP3 inflammasome activation. PND blocked inflammasome cytokines release, pyroptotic cell death, caspase-1 activation and specking of inflammasome complex. Inhibitory effect of PND was specific to NLRP3-dependent pathway via potential interaction with ATP binding motif of NLRP3. Moreover, in vivo studies showed that PND plays beneficial roles to reduce tissue inflammations through disruption of NLRP3 inflammasome and to ameliorate the development of NASH. These results provide new insight of natural products, panaxydol, for NLRP3 inflammasome inhibitor and could offer potential therapeutic candidate for reliving NASH.
Collapse
Affiliation(s)
- Mi-Yeon Kim
- Laboratory of Pathology and Physiology, College of Pharmacy, Kangwon National University, Chuncheon 24341, South Korea
| | - Birang Jeong
- Laboratory of Natural Products Chemistry, College of Pharmacy, Kangwon National University, Chuncheon 24341, South Korea
| | - Geun-Shik Lee
- College of Veterinary Medicine and Institute of Veterinary Science, Kangwon National University, Chuncheon 24341, South Korea
| | - Hongjun Jeon
- Therapeutics & Biotechnology Division, Korea Research Institute of Chemical Technology, Daejeon 34114, South Korea
| | - Yoon Mee Yang
- Multidimensional Genomics Research Center, Kangwon National University, Chuncheon 24341, South Korea; College of Pharmacy, Kangwon National University, Chuncheon 24341, South Korea
| | - Heejung Yang
- Laboratory of Natural Products Chemistry, College of Pharmacy, Kangwon National University, Chuncheon 24341, South Korea.
| | - Yong-Hyun Han
- Laboratory of Pathology and Physiology, College of Pharmacy, Kangwon National University, Chuncheon 24341, South Korea; Multidimensional Genomics Research Center, Kangwon National University, Chuncheon 24341, South Korea.
| |
Collapse
|
22
|
Teske KA, Corona C, Wilkinson J, Mamott D, Good DA, Zambrano D, Lazar DF, Cali JJ, Robers MB, O'Brien MA. Interrogating direct NLRP3 engagement and functional inflammasome inhibition using cellular assays. Cell Chem Biol 2024; 31:349-360.e6. [PMID: 37858335 DOI: 10.1016/j.chembiol.2023.09.016] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Revised: 07/20/2023] [Accepted: 09/25/2023] [Indexed: 10/21/2023]
Abstract
As a key regulator of the innate immune system, the NLRP3 inflammasome responds to a variety of environmental insults through activation of caspase-1 and release of the proinflammatory cytokines IL-1β and IL-18. Aberrant NLRP3 inflammasome function is implicated in numerous inflammatory diseases, spurring drug discovery efforts at NLRP3 as a therapeutic target. A diverse array of small molecules is undergoing preclinical/clinical evaluation with a reported mode of action involving direct modulation of the NLRP3 pathway. However, for a subset of these ligands the functional link between live-cell target engagement and pathway inhibition has yet to be fully established. Herein we present a cohort of mechanistic assays to both query direct NLRP3 engagement in cells, and functionally interrogate different nodes of NLRP3 pathway activity. This system enabled the stratification of potency for five confirmed NLRP3 inhibitors, and identification of two reported NLRP3 inhibitors that failed to demonstrate direct pathway antagonism.
Collapse
Affiliation(s)
- Kelly A Teske
- Promega Corporation, Research & Development, Madison, WI 53711, USA
| | - Cesear Corona
- Promega Corporation, Research & Development, San Luis Obispo, CA 93401, USA
| | | | - Daniel Mamott
- Promega Corporation, Research & Development, Madison, WI 53711, USA
| | - David A Good
- Promega Corporation, Research & Development, San Luis Obispo, CA 93401, USA
| | - Delia Zambrano
- Promega Corporation, Research & Development, San Luis Obispo, CA 93401, USA
| | - Dan F Lazar
- Promega Corporation, Research & Development, Madison, WI 53711, USA
| | - James J Cali
- Promega Corporation, Research & Development, Madison, WI 53711, USA
| | - Matthew B Robers
- Promega Corporation, Research & Development, Madison, WI 53711, USA.
| | - Martha A O'Brien
- Promega Corporation, Research & Development, Madison, WI 53711, USA.
| |
Collapse
|
23
|
Shen G, Lou C, Li Q, Zhao B, Luo Y, Wu F, Jiao D, Fang M, Geng Y. Edaravone dexborneol alleviates cerebral ischemia-reperfusion injury through NF-κB/NLRP3 signal pathway. Anat Rec (Hoboken) 2024; 307:372-384. [PMID: 37475155 DOI: 10.1002/ar.25296] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Revised: 06/25/2023] [Accepted: 07/08/2023] [Indexed: 07/22/2023]
Abstract
Inflammatory injury following ischemia-reperfusion (I/R) severely limits the efficacy of stroke treatment. Edaravone dexborneol (C.EDA) has been shown to reduce inflammation following a cerebral hemorrhage. However, the precise anti-inflammatory mechanism of C.EDA is unknown. In this study, we investigated whether C.EDA provides neuroprotection after I/R in rats, as well as the potential mechanisms involved. A middle cerebral artery occlusion/reperfusion (I/R) model was created using Sprague-Dawley rats. The blood flow of the central cerebral artery was monitored by a laser speckle imaging system. The neurological score was used to assess behavioral improvement. Cerebral infarction volume was measured by TTC staining. And the integrity of the blood-brain barrier was detected by Evan's blue staining. The expression of the nuclear factor kappa-B (NF-κB)/ the NOD-like receptor protein (NLRP3) inflammasome signal pathway and microglia polarization were detected by immunofluorescence and Western blotting. The cerebral blood flow ratio indicates that the cerebral I/R model was successfully established. After reperfusion for 72 h, the improvement of neurological scores, infarct volume reduction, and integrity of the blood-brain barrier was observed in I/R rats with C.EDA treatment. Meanwhile, the immunofluorescence result showed that the expression of iNOS, NLRP3, and NF-κB protein was decreased and the level of Arg1 was increased. Western blot analysis showed that the expression of NF-κB/NLRP3 signal pathway-related protein was decreased. In conclusion, this study indicates that C.EDA alleviates I/R injury by blocking the activation of the NLRP3 inflammasome and regulating the polarization of M1/M2 microglia via the NF-κB signal pathway.
Collapse
Affiliation(s)
- Guanghong Shen
- School of Basic Medical Sciences and Forensic Medicine, Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Chengjian Lou
- Department of Neurosurgery, Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu, China
| | - Qunfeng Li
- Department of Medicine, QuZhou College of Technology, Quzhou, Zhejiang, China
| | - Bingxin Zhao
- School of Basic Medical Sciences and Forensic Medicine, Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Yuhuan Luo
- Department of Pediatric, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Fei Wu
- Institute of System Medicine, Zhejiang University School of Medicine, Hangzhou, China
| | - Dian Jiao
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou, Zhejiang, China
| | - Marong Fang
- Institute of System Medicine, Zhejiang University School of Medicine, Hangzhou, China
- Children's Hospital of Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
| | - Yu Geng
- Center for Rehabilitation Medicine, Department of Neurology, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, China
| |
Collapse
|
24
|
Bonam SR, Mastrippolito D, Georgel P, Muller S. Pharmacological targets at the lysosomal autophagy-NLRP3 inflammasome crossroads. Trends Pharmacol Sci 2024; 45:81-101. [PMID: 38102020 DOI: 10.1016/j.tips.2023.11.005] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Revised: 11/14/2023] [Accepted: 11/14/2023] [Indexed: 12/17/2023]
Abstract
Many aspects of cell homeostasis and integrity are maintained by the nucleotide-binding oligomerization domain (NOD)-like receptor (NLR) family pyrin domain-containing 3 (NLRP3) inflammasome. The NLRP3 oligomeric protein complex assembles in response to exogenous and endogenous danger signals. This inflammasome has also been implicated in the pathogenesis of a range of disease conditions, particularly chronic inflammatory diseases. Given that NLRP3 modulates autophagy, which is also a key regulator of inflammasome activity, excessive inflammation may be controlled by targeting this intersecting pathway. However, specific niche areas of NLRP3-autophagy interactions and their reciprocal regulatory mechanisms remain underexplored. Consequently, we lack treatment methods specifically targeting this pivotal axis. Here, we discuss the potential of such strategies in the context of autoimmune and metabolic diseases and propose some research avenues.
Collapse
Affiliation(s)
- Srinivasa Reddy Bonam
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Dylan Mastrippolito
- CNRS-University of Strasbourg, Biotechnology and Cell Signaling, Illkirch, France; Strasbourg Institute of Drug Discovery and Development (IMS), Strasbourg, France
| | - Philippe Georgel
- CNRS-University of Strasbourg, Biotechnology and Cell Signaling, Illkirch, France; Strasbourg Institute of Drug Discovery and Development (IMS), Strasbourg, France; Fédération Hospitalo-Universitaire (FHU) OMICARE, Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg University, Strasbourg, France
| | - Sylviane Muller
- CNRS-University of Strasbourg, Biotechnology and Cell Signaling, Illkirch, France; Strasbourg Institute of Drug Discovery and Development (IMS), Strasbourg, France; Fédération Hospitalo-Universitaire (FHU) OMICARE, Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg University, Strasbourg, France; University of Strasbourg Institute for Advanced Study (USIAS), Strasbourg, France.
| |
Collapse
|
25
|
Palumbo L, Carinci M, Guarino A, Asth L, Zucchini S, Missiroli S, Rimessi A, Pinton P, Giorgi C. The NLRP3 Inflammasome in Neurodegenerative Disorders: Insights from Epileptic Models. Biomedicines 2023; 11:2825. [PMID: 37893198 PMCID: PMC10604217 DOI: 10.3390/biomedicines11102825] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Revised: 10/12/2023] [Accepted: 10/16/2023] [Indexed: 10/29/2023] Open
Abstract
Neuroinflammation represents a dynamic process of defense and protection against the harmful action of infectious agents or other detrimental stimuli in the central nervous system (CNS). However, the uncontrolled regulation of this physiological process is strongly associated with serious dysfunctional neuronal issues linked to the progression of CNS disorders. Moreover, it has been widely demonstrated that neuroinflammation is linked to epilepsy, one of the most prevalent and serious brain disorders worldwide. Indeed, NLRP3, one of the most well-studied inflammasomes, is involved in the generation of epileptic seizures, events that characterize this pathological condition. In this context, several pieces of evidence have shown that the NLRP3 inflammasome plays a central role in the pathophysiology of mesial temporal lobe epilepsy (mTLE). Based on an extensive review of the literature on the role of NLRP3-dependent inflammation in epilepsy, in this review we discuss our current understanding of the connection between NLRP3 inflammasome activation and progressive neurodegeneration in epilepsy. The goal of the review is to cover as many of the various known epilepsy models as possible, providing a broad overview of the current literature. Lastly, we also propose some of the present therapeutic strategies targeting NLRP3, aiming to provide potential insights for future studies.
