1
|
Zambrano-Vásquez OR, Cortés-Camacho F, Castañeda-Sánchez JI, Aréchaga-Ocampo E, Valle-Velázquez E, Cabrera-Angeles JC, Sánchez-Gloria JL, Sánchez-Muñoz F, Arellano-Buendia AS, Sánchez-Lozada LG, Osorio-Alonso H. Update in non-alcoholic fatty liver disease management: role of sodium-glucose cotransporter 2 inhibitors. Life Sci 2025; 372:123638. [PMID: 40246191 DOI: 10.1016/j.lfs.2025.123638] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2024] [Revised: 03/28/2025] [Accepted: 04/09/2025] [Indexed: 04/19/2025]
Abstract
Non-alcoholic fatty liver disease (NAFLD) is characterized by excessive lipid accumulation in hepatocytes without significant alcohol consumption. It is closely associated with sedentarism, hypercaloric diets, obesity, dyslipidemia, insulin resistance, type 2 diabetes mellitus, and genetic predisposition. NAFLD comprises a spectrum of liver disorders, from simple steatosis to non-alcoholic (NASH) and liver cirrhosis. The complex etiological mechanisms include oxidative stress, inflammation, apoptosis, and fibrosis; therefore, its management is challenging. Sodium-glucose cotransporter type 2 inhibitors (SGLT2i), a class of antidiabetic drugs, have emerged as promising therapeutic agents due to their ability to improve key metabolic parameters, including obesity, dyslipidemia, insulin resistance, and hyperglycemia. This review explores the cellular mechanisms by which SGLT2i, either as monotherapy or combined with other treatments, modulate signaling pathways involved in lipid and carbohydrate metabolism. Additionally, we examine their effects on oxidative stress, inflammation, fibrosis, and apoptosis, which are critical drivers of NAFLD progression. This review is intended to summarize the multiple benefits of SGLT2 inhibitors and to educate healthcare providers on the therapeutic potential of these drugs in order to foster their incorporation into effective NAFLD management plans.
Collapse
Affiliation(s)
- Oscar R Zambrano-Vásquez
- Doctorado en Ciencias Biológicas y de la Salud, Universidad Autónoma Metropolitana, Ciudad de México 04960, Mexico; Departamento de Fisiopatología Cardio-Renal, Instituto Nacional de Cardiología Ignacio Chávez, México City 14080, Mexico
| | - Fernando Cortés-Camacho
- Doctorado en Ciencias Biológicas y de la Salud, Universidad Autónoma Metropolitana, Ciudad de México 04960, Mexico; Departamento de Fisiopatología Cardio-Renal, Instituto Nacional de Cardiología Ignacio Chávez, México City 14080, Mexico
| | - Jorge I Castañeda-Sánchez
- Departamento de Sistemas Biológicos, Universidad Autónoma Metropolitana, Unidad Xochimilco, México City 04960, Mexico
| | - Elena Aréchaga-Ocampo
- Departamento de Ciencias Naturales, Universidad Autónoma Metropolitana, Unidad Cuajimalpa, México City 05348, Mexico
| | - Estefanía Valle-Velázquez
- Departamento de Fisiopatología Cardio-Renal, Instituto Nacional de Cardiología Ignacio Chávez, México City 14080, Mexico
| | - Juan C Cabrera-Angeles
- Sección de Estudios de Posgrado e Investigación, Escuela Superior de Medicina, Instituto Politécnico Nacional, México City, Mexico
| | - José L Sánchez-Gloria
- Department of Internal Medicine, Division of Nephrology, Rush University Medical Center, Chicago, IL 60612, USA
| | - Fausto Sánchez-Muñoz
- Departamento de Fisiología, Instituto Nacional de Cardiología Ignacio Chávez, México City 14080, Mexico
| | - Abraham S Arellano-Buendia
- Departamento de Fisiopatología Cardio-Renal, Instituto Nacional de Cardiología Ignacio Chávez, México City 14080, Mexico
| | - Laura G Sánchez-Lozada
- Departamento de Fisiopatología Cardio-Renal, Instituto Nacional de Cardiología Ignacio Chávez, México City 14080, Mexico
| | - Horacio Osorio-Alonso
- Departamento de Fisiopatología Cardio-Renal, Instituto Nacional de Cardiología Ignacio Chávez, México City 14080, Mexico.
| |
Collapse
|
2
|
Lemche E, Hortobágyi T, Kiecker C, Turkheimer F. Neuropathological links between T2DM and LOAD: systematic review and meta-analysis. Physiol Rev 2025; 105:1429-1486. [PMID: 40062731 DOI: 10.1152/physrev.00040.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2024] [Revised: 02/01/2025] [Accepted: 02/22/2025] [Indexed: 04/16/2025] Open
Abstract
Recent decades have described parallel neuropathological mechanisms increasing the risk for developing late-onset Alzheimer's dementia (LOAD) in type 2 diabetes mellitus (T2DM); however, still little is known of the role of diabetic encephalopathy and brain atrophy in LOAD. The aim of this systematic review is to provide a comprehensive view on diabetic encephalopathy/cerebral atrophy, taking into account neuroimaging data, neuropathology, metabolic and endocrine mechanisms, amyloid formation, brain perfusion impairments, neuroimmunology, and inflammasome activation. Key switches were identified, to further meta-analyze genomic candidate loci and epigenetic modifications. For the qualitative meta-analysis of genomic bases extracted, human linkage studies were examined; for epigenetic mechanisms, data from both human and animal studies are described. For the systematic review of pathophysiological mechanisms, 1,259 publications were evaluated and 93 gene loci extracted for candidate risk linkages. Sixty-six publications were evaluated for genomic association and descriptions of epigenomic modifications. Overall accumulated results highlight the insulin signaling system, vascular markers, inflammation and inflammasome pathways, amylin interactions, and glycosylation mechanisms. The protocol was registered with PROSPERO (ID: CRD42023440535).