Collapse
Affiliation(s)
- Laura Palumbo
- Department of Medical Sciences, Section of Experimental Medicine, University of Ferrara, 44121 Ferrara, Italy; (L.P.); (M.C.); (S.M.); (A.R.); (P.P.)
| | - Marianna Carinci
- Department of Medical Sciences, Section of Experimental Medicine, University of Ferrara, 44121 Ferrara, Italy; (L.P.); (M.C.); (S.M.); (A.R.); (P.P.)
| | - Annunziata Guarino
- Department of Neuroscience and Rehabilitation, University of Ferrara, Via Fossato di Mortara 70, 44121 Ferrara, Italy; (A.G.); (L.A.); (S.Z.)
| | - Laila Asth
- Department of Neuroscience and Rehabilitation, University of Ferrara, Via Fossato di Mortara 70, 44121 Ferrara, Italy; (A.G.); (L.A.); (S.Z.)
| | - Silvia Zucchini
- Department of Neuroscience and Rehabilitation, University of Ferrara, Via Fossato di Mortara 70, 44121 Ferrara, Italy; (A.G.); (L.A.); (S.Z.)
- Laboratory of Technologies for Advanced Therapy (LTTA), Technopole of Ferrara, 44121 Ferrara, Italy
| | - Sonia Missiroli
- Department of Medical Sciences, Section of Experimental Medicine, University of Ferrara, 44121 Ferrara, Italy; (L.P.); (M.C.); (S.M.); (A.R.); (P.P.)
- Laboratory of Technologies for Advanced Therapy (LTTA), Technopole of Ferrara, 44121 Ferrara, Italy
| | - Alessandro Rimessi
- Department of Medical Sciences, Section of Experimental Medicine, University of Ferrara, 44121 Ferrara, Italy; (L.P.); (M.C.); (S.M.); (A.R.); (P.P.)
- Laboratory of Technologies for Advanced Therapy (LTTA), Technopole of Ferrara, 44121 Ferrara, Italy
- Center of Research for Innovative Therapies in Cystic Fibrosis, University of Ferrara, 44121 Ferrara, Italy
| | - Paolo Pinton
- Department of Medical Sciences, Section of Experimental Medicine, University of Ferrara, 44121 Ferrara, Italy; (L.P.); (M.C.); (S.M.); (A.R.); (P.P.)
- Laboratory of Technologies for Advanced Therapy (LTTA), Technopole of Ferrara, 44121 Ferrara, Italy
- Center of Research for Innovative Therapies in Cystic Fibrosis, University of Ferrara, 44121 Ferrara, Italy
| | - Carlotta Giorgi
- Department of Medical Sciences, Section of Experimental Medicine, University of Ferrara, 44121 Ferrara, Italy; (L.P.); (M.C.); (S.M.); (A.R.); (P.P.)
- Laboratory of Technologies for Advanced Therapy (LTTA), Technopole of Ferrara, 44121 Ferrara, Italy
| |
Collapse
|
26
|
Li N, Jiang X, Zhang R, Ye N, Tang M, Cai X, Su K, Peng J, Zhang X, Zhao M, Wu W, Ye H. Discovery of Triazinone Derivatives as Novel, Specific, and Direct NLRP3 Inflammasome Inhibitors for the Treatment of DSS-Induced Ulcerative Colitis. J Med Chem 2023; 66:13428-13451. [PMID: 37756547 DOI: 10.1021/acs.jmedchem.3c00696] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/29/2023]
Abstract
NLRP3 is an intracellular sensor protein that causes inflammasome formation and pyroptosis in response to a wide range of stimuli. Aberrant activation of NLRP3 inflammasome has been implicated in various chronic inflammatory diseases, making it a promising target for therapeutic intervention. In this work, a series of novel triazinone inhibitors of NLRP3 inflammasome were designed and synthesized. Compound L38 was identified for its excellent activity and acceptable metabolic stability among 41 compounds. Additionally, mechanism studies indicated that L38 inhibited NLRP3 inflammasome activation and pyroptosis by suppressing gasdermin D cleavage, ASC oligomerization, and NLRP3 inflammasome assembly while leaving mitochondrial ROS production, lysosome damage, and chloride/potassium efflux unaffected. Further investigation revealed that L38 could bind to the NACHT domain to exert inflammatory properties. Importantly, L38 exhibited positive therapeutic effects in DSS-induced ulcerative colitis mouse model. Taken together, this study presents a promising inhibitor of NLRP3 inflammasome deserving further investigation.
Collapse
Affiliation(s)
- Na Li
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, 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
| | - Ruijia Zhang
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Neng Ye
- 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
| | - Xiaoying Cai
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Kaiyue Su
- 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
| | - Xinlu Zhang
- 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, Frontiers Science Center for Disease-Related Molecular Network, 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
|
27
|
Woo S, Gandhi S, Ghincea A, Saber T, Lee CJ, Ryu C. Targeting the NLRP3 inflammasome and associated cytokines in scleroderma associated interstitial lung disease. Front Cell Dev Biol 2023; 11:1254904. [PMID: 37849737 PMCID: PMC10577231 DOI: 10.3389/fcell.2023.1254904] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Accepted: 09/19/2023] [Indexed: 10/19/2023] Open
Abstract
SSc-ILD (scleroderma associated interstitial lung disease) is a complex rheumatic disease characterized in part by immune dysregulation leading to the progressive fibrotic replacement of normal lung architecture. Because improved treatment options are sorely needed, additional study of the fibroproliferative mechanisms mediating this disease has the potential to accelerate development of novel therapies. The contribution of innate immunity is an emerging area of investigation in SSc-ILD as recent work has demonstrated the mechanistic and clinical significance of the NLRP3 inflammasome and its associated cytokines of TNFα (tumor necrosis factor alpha), IL-1β (interleukin-1 beta), and IL-18 in this disease. In this review, we will highlight novel pathophysiologic insights afforded by these studies and the potential of leveraging this complex biology for clinical benefit.
Collapse
Affiliation(s)
| | | | | | | | | | - Changwan Ryu
- Department of Internal Medicine, Yale School of Medicine, Section of Pulmonary, Critical Care and Sleep Medicine, New Haven, CT, United States
| |
Collapse
|
28
|
Chen SY, Li YP, You YP, Zhang HR, Shi ZJ, Liang QQ, Yuan T, Xu R, Xu LH, Zha QB, Ou-Yang DY, He XH. Theaflavin mitigates acute gouty peritonitis and septic organ injury in mice by suppressing NLRP3 inflammasome assembly. Acta Pharmacol Sin 2023; 44:2019-2036. [PMID: 37221235 PMCID: PMC10545837 DOI: 10.1038/s41401-023-01105-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Accepted: 05/03/2023] [Indexed: 05/25/2023]
Abstract
Activation of NLR family pyrin domain-containing 3 (NLRP3) inflammasome plays important role in defending against infections, but its aberrant activation is causally linked to many inflammatory diseases, thus being a therapeutic target for these diseases. Theaflavin, one major ingredient of black tea, exhibits potent anti-inflammatory and anti-oxidative activities. In this study, we investigated the therapeutic effects of theaflavin against NLRP3 inflammasome activation in macrophages in vitro and in animal models of related diseases. We showed that theaflavin (50, 100, 200 μM) dose-dependently inhibited NLRP3 inflammasome activation in LPS-primed macrophages stimulated with ATP, nigericin or monosodium urate crystals (MSU), evidenced by reduced release of caspase-1p10 and mature interleukin-1β (IL-1β). Theaflavin treatment also inhibited pyroptosis as shown by decreased generation of N-terminal fragment of gasdermin D (GSDMD-NT) and propidium iodide incorporation. Consistent with these, theaflavin treatment suppressed ASC speck formation and oligomerization in macrophages stimulated with ATP or nigericin, suggesting reduced inflammasome assembly. We revealed that theaflavin-induced inhibition on NLRP3 inflammasome assembly and pyroptosis resulted from ameliorated mitochondrial dysfunction and reduced mitochondrial ROS production, thereby suppressing interaction between NLRP3 and NEK7 downstream of ROS. Moreover, we showed that oral administration of theaflavin significantly attenuated MSU-induced mouse peritonitis and improved the survival of mice with bacterial sepsis. Consistently, theaflavin administration significantly reduced serum levels of inflammatory cytokines including IL-1β and attenuated liver inflammation and renal injury of mice with sepsis, concomitant with reduced generation of caspase-1p10 and GSDMD-NT in the liver and kidney. Together, we demonstrate that theaflavin suppresses NLRP3 inflammasome activation and pyroptosis by protecting mitochondrial function, thus mitigating acute gouty peritonitis and bacterial sepsis in mice, highlighting a potential application in treating NLRP3 inflammasome-related diseases.
Collapse
Affiliation(s)
- Si-Yuan Chen
- Department of Immunobiology, College of Life Science and Technology, Jinan University, Guangzhou, 510632, China
| | - Ya-Ping Li
- Department of Immunobiology, College of Life Science and Technology, Jinan University, Guangzhou, 510632, China
| | - Yi-Ping You
- Department of Immunobiology, College of Life Science and Technology, Jinan University, Guangzhou, 510632, China
| | - Hong-Rui Zhang
- Department of Immunobiology, College of Life Science and Technology, Jinan University, Guangzhou, 510632, China
| | - Zi-Jian Shi
- Department of Fetal Medicine, the First Affiliated Hospital of Jinan University, Guangzhou, 510630, China
| | - Qi-Qi Liang
- Department of Immunobiology, College of Life Science and Technology, Jinan University, Guangzhou, 510632, China
| | - Tao Yuan
- Department of Immunobiology, College of Life Science and Technology, Jinan University, Guangzhou, 510632, China
| | - Rong Xu
- Department of Immunobiology, College of Life Science and Technology, Jinan University, Guangzhou, 510632, China
| | - Li-Hui Xu
- Department of Cell Biology, College of Life Science and Technology, Jinan University, Guangzhou, 510632, China
| | - Qing-Bing Zha
- Department of Fetal Medicine, the First Affiliated Hospital of Jinan University, Guangzhou, 510630, China.
- Department of Clinical Laboratory, the Fifth Affiliated Hospital of Jinan University, Heyuan, 517000, China.
| | - Dong-Yun Ou-Yang
- Department of Immunobiology, College of Life Science and Technology, Jinan University, Guangzhou, 510632, China.
| | - Xian-Hui He
- Department of Immunobiology, College of Life Science and Technology, Jinan University, Guangzhou, 510632, China.
- Department of Clinical Laboratory, the Fifth Affiliated Hospital of Jinan University, Heyuan, 517000, China.
| |
Collapse
|
29
|
Mészáros Á, Molnár K, Fazakas C, Nógrádi B, Lüvi A, Dudás T, Tiszlavicz L, Farkas AE, Krizbai IA, Wilhelm I. Inflammasome activation in peritumoral astrocytes is a key player in breast cancer brain metastasis development. Acta Neuropathol Commun 2023; 11:155. [PMID: 37749707 PMCID: PMC10521486 DOI: 10.1186/s40478-023-01646-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Accepted: 08/27/2023] [Indexed: 09/27/2023] Open
Abstract
Inflammasomes, primarily responsible for the activation of IL-1β, have emerged as critical regulators of the tumor microenvironment. By using in vivo and in vitro brain metastasis models, as well as human samples to study the role of the NLRP3 inflammasome in triple-negative breast cancer (TNBC) brain metastases, we found NLRP3 inflammasome components and IL-1β to be highly and specifically expressed in peritumoral astrocytes. Soluble factors from TNBC cells induced upregulation and activation of NLRP3 and IL-1β in astrocytes, while astrocyte-derived mediators augmented the proliferation of metastatic cells. In addition, inhibition of NLRP3 inflammasome activity using MCC950 or dampening the downstream effect of IL-1β prevented the proliferation increase in cancer cells. In vivo, MCC950 reduced IL-1β expression in peritumoral astrocytes, as well as the levels of inflammasome components and active IL-1β. Most importantly, significantly retarded growth of brain metastatic tumors was observed in mice treated with MCC950. Overall, astrocytes contribute to TNBC progression in the brain through activation of the NLRP3 inflammasome and consequent IL-1β release. We conclude that pharmacological targeting of inflammasomes may become a novel strategy in controlling brain metastatic diseases.
Collapse
Affiliation(s)
- Ádám Mészáros
- Institute of Biophysics, Biological Research Centre, ELKH (Eötvös Loránd Research Network), Temesvári Krt. 62, 6726, Szeged, Hungary
- Doctoral School of Biology, University of Szeged, Szeged, Hungary
| | - Kinga Molnár
- Institute of Biophysics, Biological Research Centre, ELKH (Eötvös Loránd Research Network), Temesvári Krt. 62, 6726, Szeged, Hungary
| | - Csilla Fazakas
- Institute of Biophysics, Biological Research Centre, ELKH (Eötvös Loránd Research Network), Temesvári Krt. 62, 6726, Szeged, Hungary
| | - Bernát Nógrádi
- Institute of Biophysics, Biological Research Centre, ELKH (Eötvös Loránd Research Network), Temesvári Krt. 62, 6726, Szeged, Hungary
- Department of Neurology, University of Szeged, Szeged, Hungary
| | - Adél Lüvi
- Institute of Biophysics, Biological Research Centre, ELKH (Eötvös Loránd Research Network), Temesvári Krt. 62, 6726, Szeged, Hungary
| | - Tamás Dudás
- Institute of Biophysics, Biological Research Centre, ELKH (Eötvös Loránd Research Network), Temesvári Krt. 62, 6726, Szeged, Hungary
- Theoretical Medicine Doctoral School, University of Szeged, Szeged, Hungary
| | | | - Attila Elek Farkas
- Institute of Biophysics, Biological Research Centre, ELKH (Eötvös Loránd Research Network), Temesvári Krt. 62, 6726, Szeged, Hungary
| | - István Adorján Krizbai
- Institute of Biophysics, Biological Research Centre, ELKH (Eötvös Loránd Research Network), Temesvári Krt. 62, 6726, Szeged, Hungary.
- Institute of Life Sciences, Vasile Goldiş Western University of Arad, Arad, Romania.
| | - Imola Wilhelm
- Institute of Biophysics, Biological Research Centre, ELKH (Eötvös Loránd Research Network), Temesvári Krt. 62, 6726, Szeged, Hungary.
- Institute of Life Sciences, Vasile Goldiş Western University of Arad, Arad, Romania.
| |
Collapse
|
30
|
Gastaldi S, Rocca C, Gianquinto E, Granieri MC, Boscaro V, Blua F, Rolando B, Marini E, Gallicchio M, De Bartolo A, Romeo N, Mazza R, Fedele F, Pagliaro P, Penna C, Spyrakis F, Bertinaria M, Angelone T. Discovery of a novel 1,3,4-oxadiazol-2-one-based NLRP3 inhibitor as a pharmacological agent to mitigate cardiac and metabolic complications in an experimental model of diet-induced metaflammation. Eur J Med Chem 2023; 257:115542. [PMID: 37290185 DOI: 10.1016/j.ejmech.2023.115542] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Revised: 05/30/2023] [Accepted: 06/01/2023] [Indexed: 06/10/2023]
Abstract
Inspired by the recent advancements in understanding the binding mode of sulfonylurea-based NLRP3 inhibitors to the NLRP3 sensor protein, we developed new NLRP3 inhibitors by replacing the central sulfonylurea moiety with different heterocycles. Computational studies evidenced that some of the designed compounds were able to maintain important interaction within the NACHT domain of the target protein similarly to the most active sulfonylurea-based NLRP3 inhibitors. Among the studied compounds, the 1,3,4-oxadiazol-2-one derivative 5 (INF200) showed the most promising results being able to prevent NLRP3-dependent pyroptosis triggered by LPS/ATP and LPS/MSU by 66.3 ± 6.6% and 61.6 ± 11.5% and to reduce IL-1β release (35.5 ± 8.8% μM) at 10 μM in human macrophages. The selected compound INF200 (20 mg/kg/day) was then tested in an in vivo rat model of high-fat diet (HFD)-induced metaflammation to evaluate its beneficial cardiometabolic effects. INF200 significantly counteracted HFD-dependent "anthropometric" changes, improved glucose and lipid profiles, and attenuated systemic inflammation and biomarkers of cardiac dysfunction (particularly BNP). Hemodynamic evaluation on Langendorff model indicate that INF200 limited myocardial damage-dependent ischemia/reperfusion injury (IRI) by improving post-ischemic systolic recovery and attenuating cardiac contracture, infarct size, and LDH release, thus reversing the exacerbation of obesity-associated damage. Mechanistically, in post-ischemic hearts, IFN200 reduced IRI-dependent NLRP3 activation, inflammation, and oxidative stress. These results highlight the potential of the novel NLRP3 inhibitor, INF200, and its ability to reverse the unfavorable cardio-metabolic dysfunction associated with obesity.
Collapse
Affiliation(s)
- Simone Gastaldi
- Department of Drug Science and Technology, University of Turin, 10125, Turin, Italy
| | - Carmine Rocca
- Cellular and Molecular Cardiovascular Pathophysiology Laboratory, Department of Biology, E. and E.S. (DiBEST), University of Calabria, 87036, Rende, Italy
| | - Eleonora Gianquinto
- Department of Drug Science and Technology, University of Turin, 10125, Turin, Italy
| | - Maria Concetta Granieri
- Cellular and Molecular Cardiovascular Pathophysiology Laboratory, Department of Biology, E. and E.S. (DiBEST), University of Calabria, 87036, Rende, Italy
| | - Valentina Boscaro
- Department of Drug Science and Technology, University of Turin, 10125, Turin, Italy
| | - Federica Blua
- Department of Drug Science and Technology, University of Turin, 10125, Turin, Italy
| | - Barbara Rolando
- Department of Drug Science and Technology, University of Turin, 10125, Turin, Italy
| | - Elisabetta Marini
- Department of Drug Science and Technology, University of Turin, 10125, Turin, Italy
| | | | - Anna De Bartolo
- Cellular and Molecular Cardiovascular Pathophysiology Laboratory, Department of Biology, E. and E.S. (DiBEST), University of Calabria, 87036, Rende, Italy
| | - Naomi Romeo
- Cellular and Molecular Cardiovascular Pathophysiology Laboratory, Department of Biology, E. and E.S. (DiBEST), University of Calabria, 87036, Rende, Italy
| | - Rosa Mazza
- Cellular and Molecular Cardiovascular Pathophysiology Laboratory, Department of Biology, E. and E.S. (DiBEST), University of Calabria, 87036, Rende, Italy
| | - Francesco Fedele
- Department of Clinical, Internal, Anesthesiology and Cardiovascular Sciences, Sapienza University of Rome, Viale del Policlinico, 155, 00161, Rome, Italy; National Institute for Cardiovascular Research (INRC), Bologna, Italy
| | - Pasquale Pagliaro
- Department of Clinical and Biological Sciences, University of Turin, Turin, Italy; National Institute for Cardiovascular Research (INRC), Bologna, Italy
| | - Claudia Penna
- Department of Clinical and Biological Sciences, University of Turin, Turin, Italy; National Institute for Cardiovascular Research (INRC), Bologna, Italy.
| | - Francesca Spyrakis
- Department of Drug Science and Technology, University of Turin, 10125, Turin, Italy.
| | - Massimo Bertinaria
- Department of Drug Science and Technology, University of Turin, 10125, Turin, Italy.
| | - Tommaso Angelone
- Cellular and Molecular Cardiovascular Pathophysiology Laboratory, Department of Biology, E. and E.S. (DiBEST), University of Calabria, 87036, Rende, Italy; National Institute for Cardiovascular Research (INRC), Bologna, Italy
| |
Collapse
|
31
|
Chen L, Yuan L, Yang J, Pan Y, Wang H. Identification of key immune-related genes associated with LPS/D-GalN-induced acute liver failure in mice based on transcriptome sequencing. PeerJ 2023; 11:e15241. [PMID: 37168540 PMCID: PMC10166078 DOI: 10.7717/peerj.15241] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Accepted: 03/28/2023] [Indexed: 05/13/2023] Open
Abstract
BACKGROUND The aim of this study was to identify key immune-related genes in acute liver failure (ALF) by constructing an ALF mouse model for transcriptome sequencing. METHODS The C57BL/6 mouse with ALF model was induced by lipopolysaccharide (LPS)/ D-galactosamine (D-GalN). After successful modelling, the liver tissues of all mice were obtained for transcriptome sequencing. The key immune-related genes in mice with ALF were identified by differential expression analysis, immune infiltration analysis, weighted gene co-expression network analysis (WGCNA), enrichment analysis, and protein-protein interaction (PPI) analysis. RESULTS An LPS/D-GalN-induced ALF mouse model was successfully constructed, and transcriptome sequencing was performed. Significant differences in the proportions of monocytes, macrophages M0, macrophages M1 and neutrophils were shown by immune infiltration analysis, and 5255 genes highly associated with these four immune cells were identified by WGCNA. These immune genes were found to be significantly enriched in the TNF signalling pathway by enrichment analysis. Finally, PPI analysis was performed on genes enriched in this pathway and three key genes (CXCL1, CXCL10 and IL1B) were screened out and revealed to be significantly upregulated in ALF. CONCLUSIONS Key immune-related genes in ALF were identified in this study, which may provide not only potential therapeutic targets for treating ALF and improving its prognosis, but also a reliable scientific basis for the immunotherapy of the disease.