Collapse
Affiliation(s)
- Erwin Lemche
- Section of Cognitive Neuropsychiatry, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, United Kingdom
- Centre for Neuroimaging Sciences, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, United Kingdom
| | - Tibor Hortobágyi
- Institute of Neuropathology, University Hospital Zurich, Zurich, Switzerland
- Department of Old Age Psychiatry, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, United Kingdom
- Department of Neurology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Clemens Kiecker
- Department for Developmental Neurobiology, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, United Kingdom
| | - Federico Turkheimer
- Centre for Neuroimaging Sciences, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, United Kingdom
| |
Collapse
|
3
|
Liu Y, Hu Y, Shan ZL. Mitochondrial DNA release mediates metabolic-associated steatohepatitis via activation of inflammatory pathways. Shijie Huaren Xiaohua Zazhi 2025; 33:344-360. [DOI: 10.11569/wcjd.v33.i5.344] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/24/2025] [Revised: 04/25/2025] [Accepted: 05/19/2025] [Indexed: 05/28/2025] Open
Affiliation(s)
- Ying Liu
- Gannan Institute of Medical Innovation and Translational Medicine, Gannan Medical University, Ganzhou 431000, Jiangxi Province, China
| | - Yang Hu
- Gannan Institute of Medical Innovation and Translational Medicine, Gannan Medical University, Ganzhou 431000, Jiangxi Province, China
| | - Zhao-Liang Shan
- Gannan Institute of Medical Innovation and Translational Medicine, Gannan Medical University, Ganzhou 431000, Jiangxi Province, China
| |
Collapse
|
4
|
Yang B, Xu J, Dao X, Huang Y, Liang J, Huang J, Gou B, Yan H, Chen N, Fan J. Aerobic Exercise and PI3K Inhibitor Ameliorate Obesity Cardiomyopathy by Alleviating Pyroptosis in Middle-Aged Mice. Int J Mol Sci 2025; 26:4935. [PMID: 40430076 DOI: 10.3390/ijms26104935] [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: 04/19/2025] [Revised: 05/15/2025] [Accepted: 05/16/2025] [Indexed: 05/29/2025] Open
Abstract
Obesity cardiomyopathy (OCM) represents a rapidly growing health concern globally, characterized by metabolic, structural, and functional abnormalities of the heart. Current research has demonstrated that inflammation plays a pivotal role in obesity-induced cardiomyopathy, and that regular exercise can ameliorate lipid disturbances and inflammatory abnormalities effectively. However, the underlying mechanisms are not fully elucidated. We investigated the effects of an 8-week aerobic exercise intervention on myocardial structure, function, and inflammation in HFD-induced obese mice. The results revealed that aerobic exercise alleviated myocardium pyroptosis and inflammation by down-regulating the PI3K/AKT signaling pathway. Furthermore, the inhibition of the PI3K pathway by LY294002, coupled with exercise, attenuated and suppressed HFD-induced myocardial impairments, inflammation, and pyroptosis, with a synergistic effect. Based on these findings, we concluded that eight weeks of aerobic exercise synergizes with the inhibition of PI3K through inflammatory and pyroptosis mechanisms to improve obesity-associated myocardial remodeling and dysfunction. Therefore, long-term regular aerobic exercise represents a potential strategy in the treatment of OCM.
Collapse
Affiliation(s)
- Bojun Yang
- Hubei Key Laboratory of Exercise Training and Monitoring, College of Sports Medicine, Wuhan Sports University, Wuhan 430079, China
| | - Jiahao Xu
- Hubei Key Laboratory of Exercise Training and Monitoring, College of Sports Medicine, Wuhan Sports University, Wuhan 430079, China
| | - Xiaoyan Dao
- Hubei Key Laboratory of Exercise Training and Monitoring, College of Sports Medicine, Wuhan Sports University, Wuhan 430079, China
| | - Yu Huang
- Hubei Key Laboratory of Exercise Training and Monitoring, College of Sports Medicine, Wuhan Sports University, Wuhan 430079, China
| | - Jiling Liang
- Hubei Key Laboratory of Exercise Training and Monitoring, College of Sports Medicine, Wuhan Sports University, Wuhan 430079, China
| | - Jielun Huang
- Hubei Key Laboratory of Exercise Training and Monitoring, College of Sports Medicine, Wuhan Sports University, Wuhan 430079, China
| | - Bo Gou
- Hubei Key Laboratory of Exercise Training and Monitoring, College of Sports Medicine, Wuhan Sports University, Wuhan 430079, China
| | - Hanyu Yan
- Hubei Key Laboratory of Exercise Training and Monitoring, College of Sports Medicine, Wuhan Sports University, Wuhan 430079, China
| | - Ning Chen
- Hubei Key Laboratory of Exercise Training and Monitoring, College of Sports Medicine, Wuhan Sports University, Wuhan 430079, China
| | - Jingjing Fan
- Hubei Key Laboratory of Exercise Training and Monitoring, College of Sports Medicine, Wuhan Sports University, Wuhan 430079, China
| |
Collapse
|
5
|
El-Sheikh H, El-Haggar S, Badawi R, Habba E. Comparative efficacy of febuxostat and vitamin E in the management of MASLD: Insights from a randomized parallel clinical study. Eur J Pharmacol 2025; 1000:177735. [PMID: 40383220 DOI: 10.1016/j.ejphar.2025.177735] [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: 03/12/2025] [Revised: 05/11/2025] [Accepted: 05/15/2025] [Indexed: 05/20/2025]
Abstract
AIM We aimed to determine whether inhibiting xanthine oxidase (XO) activity and NLRP-3 activation in the liver with febuxostat could influence the progression of metabolic dysfunction-associated steatotic liver disease (MASLD). METHODS Sixty-four MASLD patients were divided into two groups: 32 patients received 80 mg of febuxostat daily, while the other 32 patients received 400 mg of vitamin E twice daily for 24 weeks. A gastroenterologist assessed the degree of steatosis using Fibroscan and controlled attenuation parameter (CAP) measurements at baseline and after six months. Additionally, hepatic steatosis index (HSI), HAIR-score, and levels of NLRP-3, TIM-3, HOMA-IR, MDA, uric acid, lipid profile, and liver function tests were measured before and after treatment. RESULTS Improvement in steatosis was observed in 50 % of febuxostat group and 46.9 % of vitamin E group. Both groups showed a significant reduction in CAP scores, with more pronounced decrease in vitamin E group (p < 0.001) compared to febuxostat group (p = 0.001). Febuxostat group exhibited significantly lower levels of NLRP-3, MDA, TIM-3, and uric acid compared to vitamin E group. HSI, HAIR score, and liver functions improved similarly in both groups. CONCLUSION Febuxostat appears to be effective in reducing steatosis in MASLD patients, suggesting its potential as a treatment option for non-cirrhotic MASLD. CLINICALTRIALS gov identifier is NCT05574036, Registered October 6, 2022 - Retrospectively.
Collapse
Affiliation(s)
- Hadier El-Sheikh
- Department of Clinical Pharmacy, Faculty of Pharmacy, Tanta University, Al-Guiesh Street, Tanta, 31527, Egypt.
| | - Sahar El-Haggar
- Department of Clinical Pharmacy, Faculty of Pharmacy, Tanta University, Al-Guiesh Street, Tanta, 31527, Egypt.
| | - Rehab Badawi
- Department of Tropical Medicine, and Infectious Diseases, Faculty of Medicine, Tanta University, Al-Guiesh Street, Tanta, 31527, Egypt.
| | - Eslam Habba
- Department of Tropical Medicine, and Infectious Diseases, Faculty of Medicine, Tanta University, Al-Guiesh Street, Tanta, 31527, Egypt.
| |
Collapse
|
6
|
Zeng X, Yuan Y, Li Y, Hu Z, Hu S. Deciphering the NLRP3 inflammasome in diabetic encephalopathy: Molecular insights and emerging therapeutic targets. Exp Neurol 2025; 391:115304. [PMID: 40383363 DOI: 10.1016/j.expneurol.2025.115304] [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: 12/17/2024] [Revised: 05/01/2025] [Accepted: 05/11/2025] [Indexed: 05/20/2025]
Abstract
Diabetic encephalopathy (DE) is a neurological complication characterized by neuroinflammation, cognitive impairment, and memory decline, with its pathogenesis closely linked to the activation of the NLRP3 inflammasome. As a central regulator of the innate immune system, the NLRP3 inflammasome plays a pivotal role in DE progression by mediating neuroinflammation, pyroptosis, mitochondrial dysfunction, oxidative stress, endoplasmic reticulum (ER) stress, and microglial polarization. This review systematically explores the molecular mechanisms by which the NLRP3 inflammasome contributes to DE, focusing on its role in neuroinflammatory cascades and neuronal damage, as well as the diabetes-associated physiological changes that exacerbate DE pathogenesis. Furthermore, we summarize emerging therapeutic strategies targeting the NLRP3 inflammasome, including small-molecule inhibitors and bioactive compounds derived from traditional herbal medicine, highlighting their potential for DE treatment. These findings not only advance our understanding of DE but also provide a foundation for developing NLRP3-targeted pharmacological interventions.
Collapse
Affiliation(s)
- Xinyi Zeng
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330031, China; The First Clinical Medical College of Nanchang University, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330031, China
| | - Yi Yuan
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330031, China; School of Huankui Academy, Nanchang University, Nanchang, Jiangxi 330031, China
| | - Yujia Li
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330031, China; The Second Clinical Medical College of Nanchang University, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330031, China
| | - Ziyan Hu
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330031, China; The Second Clinical Medical College of Nanchang University, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330031, China
| | - Shan Hu
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330031, China.
| |
Collapse
|
7
|
Cao P, Yang Y, Zhang N, Wang B, Gong Z. Inflammasomes: novel therapeutic targets for metabolic syndrome? Front Endocrinol (Lausanne) 2025; 16:1569579. [PMID: 40433411 PMCID: PMC12106043 DOI: 10.3389/fendo.2025.1569579] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2025] [Accepted: 04/18/2025] [Indexed: 05/29/2025] Open
Abstract
Chronic inflammation is a hallmark for Metabolic Syndrome (MetS). It is also one of the most important risk factors for insulin resistance and metabolic disorders. Inflammasomes, which are intracellular multiprotein complexes within the innate immune system, regulate the production and maturation of pro-inflammatory cytokines including interleukin-1β (IL-1β) and IL-18 upon sensing pathogens or danger signals in the cytosol. A growing body of evidence indicates that inflammasomes play a pivotal role in the pathophysiology and progression of metabolic diseases, as deficiency in the key component of inflammasomes protects mice from high fat diet induced obesity and insulin resistance. Thus, in this review, we will summarize the role of inflammasomes in MetS and how to treat MetS by targeting inflammasomes. This may provide novel insights and therapeutic targets for treating metabolic disorders.
Collapse
Affiliation(s)
- Pengyu Cao
- The Second People’s Hospital of Changzhou, the Third Affiliated Hospital of Nanjing Medical University, Changzhou, Jiangsu, China
- Changzhou Medical Center, Nanjing Medical University, Changzhou, Jiangsu, China
| | - Yulin Yang
- The Second People’s Hospital of Changzhou, the Third Affiliated Hospital of Nanjing Medical University, Changzhou, Jiangsu, China
- Changzhou Medical Center, Nanjing Medical University, Changzhou, Jiangsu, China
| | - Ningning Zhang
- The Second People’s Hospital of Changzhou, the Third Affiliated Hospital of Nanjing Medical University, Changzhou, Jiangsu, China
| | - Bojian Wang
- School of Nursing, Jilin University, Changchun, Jilin, China
| | - Zhenwei Gong
- Division of Endocrinology, Department of Pediatrics, Children’s Hospital of Pittsburgh of UPMC, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| |
Collapse
|
8
|
Li Z, Cheng W, Gao K, Liang S, Ke L, Wang M, Fan J, Li D, Zhang P, Xu Z, Li N. Pyroptosis: A spoiler of peaceful coexistence between cells in degenerative bone and joint diseases. J Adv Res 2025; 71:227-262. [PMID: 38876191 DOI: 10.1016/j.jare.2024.06.010] [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: 02/17/2024] [Revised: 05/23/2024] [Accepted: 06/07/2024] [Indexed: 06/16/2024] Open
Abstract
BACKGROUND As people age, degenerative bone and joint diseases (DBJDs) become more prevalent. When middle-aged and elderly people are diagnosed with one or more disorders such as osteoporosis (OP), osteoarthritis (OA), and intervertebral disc degeneration (IVDD), it often signals the onset of prolonged pain and reduced functionality. Chronic inflammation has been identified as the underlying cause of various degenerative diseases, including DBJDs. Recently, excessive activation of pyroptosis, a form of programed cell death (PCD) mediated by inflammasomes, has emerged as a primary driver of harmful chronic inflammation. Consequently, pyroptosis has become a potential target for preventing and treating DBJDs. AIM OF REVIEW This review explored the physiological and pathological roles of the pyroptosis pathway in bone and joint development and its relation to DBJDs. Meanwhile, it elaborated the molecular mechanisms of pyroptosis within individual cell types in the bone marrow and joints, as well as the interplay among different cell types in the context of DBJDs. Furthermore, this review presented the latest compelling evidence supporting the idea of regulating the pyroptosis pathway for DBJDs treatment, and discussed the potential, limitations, and challenges of various therapeutic strategies involving pyroptosis regulation. KEY SCIENTIFIC CONCEPTS OF REVIEW In summary, an interesting identity for the unregulated pyroptosis pathway in the context of DBJDs was proposed in this review, which was undertaken as a spoiler of peaceful coexistence between cells in a degenerative environment. Over the extended course of DBJDs, pyroptosis pathway perpetuated its activity through crosstalk among pyroptosis cascades in different cell types, thus exacerbating the inflammatory environment throughout the entire bone marrow and joint degeneration environment. Correspondingly, pyroptosis regulation therapy emerged as a promising option for clinical treatment of DBJDs.