Collapse
Affiliation(s)
- Ling Chen
- Department of Infectious Disease, Zhejiang Hospital, Hangzhou, China
| | - Li Yuan
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Jingle Yang
- Department of Infectious Disease, Zhejiang Hospital, Hangzhou, China
| | - Yizhi Pan
- Department of Infectious Disease, Zhejiang Hospital, Hangzhou, China
| | - Hong Wang
- Department of Infectious Disease, Zhejiang Hospital, Hangzhou, China
| |
Collapse
|
32
|
Chen C, Smith MT. The NLRP3 inflammasome: role in the pathobiology of chronic pain. Inflammopharmacology 2023:10.1007/s10787-023-01235-8. [PMID: 37106238 DOI: 10.1007/s10787-023-01235-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Accepted: 04/01/2023] [Indexed: 04/29/2023]
Abstract
Chronic pain is not only one of the most common health problems, it is often challenging to treat adequately. Chronic pain has a high prevalence globally, affecting approximately 20% of the adult population. Chronic inflammatory pain and neuropathic (nerve) pain conditions are areas of large unmet medical need because analgesic/adjuvant agents recommended for alleviation of these types of chronic pain often lack efficacy and/or they produce dose-limiting side effects. Recent work has implicated the NLRP3 (NOD-, LRR- and pyrin domain-containing protein 3) inflammasome in the pathobiology of chronic pain, especially neuropathic and inflammatory pain conditions. NLRP3 is activated by damage-associated molecular patterns (DAMPs) and pathogen-associated molecular patterns (PAMPs). This in turn leads to recruitment and activation of caspase-1 an enzyme that cleaves the inactive IL-1β and IL-18 precursors to their respective mature pro-inflammatory cytokines (IL-1β and IL-18) for release into the cellular milieu. Caspase-1 also cleaves the pyroptosis-inducing factor, gasdermin D, that leads to oligomerization of its N-terminal fragment to form pores in the host cell membrane. This then results in cellular swelling, lysis and release of cytoplasmic contents in an inflammatory form of cell death, termed pyroptosis. The ultimate outcome may lead to the development of neuropathic pain and/or chronic inflammatory pain. In this review, we address a role for NLRP3 inflammasome activation in the pathogenesis of various chronic pain conditions.
Collapse
Affiliation(s)
- Chen Chen
- Faculty of Science, School of Chemistry and Molecular Biosciences and School of Biomedical Sciences, Faculty of Medicine, St Lucia Campus, The University of Queensland, Brisbane, Australia
- School of Biomedical Sciences, Faculty of Medicine, St Lucia Campus, The University of Queensland, Brisbane, QLD, 4072, Australia
| | - Maree T Smith
- School of Biomedical Sciences, Faculty of Medicine, St Lucia Campus, The University of Queensland, Brisbane, QLD, 4072, Australia.
| |
Collapse
|
33
|
Magagnoli J, Narendran S, Pereira F, Cummings TH, Hardin JW, Sutton SS, Ambati J. Association between Fluoxetine Use and Overall Survival among Patients with Cancer Treated with PD-1/L1 Immunotherapy. Pharmaceuticals (Basel) 2023; 16:ph16050640. [PMID: 37242422 DOI: 10.3390/ph16050640] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Revised: 04/13/2023] [Accepted: 04/17/2023] [Indexed: 05/28/2023] Open
Abstract
Checkpoint inhibitors can be a highly effective antitumor therapy but only to a subset of patients, presumably due to immunotherapy resistance. Fluoxetine was recently revealed to inhibit the NLRP3 inflammasome, and NLRP3 inhibition could serve as a target for immunotherapy resistance. Therefore, we evaluated the overall survival (OS) in patients with cancer receiving checkpoint inhibitors combined with fluoxetine. A cohort study was conducted among patients diagnosed with lung, throat (pharynx or larynx), skin, or kidney/urinary cancer treated with checkpoint inhibitor therapy. Utilizing the Veterans Affairs Informatics and Computing Infrastructure, patients were retrospectively evaluated during the period from October 2015 to June 2021. The primary outcome was overall survival (OS). Patients were followed until death or the end of the study period. There were 2316 patients evaluated, including 34 patients who were exposed to checkpoint inhibitors and fluoxetine. Propensity score weighted Cox proportional hazards demonstrated a better OS in fluoxetine-exposed patients than unexposed (HR: 0.59, 95% CI 0.371-0.936). This cohort study among cancer patients treated with checkpoint inhibitor therapy showed a significant improvement in the OS when fluoxetine was used. Because of this study's potential for selection bias, randomized trials are needed to assess the efficacy of the association of fluoxetine or another anti-NLRP3 drug to checkpoint inhibitor therapy.
Collapse
Affiliation(s)
- Joseph Magagnoli
- Dorn Research Institute, Columbia VA Health Care System, Columbia, SC 29209, USA
- Department of Clinical Pharmacy and Outcomes Sciences, College of Pharmacy, University of South Carolina, Columbia, SC 29208, USA
| | - Siddharth Narendran
- Center for Advanced Vision Science, University of Virginia School of Medicine, Charlottesville, VA 22903, USA
- Department of Ophthalmology, University of Virginia School of Medicine, Charlottesville, VA 22903, USA
| | - Felipe Pereira
- Center for Advanced Vision Science, University of Virginia School of Medicine, Charlottesville, VA 22903, USA
- Department of Ophthalmology, University of Virginia School of Medicine, Charlottesville, VA 22903, USA
| | - Tammy H Cummings
- Dorn Research Institute, Columbia VA Health Care System, Columbia, SC 29209, USA
- Department of Clinical Pharmacy and Outcomes Sciences, College of Pharmacy, University of South Carolina, Columbia, SC 29208, USA
| | - James W Hardin
- Dorn Research Institute, Columbia VA Health Care System, Columbia, SC 29209, USA
- Department of Epidemiology & Biostatistics, University of South Carolina, Columbia, SC 29208, USA
| | - S Scott Sutton
- Dorn Research Institute, Columbia VA Health Care System, Columbia, SC 29209, USA
- Department of Clinical Pharmacy and Outcomes Sciences, College of Pharmacy, University of South Carolina, Columbia, SC 29208, USA
| | - Jayakrishna Ambati
- Center for Advanced Vision Science, University of Virginia School of Medicine, Charlottesville, VA 22903, USA
- Department of Ophthalmology, University of Virginia School of Medicine, Charlottesville, VA 22903, USA
- Department of Pathology, University of Virginia School of Medicine, Charlottesville, VA 22903, USA
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia School of Medicine, Charlottesville, VA 22903, USA
| |
Collapse
|
34
|
Thapa P, Upadhyay SP, Singh V, Boinpelly VC, Zhou J, Johnson DK, Gurung P, Lee ES, Sharma R, Sharma M. Chalcone: A potential scaffold for NLRP3 inflammasome inhibitors. EUROPEAN JOURNAL OF MEDICINAL CHEMISTRY REPORTS 2023; 7:100100. [PMID: 37033416 PMCID: PMC10081147 DOI: 10.1016/j.ejmcr.2022.100100] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Overactivated NLRP3 inflammasome has been shown to associate with an increasing number of disease conditions. Activation of the NLRP3 inflammasome results in caspase-1-catalyzed formation of active pro-inflammatory cytokines (IL-1β and IL-18) resulting in pyroptosis. The multi-protein composition of the NLRP3 inflammasome and its sensitivity to several damage-associated molecular patterns (DAMPs) and pathogen-associated molecular patterns (PAMPs) make this extensively studied inflammasome an attractive target to treat chronic conditions. However, none of the known NLRP3 inhibitors has been approved for clinical use. Sulfonylurea and covalent inhibitors with electrophilic warhead (Michael acceptor) are among the prominent classes of compounds explored for their NLRP3 inhibitory effects. Chalcone, a small molecule with α, β unsaturated carbonyl group (Michael acceptor), has also been studied as a promising scaffold for the development of NLRP3 inhibitors. Low molecular weight, easy to manipulate lipophilicity and cost-effectiveness have attracted many to use chalcone scaffold for drug development. In this review, we highlight chalcone derivatives with NLRP3 inflammasome inhibitory activities. Recent developments and potential new directions summarized here will, hopefully, serve as valuable perspectives for investigators including medicinal chemists and drug discovery researchers to utilize chalcone as a scaffold for developing novel NLRP3 inflammasome inhibitors.