Collapse
Affiliation(s)
- Zhichao Li
- First College of Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan 250014, China; Department of Orthopedics, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan 250014, China; Center for Translational Medicine Research and Development, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Wenxiang Cheng
- Center for Translational Medicine Research and Development, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Kuanhui Gao
- First College of Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan 250014, China
| | - Songlin Liang
- First College of Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan 250014, China; Center for Translational Medicine Research and Development, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Liqing Ke
- Center for Translational Medicine Research and Development, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Mengjie Wang
- First College of Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan 250014, China
| | - Jilin Fan
- First College of Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan 250014, China
| | - Dandan Li
- College of Integrated Traditional Chinese and Western Medicine, Hebei University of Chinese Medicine, Shijiazhuang 050011, China
| | - Peng Zhang
- Center for Translational Medicine Research and Development, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China; Faculty of Biomedical Engineering, Shenzhen University of Advanced Technology, Shenzhen 518000, China; Key Laboratory of Biomedical Imaging Science and System, Chinese Academy of Sciences, Shenzhen, 518000 China; Shandong Zhongke Advanced Technology Co., Ltd., Jinan, 250300 China.
| | - Zhanwang Xu
- First College of Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan 250014, China; Department of Orthopedics, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan 250014, China.
| | - Nianhu Li
- First College of Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan 250014, China; Department of Orthopedics, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan 250014, China.
| |
Collapse
|
9
|
Zhang S, Lin H, Wang J, Rui J, Wang T, Cai Z, Huang S, Gao Y, Ma T, Fan R, Dai R, Li Z, Jia Y, Chen Q, He H, Tan J, Zhu S, Gu R, Dong Z, Li M, Xie E, Fu Y, Zheng J, Jiang C, Sun J, Kong W. Sensing ceramides by CYSLTR2 and P2RY6 to aggravate atherosclerosis. Nature 2025; 641:476-485. [PMID: 40049228 DOI: 10.1038/s41586-025-08792-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Accepted: 02/14/2025] [Indexed: 04/18/2025]
Abstract
Recent evidence has shown that increased levels of circulating long-chain ceramides predict atherosclerotic cardiovascular disease independently of cholesterol1,2. Although targeting ceramide signalling may provide therapeutic benefits beyond the treatment of hypercholesterolaemia, the underlying mechanism by which circulating ceramides aggravate atherosclerotic cardiovascular disease remains elusive. Here we examine whether circulating long-chain ceramides activate membrane G-protein-coupled receptors to exacerbate atherosclerosis. We perform a systematic screen that combines G-protein-signalling quantification, bioinformatic analysis of G-protein-coupled receptor expression and functional examination of NLRP3 inflammasome activation. The results suggest that CYSLTR2 and P2RY6 are potential endogenous receptors of C16:0 ceramide-induced inflammasome activation in both endothelial cells and macrophages. Inhibition of CYSLTR2 and P2RY6 genetically or pharmacologically alleviates ceramide-induced atherosclerosis aggravation. Moreover, increased ceramide levels correlate with the severity of coronary artery disease in patients with varying degrees of renal impairment. Notably, CYSLTR2 and P2RY6 deficiency mitigates chronic-kidney-disease-aggravated atherosclerosis in mice without affecting cholesterol or ceramide levels. Structural analyses of ceramide-CYSLTR2-Gq complexes reveal that both C16:0 and C20:0 ceramides bind in an inclined channel-like ligand-binding pocket on CYSLTR2. We further reveal an unconventional mechanism underlying ceramide-induced CYSLTR2 activation and the CYSLTR2-Gq interface. Overall, our study provides structural and molecular mechanisms of how long-chain ceramides initiate transmembrane Gq and inflammasome signalling through direct binding to CYSLTR2 and P2RY6 receptors. Therefore, blocking these signals may provide a new therapeutic potential to treat atherosclerosis-related diseases.
Collapse
MESH Headings
- Animals
- Atherosclerosis/metabolism
- Atherosclerosis/pathology
- Humans
- Mice
- Ceramides/metabolism
- Ceramides/blood
- Ceramides/chemistry
- Male
- Inflammasomes/metabolism
- Receptors, Leukotriene/metabolism
- Receptors, Leukotriene/chemistry
- Receptors, Leukotriene/genetics
- Receptors, Leukotriene/deficiency
- Receptors, Purinergic P2/metabolism
- Receptors, Purinergic P2/chemistry
- Receptors, Purinergic P2/genetics
- Receptors, Purinergic P2/deficiency
- Female
- NLR Family, Pyrin Domain-Containing 3 Protein/metabolism
- Coronary Artery Disease/pathology
- Coronary Artery Disease/metabolism
- Macrophages/metabolism
- Renal Insufficiency, Chronic/complications
- Renal Insufficiency, Chronic/metabolism
- Renal Insufficiency, Chronic/pathology
- Signal Transduction
- Mice, Inbred C57BL
- Models, Molecular
Collapse
Affiliation(s)
- Siting Zhang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, P. R. China
- State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, P. R. China
| | - Hui Lin
- New Cornerstone Science Laboratory, Advanced Medical Research Institute, Cheeloo College of Medicine, Shandong University, Jinan, China
- Department of Periodontology, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University and Shandong Key Laboratory of Oral Tissue Regeneration, Jinan, China
- Department of Biophysics, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, P. R. China
| | - Jiale Wang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, P. R. China
- State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, P. R. China
- Department of Biophysics, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, P. R. China
| | - Jingyu Rui
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, P. R. China
| | - Tengwei Wang
- New Cornerstone Science Laboratory, Advanced Medical Research Institute, Cheeloo College of Medicine, Shandong University, Jinan, China
- School of Pharmacy, Binzhou Medical University, Yantai, China
| | - Zeyu Cai
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, P. R. China
- State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, P. R. China
| | - Shenming Huang
- New Cornerstone Science Laboratory, Advanced Medical Research Institute, Cheeloo College of Medicine, Shandong University, Jinan, China
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Jinan University, Guangzhou, China
| | - Yanxiang Gao
- Department of Cardiology, Peking University China-Japan Friendship School of Clinical Medicine, Beijing, China
| | - Tianfeng Ma
- Department of Vascular and Endovascular Surgery, the First Medical Centre, Chinese PLA General Hospital, Beijing, China
| | - Rui Fan
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, P. R. China
- State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, P. R. China
- Department of Biomedical Informatics, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, P. R. China
| | - Rongbo Dai
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, P. R. China
- State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, P. R. China
| | - Zhiqing Li
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, P. R. China
- State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, P. R. China
| | - Yiting Jia
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, P. R. China
- State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, P. R. China
- Department of Pharmacology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, P. R. China
| | - Qiang Chen
- Department of Cardiology, Peking University China-Japan Friendship School of Clinical Medicine, Beijing, China
| | - HaoMing He
- Department of Cardiology, Peking University China-Japan Friendship School of Clinical Medicine, Beijing, China
| | - Jiaai Tan
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, P. R. China
- State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, P. R. China
| | - Shirong Zhu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, P. R. China
- State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, P. R. China
| | - Rui Gu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, P. R. China
- State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, P. R. China
| | - Zhigang Dong
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, P. R. China
- State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, P. R. China
| | - Meihong Li
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, P. R. China
- State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, P. R. China
| | - Enmin Xie
- Department of Cardiology, Peking University China-Japan Friendship School of Clinical Medicine, Beijing, China
| | - Yi Fu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, P. R. China
- State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, P. R. China
| | - Jingang Zheng
- Department of Cardiology, Peking University China-Japan Friendship School of Clinical Medicine, Beijing, China.
| | - Changtao Jiang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, P. R. China.
- State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, P. R. China.
| | - Jinpeng Sun
- State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, P. R. China.
- New Cornerstone Science Laboratory, Advanced Medical Research Institute, Cheeloo College of Medicine, Shandong University, Jinan, China.
- Department of Biophysics, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, P. R. China.
- NHC Key Laboratory of Otorhinolaryngology, Qilu Hospital of Shandong University, Shandong, China.
| | - Wei Kong
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, P. R. China.
- State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, P. R. China.
| |
Collapse
|
10
|
Baumer Y, Irei J, Boisvert WA. Cholesterol crystals in the pathogenesis of atherosclerosis. Nat Rev Cardiol 2025; 22:315-332. [PMID: 39558130 DOI: 10.1038/s41569-024-01100-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 10/23/2024] [Indexed: 11/20/2024]
Abstract
The presence of cholesterol crystals (CCs) in tissues was first described more than 100 years ago. CCs have a pathogenic role in various cardiovascular diseases, including myocardial infarction, aortic aneurysm and, most prominently, atherosclerosis. Although the underlying mechanisms and signalling pathways involved in CC formation are incompletely understood, numerous studies have highlighted the existence of CCs at various stages of atheroma progression. In this Review, we summarize the mechanisms underlying CC formation and the role of CCs in cardiovascular disease. In particular, we explore the established links between lipid metabolism across various cell types and the formation of CCs, with a focus on CC occurrence in the vasculature. We also discuss CC-induced inflammation as one of the pathogenic features of CCs in the atheroma. Finally, we summarize the therapeutic strategies aimed at reducing CC-mediated atherosclerotic burden, including approaches to inhibit CC formation in the vasculature or to mitigate the inflammatory response triggered by CCs. Addressing CC formation might emerge as a crucial component in our broader efforts to combat cardiovascular disease.
Collapse
Affiliation(s)
- Yvonne Baumer
- Social Determinants of Obesity and Cardiovascular Risk Laboratory, NIH, NHLBI, Bethesda, MD, USA
| | - Jason Irei
- Center for Cardiovascular Research, John A. Burns School of Medicine, University of Hawaii, Honolulu, HI, USA
| | - William A Boisvert
- Center for Cardiovascular Research, John A. Burns School of Medicine, University of Hawaii, Honolulu, HI, USA.
| |
Collapse
|
11
|
Thornton P, Reader V, Digby Z, Doedens J, Lindsay N, Clarke N, Watt AP. The NLRP3 inhibitor NT-0796 enhances and sustains GLP-1R agonist-mediated weight loss in a murine diet-induced obesity model. Obesity (Silver Spring) 2025. [PMID: 40304241 DOI: 10.1002/oby.24305] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/03/2025] [Revised: 03/11/2025] [Accepted: 03/29/2025] [Indexed: 05/02/2025]
Abstract
OBJECTIVE In order to investigate whether a central nervous system penetrant anti-inflammatory could augment or sustain obesity treatment with semaglutide (Wegovy), a glucagon-like peptide-1 receptor (GLP-1R) agonist, we tested two hypotheses in models of diet-induced obesity (DIO): 1) a centrally penetrant NLPR3 inhibitor, NT-0796, drives enhanced weight loss when combined with low-dose semaglutide, compared to monotherapy; and 2) NT-0796 monotherapy sustains weight loss induced by semaglutide. METHODS Mice fed a standard high-fat or a polyunsaturated fatty acid diet served as models of DIO and were dosed with low-dose semaglutide, NT-0796, or combinations. Body weight, food intake, peripheral inflammatory markers, and hypothalamic glial fibrillary acidic protein expression were assessed. RESULTS Combined dosing of NT-0796 with semaglutide drove greater weight loss than either monotherapy alone, and this effect was enhanced in mice consuming the polyunsaturated fatty acid diet. In addition, NT-0796 sharply limited weight regain following cessation of semaglutide therapy and normalized markers of both peripheral inflammation and hypothalamic astrogliosis to a far greater extent than either semaglutide or calorie restriction. CONCLUSIONS Alleviation of obesity-associated inflammation via NLRP3 inhibition 1) constitutes an effective weight-loss strategy as monotherapy in mice with DIO, 2) augments the weight-loss efficacy of a subtherapeutic dose of semaglutide, and 3) blocks recovery of lost weight following cessation of semaglutide.