Collapse
Affiliation(s)
- Pritam Thapa
- Drug Discovery Program, Midwest Veterans’ Biomedical Research Foundation, KCVA Medical Center, Kansas City, MO, 64128, USA
| | - Sunil P. Upadhyay
- Drug Discovery Program, Midwest Veterans’ Biomedical Research Foundation, KCVA Medical Center, Kansas City, MO, 64128, USA
| | - Vikas Singh
- Division of Neurology, KCVA Medical Center, Kansas City, MO, USA
| | - Varun C. Boinpelly
- Renal Research Laboratory, Kansas City VA Medical Center, Kansas City, MO, USA
| | - Jianping Zhou
- Renal Research Laboratory, Kansas City VA Medical Center, Kansas City, MO, USA
| | - David K. Johnson
- Department of Computational Chemical Biology Core, Molecular Graphics and Modeling Core, University of Kansas, KS, 66047, USA
| | - Prajwal Gurung
- Inflammation Program, University of Iowa, Iowa City, IA, 52242, USA
| | - Eung Seok Lee
- College of Pharmacy, Yeungnam University, Gyeongsan, 712-749, Republic of Korea
| | - Ram Sharma
- Drug Discovery Program, Midwest Veterans’ Biomedical Research Foundation, KCVA Medical Center, Kansas City, MO, 64128, USA
| | - Mukut Sharma
- Drug Discovery Program, Midwest Veterans’ Biomedical Research Foundation, KCVA Medical Center, Kansas City, MO, 64128, USA
- Renal Research Laboratory, Kansas City VA Medical Center, Kansas City, MO, USA
| |
Collapse
|
35
|
Mohan J, Ghazi T, Mazibuko MS, Chuturgoon AA. Antiretrovirals Promote Insulin Resistance in HepG2 Liver Cells through miRNA Regulation and Transcriptional Activation of the NLRP3 Inflammasome. Int J Mol Sci 2023; 24:ijms24076267. [PMID: 37047241 PMCID: PMC10094183 DOI: 10.3390/ijms24076267] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Revised: 03/23/2023] [Accepted: 03/24/2023] [Indexed: 03/29/2023] Open
Abstract
Metabolic syndrome (MetS) is a non-communicable disease characterized by a cluster of metabolic irregularities. Alarmingly, the prevalence of MetS in people living with Human Immunodeficiency Virus (HIV) and antiretroviral (ARV) usage is increasing rapidly. Insulin resistance is a common characteristic of MetS that leads to the development of Type 2 diabetes mellitus (T2DM). The progression of insulin resistance is strongly linked to inflammasome activation. This study aimed to draw links between the combinational use of Tenofovir disoproxil fumarate (TDF), Lamivudine (3TC), and Dolutegravir (DTG), and inflammasome activation and subsequent promotion of insulin resistance following a 120 h treatment period in HepG2 liver in vitro cell model. Furthermore, we assess microRNA (miR-128a) expression as a negative regulator of the IRS1/AKT signaling pathway. The relative expression of phosphorylated IRS1 was determined by Western blot. Transcript levels of NLRP3, IL-1β, JNK, IRS1, AKT, PI3K, and miR-128a were assessed using quantitative PCR (qPCR). Caspase-1 activity was measured using luminometry. Following exposure to ARVs for 120 h, NLRP3 mRNA expression (p = 0.0500) and caspase-1 activity (p < 0.0001) significantly increased. This was followed by a significant elevation in IL-1β in mRNA expression (p = 0.0015). Additionally, JNK expression (p = 0.0093) was upregulated with coinciding increases in p-IRS1 protein expression (p < 0.0001) and decreased IRS1 mRNA expression (p = 0.0004). Consequently, decreased AKT (p = 0.0005) and PI3K expressions (p = 0.0007) were observed. Interestingly miR-128a expression was significantly upregulated. The results indicate that combinational use of ARVs upregulates inflammasome activation and promotes insulin resistance through dysregulation of the IRS1/PI3K/AKT insulin signaling pathway.
Collapse
|
36
|
Rennie C, Huang Y, Siwakoti P, Du Z, Padula M, Bao G, Tuch BE, Xu X, McClements L. In vitro evaluation of a hybrid drug delivery nanosystem for fibrosis prevention in cell therapy for Type 1 diabetes. Nanomedicine (Lond) 2023; 18:53-66. [PMID: 36938861 DOI: 10.2217/nnm-2022-0231] [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: 03/21/2023] Open
Abstract
Background: Implantation of insulin-secreting cells has been trialed as a treatment for Type 1 diabetes mellitus; however, the host immunogenic response limits their effectiveness. Methodology: The authors developed a core-shell nanostructure of upconversion nanoparticle-mesoporous silica for controlled local delivery of an immunomodulatory agent, MCC950, using near-infrared light and validated it in in vitro models of fibrosis. Results: The individual components of the nanosystem did not affect the proliferation of insulin-secreting cells, unlike fibroblast proliferation (p < 0.01). The nanosystem is effective at releasing MCC950 and preventing fibroblast differentiation (p < 0.01), inflammation (IL-6 expression; p < 0.05) and monocyte adhesion (p < 0.01). Conclusion: This MCC950-loaded nanomedicine system could be used in the future together with insulin-secreting cell implants to increase their longevity as a curative treatment for Type 1 diabetes mellitus.
Collapse
Affiliation(s)
- Claire Rennie
- School of Life Sciences, Faculty of Science, University of Technology Sydney, Ultimo, NSW, 2007, Australia
| | - Yanan Huang
- School of Mathematical and Physical Sciences, Faculty of Science, University of Technology Sydney, Ultimo, NSW, 2007, Australia
| | - Prakriti Siwakoti
- School of Life Sciences, Faculty of Science, University of Technology Sydney, Ultimo, NSW, 2007, Australia
| | - Ziqing Du
- School of Mathematical and Physical Sciences, Faculty of Science, University of Technology Sydney, Ultimo, NSW, 2007, Australia
| | - Matthew Padula
- School of Life Sciences, Faculty of Science, University of Technology Sydney, Ultimo, NSW, 2007, Australia
| | - Guochen Bao
- Institute for Biomedical Materials & Devices, Faculty of Science, University of Technology Sydney, Ultimo, NSW, 2007, Australia
| | - Bernard E Tuch
- Department of Diabetes, Central Clinical School, Faculty of Medicine, Nursing & Health Sciences, Monash University, Victoria, 3004, Australia.,Australian Foundation for Diabetes Research, 2000, NSW, Australia
| | - Xiaoxue Xu
- School of Mathematical and Physical Sciences, Faculty of Science, University of Technology Sydney, Ultimo, NSW, 2007, Australia.,School of Biomedical Engineering, Faculty of Engineering and IT, University of Technology Sydney, Ultimo, NSW, 2007, Australia
| | - Lana McClements
- School of Life Sciences, Faculty of Science, University of Technology Sydney, Ultimo, NSW, 2007, Australia.,Institute for Biomedical Materials & Devices, Faculty of Science, University of Technology Sydney, Ultimo, NSW, 2007, Australia
| |
Collapse
|
37
|
Enosi Tuipulotu D, Feng S, Pandey A, Zhao A, Ngo C, Mathur A, Lee J, Shen C, Fox D, Xue Y, Kay C, Kirkby M, Lo Pilato J, Kaakoush NO, Webb D, Rug M, Robertson AAB, Tessema MB, Pang S, Degrandi D, Pfeffer K, Augustyniak D, Blumenthal A, Miosge LA, Brüstle A, Yamamoto M, Reading PC, Burgio G, Man SM. Immunity against Moraxella catarrhalis requires guanylate-binding proteins and caspase-11-NLRP3 inflammasomes. EMBO J 2023; 42:e112558. [PMID: 36762431 PMCID: PMC10015372 DOI: 10.15252/embj.2022112558] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2022] [Revised: 01/17/2023] [Accepted: 01/19/2023] [Indexed: 02/11/2023] Open
Abstract
Moraxella catarrhalis is an important human respiratory pathogen and a major causative agent of otitis media and chronic obstructive pulmonary disease. Toll-like receptors contribute to, but cannot fully account for, the complexity of the immune response seen in M. catarrhalis infection. Using primary mouse bone marrow-derived macrophages to examine the host response to M. catarrhalis infection, our global transcriptomic and targeted cytokine analyses revealed activation of immune signalling pathways by both membrane-bound and cytosolic pattern-recognition receptors. We show that M. catarrhalis and its outer membrane vesicles or lipooligosaccharide (LOS) can activate the cytosolic innate immune sensor caspase-4/11, gasdermin-D-dependent pyroptosis, and the NLRP3 inflammasome in human and mouse macrophages. This pathway is initiated by type I interferon signalling and guanylate-binding proteins (GBPs). We also show that inflammasomes and GBPs, particularly GBP2, are required for the host defence against M. catarrhalis in mice. Overall, our results reveal an essential role for the interferon-inflammasome axis in cytosolic recognition and immunity against M. catarrhalis, providing new molecular targets that may be used to mitigate pathological inflammation triggered by this pathogen.
Collapse
Affiliation(s)
- Daniel Enosi Tuipulotu
- Division of Immunology and Infectious Disease, The John Curtin School of Medical ResearchThe Australian National UniversityCanberraACTAustralia
| | - Shouya Feng
- Division of Immunology and Infectious Disease, The John Curtin School of Medical ResearchThe Australian National UniversityCanberraACTAustralia
| | - Abhimanu Pandey
- Division of Immunology and Infectious Disease, The John Curtin School of Medical ResearchThe Australian National UniversityCanberraACTAustralia
| | - Anyang Zhao
- Division of Immunology and Infectious Disease, The John Curtin School of Medical ResearchThe Australian National UniversityCanberraACTAustralia
| | - Chinh Ngo
- Division of Immunology and Infectious Disease, The John Curtin School of Medical ResearchThe Australian National UniversityCanberraACTAustralia
| | - Anukriti Mathur
- Division of Immunology and Infectious Disease, The John Curtin School of Medical ResearchThe Australian National UniversityCanberraACTAustralia
| | - Jiwon Lee
- Centre for Advanced MicroscopyThe Australian National UniversityCanberraACTAustralia
| | - Cheng Shen
- Division of Immunology and Infectious Disease, The John Curtin School of Medical ResearchThe Australian National UniversityCanberraACTAustralia
| | - Daniel Fox
- Division of Immunology and Infectious Disease, The John Curtin School of Medical ResearchThe Australian National UniversityCanberraACTAustralia
| | - Yansong Xue
- Division of Immunology and Infectious Disease, The John Curtin School of Medical ResearchThe Australian National UniversityCanberraACTAustralia
| | - Callum Kay
- Division of Immunology and Infectious Disease, The John Curtin School of Medical ResearchThe Australian National UniversityCanberraACTAustralia
| | - Max Kirkby
- Division of Immunology and Infectious Disease, The John Curtin School of Medical ResearchThe Australian National UniversityCanberraACTAustralia
| | - Jordan Lo Pilato
- Division of Immunology and Infectious Disease, The John Curtin School of Medical ResearchThe Australian National UniversityCanberraACTAustralia
| | | | - Daryl Webb
- Centre for Advanced MicroscopyThe Australian National UniversityCanberraACTAustralia
| | - Melanie Rug
- Centre for Advanced MicroscopyThe Australian National UniversityCanberraACTAustralia
| | - Avril AB Robertson
- School of Chemistry and Molecular BiosciencesThe University of QueenslandBrisbaneQLDAustralia
| | - Melkamu B Tessema
- Department of Microbiology and ImmunologyThe University of Melbourne, The Peter Doherty Institute for Infection and ImmunityMelbourneVICAustralia
| | - Stanley Pang
- Antimicrobial Resistance and Infectious Diseases (AMRID) Research LaboratoryMurdoch UniversityMurdochWAAustralia
- Department of Microbiology, PathWest Laboratory Medicine‐WAFiona Stanley HospitalMurdochWAAustralia
| | - Daniel Degrandi
- Institute of Medical Microbiology and Hospital HygieneHeinrich‐Heine‐University DüsseldorfDüsseldorfGermany
| | - Klaus Pfeffer
- Institute of Medical Microbiology and Hospital HygieneHeinrich‐Heine‐University DüsseldorfDüsseldorfGermany
| | - Daria Augustyniak
- Department of Pathogen Biology and Immunology, Faculty of Biological SciencesUniversity of WroclawWroclawPoland
| | - Antje Blumenthal
- Frazer InstituteThe University of QueenslandQLDBrisbaneAustralia
| | - Lisa A Miosge
- Division of Immunology and Infectious Disease, The John Curtin School of Medical ResearchThe Australian National UniversityCanberraACTAustralia
| | - Anne Brüstle
- Division of Immunology and Infectious Disease, The John Curtin School of Medical ResearchThe Australian National UniversityCanberraACTAustralia
| | - Masahiro Yamamoto
- Department of Immunoparasitology, Research Institute for Microbial DiseasesOsaka UniversityOsakaJapan
- Laboratory of Immunoparasitology, WPI Immunology Frontier Research CenterOsaka UniversityOsakaJapan
| | - Patrick C Reading
- Department of Microbiology and ImmunologyThe University of Melbourne, The Peter Doherty Institute for Infection and ImmunityMelbourneVICAustralia
- WHO Collaborating Centre for Reference and Research on InfluenzaVictorian Infectious Diseases Reference Laboratory, The Peter Doherty Institute for Infection and ImmunityMelbourneVICAustralia
| | - Gaetan Burgio
- Division of Immunology and Infectious Disease, The John Curtin School of Medical ResearchThe Australian National UniversityCanberraACTAustralia
| | - Si Ming Man
- Division of Immunology and Infectious Disease, The John Curtin School of Medical ResearchThe Australian National UniversityCanberraACTAustralia
| |
Collapse
|
38
|
Gregory GE, Munro KJ, Couper KN, Pathmanaban ON, Brough D. The NLRP3 inflammasome as a target for sensorineural hearing loss. Clin Immunol 2023; 249:109287. [PMID: 36907540 DOI: 10.1016/j.clim.2023.109287] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Revised: 02/08/2023] [Accepted: 02/24/2023] [Indexed: 03/14/2023]
Abstract
Sensorineural hearing loss is the most common type of hearing loss in adults and occurs due to damage of the inner ear caused by a range of factors including ageing, excessive noise, toxins, and cancer. Auto-inflammatory disease is also a cause of hearing loss and there is evidence that inflammation could contribute to hearing loss in other conditions. Within the inner ear there are resident macrophage cells that respond to insults and whose activation correlates with damage. The NLRP3 inflammasome is a multi-molecular pro-inflammatory protein complex that forms in activated macrophages and may contribute to hearing loss. The aim of this article is to discuss the evidence for the NLRP3 inflammasome and associated cytokines as potential therapeutic targets for sensorineural hearing loss in conditions ranging from auto-inflammatory disease to tumour-induced hearing loss in vestibular schwannoma.