Collapse
|
12
|
Paik S, Kim JK, Shin HJ, Park EJ, Kim IS, Jo EK. Updated insights into the molecular networks for NLRP3 inflammasome activation. Cell Mol Immunol 2025:10.1038/s41423-025-01284-9. [PMID: 40307577 DOI: 10.1038/s41423-025-01284-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2024] [Accepted: 03/17/2025] [Indexed: 05/02/2025] Open
Abstract
Over the past decade, significant advances have been made in our understanding of how NACHT-, leucine-rich-repeat-, and pyrin domain-containing protein 3 (NLRP3) inflammasomes are activated. These findings provide detailed insights into the transcriptional and posttranslational regulatory processes, the structural-functional relationship of the activation processes, and the spatiotemporal dynamics of NLRP3 activation. Notably, the multifaceted mechanisms underlying the licensing of NLRP3 inflammasome activation constitute a focal point of intense research. Extensive research has revealed the interactions of NLRP3 and its inflammasome components with partner molecules in terms of positive and negative regulation. In this Review, we provide the current understanding of the complex molecular networks that play pivotal roles in regulating NLRP3 inflammasome priming, licensing and assembly. In addition, we highlight the intricate and interconnected mechanisms involved in the activation of the NLRP3 inflammasome and the associated regulatory pathways. Furthermore, we discuss recent advances in the development of therapeutic strategies targeting the NLRP3 inflammasome to identify potential therapeutics for NLRP3-associated inflammatory diseases. As research continues to uncover the intricacies of the molecular networks governing NLRP3 activation, novel approaches for therapeutic interventions against NLRP3-related pathologies are emerging.
Collapse
Affiliation(s)
- Seungwha Paik
- Department of Microbiology, Chungnam National University College of Medicine, Daejeon, Republic of Korea
- Department of Medical Science, Chungnam National University College of Medicine, Daejeon, Republic of Korea
- System Network Inflammation Control Research Center, Chungnam National University College of Medicine, Daejeon, Republic of Korea
- Biomedical Research Institute, Chungnam National University Hospital, Daejeon, Republic of Korea
| | - Jin Kyung Kim
- Department of Microbiology, Keimyung University School of Medicine, Daegu, Republic of Korea
| | - Hyo Jung Shin
- Department of Medical Science, Chungnam National University College of Medicine, Daejeon, Republic of Korea
- Department of Biochemistry and Cell Biology, Eulji University School of Medicine, Daejeon, Republic of Korea
- Brain Research Institute, Chungnam National University College of Medicine, Daejeon, Republic of Korea
| | - Eun-Jin Park
- Department of Microbiology, Chungnam National University College of Medicine, Daejeon, Republic of Korea
- Department of Medical Science, Chungnam National University College of Medicine, Daejeon, Republic of Korea
| | - In Soo Kim
- Department of Medical Science, Chungnam National University College of Medicine, Daejeon, Republic of Korea
- Biomedical Research Institute, Chungnam National University Hospital, Daejeon, Republic of Korea
- Department of Pharmacology, Chungnam National University College of Medicine, Daejeon, Republic of Korea
| | - Eun-Kyeong Jo
- Department of Microbiology, Chungnam National University College of Medicine, Daejeon, Republic of Korea.
- Department of Medical Science, Chungnam National University College of Medicine, Daejeon, Republic of Korea.
- Biomedical Research Institute, Chungnam National University Hospital, Daejeon, Republic of Korea.
| |
Collapse
|
13
|
Wang R, Gao Y, Wang Y, Zhang Y, Yang R. LncRNA29RIK in macrophages promotes LPS-mediated sensitivity to obesity. Front Immunol 2025; 16:1574507. [PMID: 40356927 PMCID: PMC12066258 DOI: 10.3389/fimmu.2025.1574507] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2025] [Accepted: 03/25/2025] [Indexed: 05/15/2025] Open
Abstract
Lipopolysaccharide (LPS, endotoxin) -mediated signaling of caspase-4 (human) and -11 (rodent) can induce the maturation of inflammatory cytokine IL-1β and cell pyroptosis, which is associated with the pathophysiology of many diseases such as obesity. However, the process by which LPS induces inflammation through caspase 4/11 is not fully understood. We found here that lncRNA29RIK plays a key role in LPS-mediated maturation of inflammatory cytokine IL-1β and pyroptosis of macrophages. Mechanistic ally, the binding of caspase 4/11 to LPS requires lncRNARIK to cause activation of the caspase 4/11 complex, which ultimately caused inflammation to promote sensitivity to high fat diet (HFD) -mediated obesity. Notably, lncRNA29RIK expression can be up-regulated by LPS. This lncRNA29 is highly conserved between humans and mice. Taken together, these results suggest that lncRNA29RIK determines the occurrence and progression of LPS-related diseases such as obesity.
Collapse
Affiliation(s)
- Rong Wang
- Department of Immunology, Nankai University School of Medicine, Nankai University, Tianjin, China
| | - Yunhuan Gao
- Department of Immunology, Nankai University School of Medicine, Nankai University, Tianjin, China
| | - Ya Wang
- Department of Immunology, Nankai University School of Medicine, Nankai University, Tianjin, China
| | - Yuan Zhang
- Department of Immunology, Nankai University School of Medicine, Nankai University, Tianjin, China
| | - Rongcun Yang
- Department of Immunology, Nankai University School of Medicine, Nankai University, Tianjin, China
- Translational Medicine Institute, Affiliated Tianjin Union Medical Center of Nankai University, Tianjin, China
- State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin, China
| |
Collapse
|
14
|
Shi R, Zhuang X, Liu T, Yao SN, Xue FS. The Role of NLRP3 Inflammasome in Oral Squamous Cell Carcinoma. J Inflamm Res 2025; 18:5601-5609. [PMID: 40303006 PMCID: PMC12039833 DOI: 10.2147/jir.s512770] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2024] [Accepted: 03/21/2025] [Indexed: 05/02/2025] Open
Abstract
Background Oral squamous cell carcinoma (OSCC) is the most common malignant tumor in the head and neck. More and more evidence emphasizes the importance of inflammation in the progression of OSCC. The main signaling pathway of acute and chronic inflammation consists of the activation of the NLR family pyrin domain containing 3 (NLRP3) inflammasome. Objective This review focuses on the role of NLRP3 immune kinase body and giving a contribution to the development of new treatment strategies against OSCC. Conclusion The NLRP3 inflammasome plays a vital role in the pathogenesis and development of OSCC and may serve as a promising therapeutic target for autoimmune diseases.
Collapse
Affiliation(s)
- Rui Shi
- Department of Oral and Maxillofacial Reconstruction, The Affiliated Hospital of Qingdao University 266600, Qingdao, 266555, People’s Republic of China
- School of Stomatology of Qingdao University, Qingdao, 266555, People’s Republic of China
| | - Xuan Zhuang
- Cardiac Surgery Intensive Care Unit Department, the Affiliated Hospital of Qingdao University, Qingdao, 266555, People’s Republic of China
| | - Tong Liu
- The Affiliated Tai’an City Central Hospital of Qingdao University, Taian, 271000, People’s Republic of China
| | - Song-nan Yao
- Cardiac Surgery Intensive Care Unit Department, the Affiliated Hospital of Qingdao University, Qingdao, 266555, People’s Republic of China
| | - Feng-shan Xue
- Cardiac Surgery Intensive Care Unit Department, the Affiliated Hospital of Qingdao University, Qingdao, 266555, People’s Republic of China
| |
Collapse
|
15
|
Li W, Liu T, Chen Y, Sun Y, Li C, Dong Y. Regulation and therapeutic potential of NLRP3 inflammasome in intestinal diseases. J Leukoc Biol 2025; 117:qiaf014. [PMID: 40276926 DOI: 10.1093/jleuko/qiaf014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2024] [Indexed: 04/26/2025] Open
Abstract
The NOD-like receptor family, particularly the protein 3 that contains the pyrin domain (NLRP3), is an intracellular sensing protein complex responsible for detecting patterns associated with pathogens and injuries. NLRP3 plays a crucial role in the innate immune response. Currently, a wide range of research has indicated the crucial importance of NLRP3 in various inflammatory conditions. Similarly, the NLRP3 inflammasome plays a significant role in preserving intestinal balance and impacting the advancement of diseases. In addition, several randomized trials have demonstrated the safety and efficacy of targeting NLRP3 in the treatment of colitis, colorectal cancer, and related diseases. This review explores the mechanisms of NLRP3 assembly and activation in the gut. We describe its pathological significance in intestinal diseases. Finally, we summarize current and future therapeutic approaches targeting NLRP3 for intestinal diseases.
Collapse
Affiliation(s)
- Wenxue Li
- National Key Laboratory of Veterinary Public Health and Safety, College of Veterinary Medicine, China Agricultural University, Haidian, Beijing 100193, China
| | - Tianya Liu
- National Key Laboratory of Veterinary Public Health and Safety, College of Veterinary Medicine, China Agricultural University, Haidian, Beijing 100193, China
| | - Yaoxing Chen
- National Key Laboratory of Veterinary Public Health and Safety, College of Veterinary Medicine, China Agricultural University, Haidian, Beijing 100193, China
| | - Yan Sun
- Department of Horticulture and Landscape Architecture, Jiangsu Agri-Animal Husbandry Vocational College, Taizhou 225300, China
| | - Chengzhong Li
- Department of Horticulture and Landscape Architecture, Jiangsu Agri-Animal Husbandry Vocational College, Taizhou 225300, China
| | - Yulan Dong
- National Key Laboratory of Veterinary Public Health and Safety, College of Veterinary Medicine, China Agricultural University, Haidian, Beijing 100193, China
| |
Collapse
|
16
|
Iwaki T, Kurano M, Sumitani M, Niimi A, Nomiya A, Kamei J, Taguchi S, Yamada Y, Sato Y, Nakamura M, Yamada D, Minagawa T, Fukuhara H, Kume H, Homma Y, Akiyama Y. Lipidomic analysis coupled with machine learning identifies unique urinary lipid signatures in patients with interstitial cystitis/bladder pain syndrome. World J Urol 2025; 43:233. [PMID: 40249505 PMCID: PMC12008056 DOI: 10.1007/s00345-025-05628-y] [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: 02/12/2025] [Accepted: 04/09/2025] [Indexed: 04/19/2025] Open
Abstract
PURPOSE To identify biomarkers for diagnosis and classification of interstitial cystitis/bladder pain syndrome (IC/BPS) by urinary lipidomics coupled with machine learning. METHODS Urine samples from 138 patients with IC/BPS, including 116 with Hunner lesion (HL) and 22 with no HL, and 71 controls were assessed by lipid chromatography-tandem mass spectrometry. Single and paired lipid analyses of differentially expressed lipids in each group were conducted to assess their diagnostic ability. Machine learning models were constructed based on the identified urinary lipids and patient demographic data, and a five-fold cross-validation method was applied for internal validation. Levels of urinary lipids were adjusted to account for urinary creatinine levels. RESULTS A total of 218 urinary lipids were identified. Single lipid analysis revealed that urinary levels of C24 ceramide and LPC (14:0) distinguished HL and no HL, with an area under the receiver operating characteristics curve of 0.792 and 0.656, respectively. Paired lipid analysis revealed that summed urinary levels of C24 ceramide and LPI (18:3), and subtraction of PG (36:5) from PC (38:2) distinguished HL and no HL even more accurately, with an area under the curve of 0.805 and 0.752, respectively. A machine learning model distinguished HL and no HL, with the highest area under the curve being 0.873 and 0.750, respectively. Limitations include the opaque black box nature of machine learning techniques. CONCLUSIONS Urinary levels of C24 ceramide, along with those of C24 ceramide plus LPI (18:3), could be potential biomarkers for HL. Machine learning-coupled urinary lipidomics may play an important role in the next-generation AI- driven diagnostic systems for IC/BPS.
Collapse
Affiliation(s)
- Takuya Iwaki
- Department of Urology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
- Department of Urology, Chiba Tokushukai Hospital, Chiba, Japan
| | - Makoto Kurano
- Department of Clinical Laboratory Medicine, The University of Tokyo, Tokyo, Japan.
| | - Masahiko Sumitani
- Department of Pain and Palliative Medicine, The University of Tokyo Hospital, Tokyo, Japan
| | - Aya Niimi
- Department of Urology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Akira Nomiya
- Department of Urology, Japan Organization of Occupational Health and Safety, Kanto Rosai Hospital, Kanagawa, Japan
| | - Jun Kamei
- Department of Urology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Satoru Taguchi
- Department of Urology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Yuta Yamada
- Department of Urology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Yusuke Sato
- Department of Urology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
- Department of Urology, Tokyo Metropolitan Tama Medical Center, Tokyo, Japan
| | - Masaki Nakamura
- Department of Urology, NTT Medical Center Tokyo, Tokyo, Japan
| | - Daisuke Yamada
- Department of Urology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Tomonori Minagawa
- Department of Urology, Shinshu University School of Medicine, Nagano, Japan
| | - Hiroshi Fukuhara
- Department of Urology, Kyorin University School of Medicine, Tokyo, Japan
| | - Haruki Kume
- Department of Urology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Yukio Homma
- Department of Interstitial Cystitis Medicine, Kyorin University School of Medicine, Tokyo, Japan
| | - Yoshiyuki Akiyama
- Department of Urology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan.