Collapse
Affiliation(s)
- Grace E Gregory
- Division of Neuroscience, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, Manchester, UK; Geoffrey Jefferson Brain Research Centre, The Manchester Academic Health Science Centre, Northern Care Alliance NHS Group, University of Manchester, Manchester, UK; The Lydia Becker Institute of Immunology and Inflammation, University of Manchester, Manchester, UK
| | - Kevin J Munro
- Manchester Centre for Audiology and Deafness, School of Health Sciences, University of Manchester, Manchester, UK; Manchester University Hospitals NHS Foundation Trust, Manchester Academic Health Science Centre, Manchester, UK
| | - Kevin N Couper
- Geoffrey Jefferson Brain Research Centre, The Manchester Academic Health Science Centre, Northern Care Alliance NHS Group, University of Manchester, Manchester, UK; The Lydia Becker Institute of Immunology and Inflammation, University of Manchester, Manchester, UK
| | - Omar N Pathmanaban
- Division of Neuroscience, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, Manchester, UK; Geoffrey Jefferson Brain Research Centre, The Manchester Academic Health Science Centre, Northern Care Alliance NHS Group, University of Manchester, Manchester, UK; The Lydia Becker Institute of Immunology and Inflammation, University of Manchester, Manchester, UK; Department of Neurosurgery, Manchester Centre for Clinical Neurosciences, Salford Royal Hospital NHS Foundation Trust, Salford, UK.
| | - David Brough
- Division of Neuroscience, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, Manchester, UK; Geoffrey Jefferson Brain Research Centre, The Manchester Academic Health Science Centre, Northern Care Alliance NHS Group, University of Manchester, Manchester, UK; The Lydia Becker Institute of Immunology and Inflammation, University of Manchester, Manchester, UK.
| |
Collapse
|
39
|
Dobrev D, Heijman J, Hiram R, Li N, Nattel S. Inflammatory signalling in atrial cardiomyocytes: a novel unifying principle in atrial fibrillation pathophysiology. Nat Rev Cardiol 2023; 20:145-167. [PMID: 36109633 PMCID: PMC9477170 DOI: 10.1038/s41569-022-00759-w] [Citation(s) in RCA: 122] [Impact Index Per Article: 61.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 07/26/2022] [Indexed: 02/08/2023]
Abstract
Inflammation has been implicated in atrial fibrillation (AF), a very common and clinically significant cardiac rhythm disturbance, but its precise role remains poorly understood. Work performed over the past 5 years suggests that atrial cardiomyocytes have inflammatory signalling machinery - in particular, components of the NLRP3 (NACHT-, LRR- and pyrin domain-containing 3) inflammasome - that is activated in animal models and patients with AF. Furthermore, work in animal models suggests that NLRP3 inflammasome activation in atrial cardiomyocytes might be a sufficient and necessary condition for AF occurrence. In this Review, we evaluate the evidence for the role and pathophysiological significance of cardiomyocyte NLRP3 signalling in AF. We first summarize the evidence for a role of inflammation in AF and review the biochemical properties of the NLRP3 inflammasome, as defined primarily in studies of classic inflammation. We then briefly consider the broader evidence for a role of inflammatory signalling in heart disease, particularly conditions that predispose individuals to develop AF. We provide a detailed discussion of the available information about atrial cardiomyocyte NLRP3 inflammasome signalling in AF and related conditions and evaluate the possibility that similar signalling might be important in non-myocyte cardiac cells. We then review the evidence on the role of active resolution of inflammation and its potential importance in suppressing AF-related inflammatory signalling. Finally, we consider the therapeutic potential and broader implications of this new knowledge and highlight crucial questions to be addressed in future research.
Collapse
Affiliation(s)
- Dobromir Dobrev
- Institute of Pharmacology, West German Heart and Vascular Center, University Duisburg-Essen, Duisburg, Germany
- Department of Medicine and Research Center, Montreal Heart Institute and Université de Montréal, Montréal, Canada
- Department of Molecular Physiology & Biophysics, Baylor College of Medicine, Houston, TX, USA
| | - Jordi Heijman
- Department of Cardiology, Cardiovascular Research Institute Maastricht, Faculty of Health, Medicine, and Life Sciences, Maastricht University, Maastricht, Netherlands
| | - Roddy Hiram
- Department of Medicine and Research Center, Montreal Heart Institute and Université de Montréal, Montréal, Canada
| | - Na Li
- Department of Molecular Physiology & Biophysics, Baylor College of Medicine, Houston, TX, USA
- Department of Medicine, Section of Cardiovascular Research, Baylor College of Medicine, Houston, TX, USA
- Cardiovascular Research Institute, Baylor College of Medicine, Houston, TX, USA
| | - Stanley Nattel
- Institute of Pharmacology, West German Heart and Vascular Center, University Duisburg-Essen, Duisburg, Germany.
- Department of Medicine and Research Center, Montreal Heart Institute and Université de Montréal, Montréal, Canada.
- IHU LIRYC and Fondation Bordeaux Université, Bordeaux, France.
- Department of Pharmacology and Therapeutics, McGill University, Montreal, Quebec, Canada.
| |
Collapse
|
40
|
Charan HV, Dwivedi DK, Khan S, Jena G. Mechanisms of NLRP3 inflammasome-mediated hepatic stellate cell activation: Therapeutic potential for liver fibrosis. Genes Dis 2023; 10:480-494. [PMID: 37223529 PMCID: PMC10201559 DOI: 10.1016/j.gendis.2021.12.006] [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: 06/12/2021] [Revised: 11/09/2021] [Accepted: 12/01/2021] [Indexed: 01/18/2023] Open
Abstract
The liver injury leads to an inflammatory response, which causes the activation of hepatic stellate cells (HSCs) that further secrete ECM proteins and play an important role in liver fibrosis. Moreover, the inflammatory response is a driving force for fibrogenesis, which is triggered by many types of injuries. Exaggerated inflammatory immune responses are mediated by cytoplasmic protein complexes known as inflammasomes, which are involved in many chronic liver diseases. Inflammasomes are pattern recognition receptors (PRRs) that can sense any microbial motifs known as pathogen-associated molecular patterns (PAMPs), and host- or environmental-derived stress signals known as damage-associated molecular patterns (DAMPs). The inflammasomes cause caspase-mediated proteolytic cleavage of pro-IL-1β and pro-IL-18 into active IL-1β and IL-18. In this review, we provide a comprehensive summary of the important roles of NLRP3 inflammasome in the pathogenesis of liver fibrosis with an emphasis on several direct and indirect pathways responsible for the NLRP3 inflammasome-mediated HSCs activation and fibrogenesis. In addition, we discuss the general pharmacological and genetics strategies for the inhibition of NLRP3 inflammasome activation and its downstream signaling with examples of emerging pharmacotherapeutics, targeting the NLRP3 inflammasome signaling as well as a possible way to develop effective and safer NLRP3 inflammasome inhibitors.
Collapse
Affiliation(s)
- Harsh Vardhan Charan
- Facility for Risk Assessment and Intervention Studies, Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, Sector-67, S.A.S. Nagar, Punjab 160062, India
| | - Durgesh Kumar Dwivedi
- Facility for Risk Assessment and Intervention Studies, Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, Sector-67, S.A.S. Nagar, Punjab 160062, India
| | - Sabbir Khan
- Facility for Risk Assessment and Intervention Studies, Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, Sector-67, S.A.S. Nagar, Punjab 160062, India
- Department of Neuro-Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Gopabandhu Jena
- Facility for Risk Assessment and Intervention Studies, Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, Sector-67, S.A.S. Nagar, Punjab 160062, India
| |
Collapse
|
41
|
Abstract
PURPOSE OF REVIEW This short review is intended to highlight the potential role of inflammation as a key pathological driver, rather than a mere consequence, of nephrolithiasis. Although there is clearly a strong likelihood that the relationship is bidirectional, and that kidney stone-triggered inflammation can establish a vicious cycle of tissue injury and stone formation. RECENT FINDINGS These consist of data from both recent preclinical and clinical studies demonstrating the importance of inflammation in models of stone disease and in kidney tissue from patients with nephrolithiasis, and as a potential driver of disease recurrence and a suitable treatment target. In particular, the role of immune cells and their relationship to the NLRP3 inflammasome is becoming clearer, as well as the potential contribution to tissue injury and stone formation of the pro-inflammatory cytokines interleukin-1β and interleukin-18. SUMMARY This concept is not new and raises the possibility that targeting inflammation directly may prove to be a novel and suitable means of treatment for at least some types of kidney stone, and in certain clinical settings, both acutely and as prevention, especially in those patients experiencing recurrent stone episodes and/or who have a well defined metabolic cause such as uric acid or calcium oxalate stones.
Collapse
Affiliation(s)
- Giovanna Capolongo
- Department of Translational Medical Sciences, University of Campania "L. Vanvitelli", Naples
| | - Pietro Manuel Ferraro
- Dipartimento Universitario di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, Roma, Italia
| | - Robert Unwin
- Department of Renal Medicine, Royal Free Hospital, University College London (UCL), London, UK
| |
Collapse
|
42
|
Franco-Trepat E, Alonso-Pérez A, Guillán-Fresco M, López-Fagúndez M, Pazos-Pérez A, Crespo-Golmar A, Belén Bravo S, López-López V, Jorge-Mora A, Cerón-Carrasco JP, Lois Iglesias A, Gómez R. β Boswellic Acid Blocks Articular Innate Immune Responses: An In Silico and In Vitro Approach to Traditional Medicine. Antioxidants (Basel) 2023; 12:371. [PMID: 36829930 PMCID: PMC9952103 DOI: 10.3390/antiox12020371] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Revised: 01/25/2023] [Accepted: 01/30/2023] [Indexed: 02/08/2023] Open
Abstract
Osteoarthritis (OA) is hallmarked as a silent progressive rheumatic disease of the whole joint. The accumulation of inflammatory and catabolic factors such as IL6, TNFα, and COX2 drives the OA pathophysiology into cartilage degradation, synovia inflammation, and bone destruction. There is no clinical available OA treatment. Although traditional ayurvedic medicine has been using Boswellia serrata extracts (BSE) as an antirheumatic treatment for a millennium, none of the BSE components have been clinically approved. Recently, β boswellic acid (BBA) has been shown to reduce in vivo OA-cartilage loss through an unknown mechanism. We used computational pharmacology, proteomics, transcriptomics, and metabolomics to present solid evidence of BBA therapeutic properties in mouse and primary human OA joint cells. Specifically, BBA binds to the innate immune receptor Toll-like Receptor 4 (TLR4) complex and inhibits both TLR4 and Interleukin 1 Receptor (IL1R) signaling in OA chondrocytes, osteoblasts, and synoviocytes. Moreover, BBA inhibition of TLR4/IL1R downregulated reactive oxygen species (ROS) synthesis and MAPK p38/NFκB, NLRP3, IFNαβ, TNF, and ECM-related pathways. Altogether, we present a solid bulk of evidence that BBA blocks OA innate immune responses and could be transferred into the clinic as an alimentary supplement or as a therapeutic tool after clinical trial evaluations.
Collapse
Affiliation(s)
- Eloi Franco-Trepat
- Musculoskeletal Pathology Group, Institute IDIS, Santiago University Clinical Hospital, 15706 Santiago de Compostela, Spain
| | - Ana Alonso-Pérez
- Musculoskeletal Pathology Group, Institute IDIS, Santiago University Clinical Hospital, 15706 Santiago de Compostela, Spain
| | - María Guillán-Fresco
- Musculoskeletal Pathology Group, Institute IDIS, Santiago University Clinical Hospital, 15706 Santiago de Compostela, Spain
| | - Miriam López-Fagúndez
- Musculoskeletal Pathology Group, Institute IDIS, Santiago University Clinical Hospital, 15706 Santiago de Compostela, Spain
| | - Andrés Pazos-Pérez
- Musculoskeletal Pathology Group, Institute IDIS, Santiago University Clinical Hospital, 15706 Santiago de Compostela, Spain
| | - Antía Crespo-Golmar
- Musculoskeletal Pathology Group, Institute IDIS, Santiago University Clinical Hospital, 15706 Santiago de Compostela, Spain
| | - Susana Belén Bravo
- Musculoskeletal Pathology Group, Institute IDIS, Santiago University Clinical Hospital, 15706 Santiago de Compostela, Spain
| | - Verónica López-López
- Musculoskeletal Pathology Group, Institute IDIS, Santiago University Clinical Hospital, 15706 Santiago de Compostela, Spain
| | - Alberto Jorge-Mora
- Musculoskeletal Pathology Group, Institute IDIS, Santiago University Clinical Hospital, 15706 Santiago de Compostela, Spain
| | - José P. Cerón-Carrasco
- Centro Universitario de la Defensa, Universidad Politécnica de Cartagena, C/Coronel López Peña S/N, Base Aérea de San Javier, Santiago de La Ribera, 30720 Murcia, Spain
| | - Ana Lois Iglesias
- Musculoskeletal Pathology Group, Institute IDIS, Santiago University Clinical Hospital, 15706 Santiago de Compostela, Spain
| | - Rodolfo Gómez
- Musculoskeletal Pathology Group, Institute IDIS, Santiago University Clinical Hospital, 15706 Santiago de Compostela, Spain
| |
Collapse
|
43
|
Ioannou GN, Horn CL, Kothari V, Yeh MM, Shyu I, Lee SP, Savard CE. Genetic deletion or pharmacologic inhibition of the Nlrp3 inflammasome did not ameliorate experimental NASH. J Lipid Res 2023; 64:100330. [PMID: 36641116 PMCID: PMC9944495 DOI: 10.1016/j.jlr.2023.100330] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Revised: 11/30/2022] [Accepted: 12/09/2022] [Indexed: 01/13/2023] Open
Abstract
It has been postulated that inflammasomes, in particular the NLRP3 (NLR family pyrin domain containing 3) inflammasome, mediate the necroinflammation and fibrosis that characterize nonalcoholic steatohepatitis (NASH) by engaging innate immune responses. We aimed to investigate the impact of genetic deletion or pharmacologic inhibition of the NLRP3 inflammasome on experimental steatohepatitis. Global Nlrp3 KO (expected to inhibit the NLRP3 inflammasome) or Casp1 KO (expected to inhibit all inflammasomes) mice were compared to wild type controls after 6 months on a high-fat, high-cholesterol (HFHC, 1% cholesterol) diet known to induce fibrosing steatohepatitis. Additionally, wildtype mice on a HFHC diet (0.75% or 0.5% cholesterol) for 6 months were either treated or not treated with an oral, pharmacologic inhibitor of Nlrp3 (MCC950) that was delivered in the drinking water (0.3 mg/ml). We found that genetic deletion or pharmacologic inhibition of the NLRP3 inflammasome did not ameliorate any of the histological components of fibrosing NASH in HFHC-fed mice. Collectively, these results do not support NLRP3 inhibition as a potential target for human NASH.
Collapse
Affiliation(s)
- George N Ioannou
- Division of Gastroenterology, Department of Medicine, Veterans Affairs Puget Sound Health Care System, Seattle, WA, USA; Division of Gastroenterology, Department of Medicine, University of Washington, Seattle WA, USA; Research and Development, Veterans Affairs Puget Sound Health Care System, Seattle, WA, USA.
| | - Christian L Horn
- Division of Gastroenterology and Hepatology, Department of Medicine, San Antonio Military Medical Center, Fort Sam Houston, TX, USA
| | - Vishal Kothari
- Department of Medicine, Division of Metabolism, Endocrinology and Nutrition, UW Medicine Diabetes Institute, Seattle, WA, USA
| | - Matthew M Yeh
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, USA
| | - Irene Shyu
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, USA
| | - Sum P Lee
- Division of Gastroenterology, Department of Medicine, University of Washington, Seattle WA, USA
| | - Christopher E Savard
- Division of Gastroenterology, Department of Medicine, Veterans Affairs Puget Sound Health Care System, Seattle, WA, USA; Division of Gastroenterology, Department of Medicine, University of Washington, Seattle WA, USA; Research and Development, Veterans Affairs Puget Sound Health Care System, Seattle, WA, USA
| |
Collapse
|
44
|
NLRP3 Inflammasome in Atherosclerosis: Putting Out the Fire of Inflammation. Inflammation 2023; 46:35-46. [PMID: 35953687 DOI: 10.1007/s10753-022-01725-x] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Revised: 07/25/2022] [Accepted: 07/29/2022] [Indexed: 11/05/2022]
Abstract
Atherosclerosis (AS) is a chronic inflammatory disease with thickening or hardening of the arteries, which led to the built-up of plaques in the inner lining of an artery. Among all the clarified pathogenesis, the over-activation of inflammatory reaction is one of the most acknowledged one. The nucleotide-binding domain leucine-rich repeat (NLR) and pyrin domain containing receptor 3 (NLRP3) inflammasome, as a vital and special form of inflammation and innate immunity, has been widely revealed to participate in the onset and development of AS. This review will introduce the process of the pathogenesis and progression of AS, and will describe the biological features of the NLRP3 inflammasome. Furthermore, the role of the NLRP3 inflammasome in AS and the possible mechanisms will be discussed. In addition, several kinds of agents with the effect of anti-atherosclerotic taking advantage of the NLRP3 inflammasome intervention will be described and discussed in detail, including natural compounds (baicalin, dihydromyricetin, luteolin, 5-deoxy-rutaecarpine (R3) and Salvianolic acid A, etc.), microRNAs (microRNA-30c-5p, microRNA-9, microRNA-146a-5p, microRNA-16-5p and microRNA-181a, etc.), and autophagy regulators (melatonin, dietary PUFA and arglabin, etc.). We aim to provide novel insights in the exploration of the specific mechanisms of AS and the development of new treatments of AS.
Collapse
|
45
|
The NLRP3 Inflammasome in Age-Related Cerebral Small Vessel Disease Manifestations: Untying the Innate Immune Response Connection. LIFE (BASEL, SWITZERLAND) 2023; 13:life13010216. [PMID: 36676165 PMCID: PMC9866483 DOI: 10.3390/life13010216] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Revised: 01/07/2023] [Accepted: 01/09/2023] [Indexed: 01/14/2023]
Abstract
In this narrative review, we present the evidence on nucleotide-binding and oligomerization (NOD) domain-like receptor (NLR) family pyrin domain (PYD)-containing 3 (NLRP3) inflammasome activation for its putative roles in the elusive pathomechanism of aging-related cerebral small vessel disease (CSVD). Although NLRP3 inflammasome-interleukin (IL)-1β has been implicated in the pathophysiology of coronary artery disease, its roles in cerebral arteriothrombotic micro-circulation disease such as CSVD remains unexplored. Here, we elaborate on the current manifestations of CSVD and its' complex pathogenesis and relate the array of activators and aberrant activation involving NLRP3 inflammasome with this condition. These neuroinflammatory insights would expand on our current understanding of CSVD clinical (and subclinical) heterogenous manifestations whilst highlighting plausible NLRP3-linked therapeutic targets.
Collapse
|
46
|
Ishabiyi FO, Ogidi JO, Olukade BA, Amorha CC, El-Sharkawy LY, Okolo CC, Adeniyi TM, Atasie NH, Ibrahim A, Balogun TA. Computational Evaluation of Azadirachta indica-Derived Bioactive Compounds as Potential Inhibitors of NLRP3 in the Treatment of Alzheimer's Disease. J Alzheimers Dis 2023; 94:S67-S85. [PMID: 36683510 PMCID: PMC10473084 DOI: 10.3233/jad-221020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
BACKGROUND The development of therapeutic agents against Alzheimer's disease (AD) has stalled recently. Drug candidates targeting amyloid-β (Aβ) deposition have often failed clinical trials at different stages, prompting the search for novel targets for AD therapy. The NLRP3 inflammasome is an integral part of innate immunity, contributing to neuroinflammation and AD pathophysiology. Thus, it has become a promising new target for AD therapy. OBJECTIVE The study sought to investigate the potential of bioactive compounds derived from Azadirachta-indica to inhibit the NLRP3 protein implicated in the pathophysiology of AD. METHODS Structural bioinformatics via molecular docking and density functional theory (DFT) analysis was utilized for the identification of novel NLRP3 inhibitors from A. indica bioactive compounds. The compounds were further subjected to pharmacokinetic and drug-likeness analysis. Results obtained from the compounds were compared against that of oridonin, a known NLRP3 inhibitor. RESULTS The studied compounds optimally saturated the binding site of the NLRP3 NACHT domain, forming principal interactions with the different amino acids at its binding site. The studied compounds also demonstrated better bioactivity and chemical reactivity as ascertained by DFT analysis and all the compounds except 7-desacetyl-7-benzoylazadiradione, which had two violations, conformed to Lipinski's rule of five. CONCLUSION In silico studies show that A. indica derived compounds have better inhibitory potential against NLRP3 and better pharmacokinetic profiles when compared with the reference ligand (oridonin). These compounds are thus proposed as novel NLRP3 inhibitors for the treatment of AD. Further wet-lab studies are needed to confirm the potency of the studied compounds.
Collapse
Affiliation(s)
- Felix Oluwasegun Ishabiyi
- Faculty of Pharmacy, University of Ibadan, Ibadan, Nigeria
- Institute of Bioinformatics and Molecular Therapeutics, Oshogbo, Osun State, Nigeria
| | - James Okwudirichukwu Ogidi
- Faculty of Pharmacy, University of Nigeria, Nsukka, Enugu, Nigeria
- Institute of Bioinformatics and Molecular Therapeutics, Oshogbo, Osun State, Nigeria
| | - Baliqis Adejoke Olukade
- Physiology Department, Faculty of Basic Medical Sciences, Olabisi Onabanjo University, Sagamu Campus, Nigeria
- Institute of Bioinformatics and Molecular Therapeutics, Oshogbo, Osun State, Nigeria
| | - Chizoba Christabel Amorha
- Department of Biochemistry, Faculty of Basic Medical Sciences, University of Ibadan, Ibadan, Nigeria
- Institute of Bioinformatics and Molecular Therapeutics, Oshogbo, Osun State, Nigeria
| | - Lina Y. El-Sharkawy
- Division of Pharmacy and Optometry, School of Health Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, The University of Manchester, United Kingdom
- Institute of Bioinformatics and Molecular Therapeutics, Oshogbo, Osun State, Nigeria
| | - Chukwuemeka Calistus Okolo
- Department of Veterinary Medicine University of Nigeria, Nsukka, Nigeria
- Institute of Bioinformatics and Molecular Therapeutics, Oshogbo, Osun State, Nigeria
| | - Titilope Mary Adeniyi
- Department of Biochemistry, Adekunle Ajasin University, Akungba Akoko, Ondo State, Nigeria
- Institute of Bioinformatics and Molecular Therapeutics, Oshogbo, Osun State, Nigeria
| | - Nkechi Hope Atasie
- Nigerian Correctional Services, Enugu Custodial Center, Enugu State, Nigeria
- Institute of Bioinformatics and Molecular Therapeutics, Oshogbo, Osun State, Nigeria
| | - Abdulwasiu Ibrahim
- Department of Biochemistry, Drosophila Laboratory, Faculty of Basic Medical Sciences, University of Ibadan, Ibadan, Nigeria
- Institute of Bioinformatics and Molecular Therapeutics, Oshogbo, Osun State, Nigeria
| | | |
Collapse
|
47
|
Liu Y, Wang D, Li T, Yang F, Li Z, Bai X, Wang Y. The role of NLRP3 inflammasome in inflammation-related skeletal muscle atrophy. Front Immunol 2022; 13:1035709. [PMID: 36405697 PMCID: PMC9668849 DOI: 10.3389/fimmu.2022.1035709] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2022] [Accepted: 10/13/2022] [Indexed: 04/04/2024] Open
Abstract
Skeletal muscle atrophy is a common complication in survivors of sepsis, which affects the respiratory and motor functions of patients, thus severely impacting their quality of life and long-term survival. Although several advances have been made in investigations on the pathogenetic mechanism of sepsis-induced skeletal muscle atrophy, the underlying mechanisms remain unclear. Findings from recent studies suggest that the nucleotide-binding and oligomerisation domain (NOD)-like receptor family pyrin domain containing 3 (NLRP3) inflammasome, a regulator of inflammation, may be crucial in the development of skeletal muscle atrophy. NLRP3 inhibitors contribute to the inhibition of catabolic processes, skeletal muscle atrophy and cachexia-induced inflammation. Here, we review the mechanisms by which NLRP3 mediates these responses and analyse how NLRP3 affects muscle wasting during inflammation.
Collapse
Affiliation(s)
- Yukun Liu
- Department of Plastic and Cosmetic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Dongfang Wang
- Trauma Center/Department of Emergency and Traumatic Surgery, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Tianyu Li
- Trauma Center/Department of Emergency and Traumatic Surgery, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Fan Yang
- Trauma Center/Department of Emergency and Traumatic Surgery, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zhanfei Li
- Trauma Center/Department of Emergency and Traumatic Surgery, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiangjun Bai
- Trauma Center/Department of Emergency and Traumatic Surgery, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yuchang Wang
- Trauma Center/Department of Emergency and Traumatic Surgery, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
48
|
Oronsky B, Caroen S, Abrouk N, Reid TR. RRx-001 and the "Right stuff": Protection and treatment in outer space. LIFE SCIENCES IN SPACE RESEARCH 2022; 35:69-75. [PMID: 36336372 DOI: 10.1016/j.lssr.2022.05.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Revised: 04/26/2022] [Accepted: 05/09/2022] [Indexed: 06/16/2023]
Abstract
From antibiotics to aspirin to antimalarials and to anticancer agents, about half of the world's best-selling drugs are derived from nature. However, accelerating climatic disruption, habitat destruction, pollution, and biodiversity loss all negatively impact the potential of natural sources to continue to serve as repositories of novel pharmaceuticals. On that basis, the final frontier for drug development is perhaps not the rainforests, coral reefs, and other natural habitats but rather the aerospace industry with its virtually unlimited and inexhaustible man-made 'library' of potentially bioactive compounds. The first aerospace-sourced therapeutic to reach the clinic is RRx-001, an inhibitor of the NOD-like receptor - Nucleotide-binding oligomerization domain with Leucine rich Repeat and Pyrin domain (NLRP3) inflammasome in a Phase 3 trial for the treatment of small cell lung cancer (SCLC) and in a soon-to-start Phase 3 trial for protection against chemoradiotherapy-induced severe oral mucositis in first line head and neck cancer. As manned missions to the Moon, Mars, and asteroids as well as space tourism beckon, it is perhaps fitting that a compound like RRx-001, which is derived from 1,3,3-Trinitroazetidine (TNAZ), an explosive propellant for rockets, is a potential "all purpose" option to mitigate the major biomedical effects of space radiation exposures including cancer development and other tissue degenerations both within mission and after mission. This article highlights the promise of RRx-001 to attenuate the acute and late effects of radiation exposure on astronauts including the development of cancer.
Collapse
|
49
|
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
|
50
|
Su X, Liu B, Wang S, Wang Y, Zhang Z, Zhou H, Li F. NLRP3 inflammasome: A potential therapeutic target to minimize renal ischemia/reperfusion injury during transplantation. Transpl Immunol 2022; 75:101718. [PMID: 36126906 DOI: 10.1016/j.trim.2022.101718] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Revised: 09/09/2022] [Accepted: 09/13/2022] [Indexed: 12/30/2022]
Abstract
Renal transplantation is currently the best treatment option for patients with end-stage kidney disease. Ischemia/reperfusion injury (IRI), which is an inevitable event during renal transplantation, has a profound impact on the function of transplanted kidneys. It has been well demonstrated that innate immune system plays an important role in the process of renal IRI. As a critical component of innate immune system, Nod-like receptor family pyrin domain-containing 3 (NLRP3) inflammasome has received great attention from scientific community over the past decade. The main function of NLRP3 inflammasome is mediating activation of caspase-1 and maturation of interleukin (IL)-1β and IL-18. In this review, we summarize the associated molecular signaling events about NLRP3 inflammasome in renal IRI, and highlight the possibility of targeting NLRP3 inflammasome to minimize renal IRI during transplantation.
Collapse
Affiliation(s)
- Xiaochen Su
- Department of Urology, the First Hospital of Jilin University, Changchun 130021, Jilin, China
| | - Bin Liu
- Department of Urology, the First Hospital of Jilin University, Changchun 130021, Jilin, China
| | - Shangguo Wang
- Department of Urology, the First Hospital of Jilin University, Changchun 130021, Jilin, China
| | - Yuxiong Wang
- Department of Urology, the First Hospital of Jilin University, Changchun 130021, Jilin, China
| | - Zehua Zhang
- Department of Urology, the First Hospital of Jilin University, Changchun 130021, Jilin, China
| | - Honglan Zhou
- Department of Urology, the First Hospital of Jilin University, Changchun 130021, Jilin, China.
| | - Faping Li
- Department of Urology, the First Hospital of Jilin University, Changchun 130021, Jilin, China.
| |
Collapse
|