- Department of Urology, Shinshu University School of Medicine, Nagano, Japan.
| |
Collapse
|
17
|
Wang X, Xie Z, Zhang J, Chen Y, Li Q, Yang Q, Chen X, Liu B, Xu S, Dong Y. Interaction between lipid metabolism and macrophage polarization in atherosclerosis. iScience 2025; 28:112168. [PMID: 40201117 PMCID: PMC11978336 DOI: 10.1016/j.isci.2025.112168] [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] [Indexed: 04/10/2025] Open
Abstract
Atherosclerosis (AS) is a chronic inflammatory condition associated with lipid deposition. The interaction between abnormal lipid metabolism and the inflammatory response has been identified as the underlying cause of AS. Lipid metabolism disorders are considered the basis of atherosclerotic lesion formation and macrophages are involved in the entire process of AS formation. Macrophages have a high degree of plasticity, and the change of their polarization direction can determine the progress or regression of AS. The disturbances in bioactive lipid metabolism affect the polarization of different phenotypes of macrophages, thus, affecting lipid metabolism and the expression of key signal factors. Therefore, understanding the interaction between lipid metabolism and macrophages as well as their key targets is important for preventing and treating AS and developing new drugs. Recent studies have shown that traditional Chinese medicines play a positive role in the prevention and treatment of AS, providing a basis for clinical individualized treatment.
Collapse
Affiliation(s)
- Xinge Wang
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, China; School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
- Guang’ anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China
| | - Zheng Xie
- Guang’ anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China
| | - Jing Zhang
- Tianjin State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Ying Chen
- Institute of Chinese Materia Medica China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Qi Li
- Institute of Chinese Materia Medica China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Qing Yang
- Institute of Chinese Materia Medica China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Xu Chen
- Guang’ anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China
| | - Bing Liu
- Guang’ anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China
| | - Shijun Xu
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, China; School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Yu Dong
- Guang’ anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China
| |
Collapse
|
18
|
Bielach-Bazyluk A, Bossowski F, Skorupska M, Mysliwiec H, Bossowski AT, Flisiak I. Psoriasis in Obese Adolescents with Diabetes-From Common Molecular Background to Vicious Circle of Metabolic Syndrome-Case Report and Review of Literature. Cells 2025; 14:610. [PMID: 40277935 PMCID: PMC12026325 DOI: 10.3390/cells14080610] [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: 03/17/2025] [Revised: 04/10/2025] [Accepted: 04/14/2025] [Indexed: 04/26/2025] Open
Abstract
Psoriasis and type 1 diabetes mellitus (T1DM) are chronic autoimmune diseases sharing common immunological pathways, particularly the involvement of interleukin 17 (IL-17), driving Th17-mediated inflammation. This review explores the overlap between psoriasis, obesity, T1DM, and necrobiosis lipoidica (NL), a skin condition associated with diabetes. Obesity exacerbates inflammation through immune cell activation in adipose tissue and the release of proinflammatory adipokines, such as leptin, resistin, and IL-18, which enhance autoimmune responses and insulin resistance. Leptin promotes the differentiation of Th1 and Th17 cells, which are central to autoimmune responses in both psoriasis and T1DM. The coexistence of psoriasis, T1DM, and insulin resistance further complicates metabolic control, increasing the risk of complications like diabetic nephropathy and cardiovascular disease. Biologic treatments targeting IL-17A and IL-17F offer promising therapeutic options for managing both skin and metabolic symptoms. The early identification and management of metabolic risk factors, along with personalized interventions, are essential to improve clinical outcomes in patients with psoriasis and T1DM, particularly in obese individuals. This case report and review highlight the complex interplay of these conditions and emphasize the need for integrated treatment strategies.
Collapse
Affiliation(s)
- Angelika Bielach-Bazyluk
- Department of Dermatology and Venereology, Medical University of Bialystok, 15-540 Bialystok, Poland; (H.M.)
| | - Filip Bossowski
- Students’ Scientific Society at the Department of Dermatology and Venereology, Medical University of Bialystok, 15-540 Bialystok, Poland
| | - Magdalena Skorupska
- Students’ Scientific Society at the Department of Dermatology and Venereology, Medical University of Bialystok, 15-540 Bialystok, Poland
| | - Hanna Mysliwiec
- Department of Dermatology and Venereology, Medical University of Bialystok, 15-540 Bialystok, Poland; (H.M.)
| | - Artur Tadeusz Bossowski
- Department of Pediatrics, Endocrinology, Diabetology with Cardiology Divisions, Medical University of Bialystok, 15-274 Bialystok, Poland
| | - Iwona Flisiak
- Department of Dermatology and Venereology, Medical University of Bialystok, 15-540 Bialystok, Poland; (H.M.)
| |
Collapse
|
19
|
Son S, Xu C, Jang J, Dinh M, Skorobogatko Y, Fu H, Valentine JM, An G, Ying W, Yu RT, Downes M, Evans RM, Saltiel AR. Sympathetic activation of white adipose tissue recruits neutrophils to limit energy expenditure. RESEARCH SQUARE 2025:rs.3.rs-6414640. [PMID: 40321773 PMCID: PMC12047989 DOI: 10.21203/rs.3.rs-6414640/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 05/08/2025]
Abstract
Adipose tissue maintains energy homeostasis by storing lipids during nutrient surplus and releasing them through lipolysis in times of energy demand. While lipolysis is essential for short term metabolic adaptation, prolonged metabolic stress requires adaptive changes that preserve energy reserves. Here, we report that β-adrenergic activation of adipocytes induces a transient and depot-specific infiltration of neutrophils into white adipose tissue (WAT), particularly in lipid-rich visceral WAT. Neutrophil recruitment requires the stimulation of both lipolysis and p38 MAPK activation in adipocytes. Recruited neutrophils locally secrete IL-1β, which suppresses lipolysis and limits excessive energy expenditure. Neutrophil depletion or blockade of IL-1β production increased lipolysis, leading to reduced WAT mass upon repeated β3-adrenergic stimulation. Together, these findings reveal an unexpected role of neutrophil-derived IL-1β in preserving lipid stores during metabolic stress, highlighting a physiological function of innate immune cells in maintaining energy homeostasis.
Collapse
Affiliation(s)
- Seunghwan Son
- Division of Endocrinology and Metabolism, Department of Medicine and Pharmacology, University of California San Diego, San Diego, CA, USA
| | - Cindy Xu
- Division of Endocrinology and Metabolism, Department of Medicine and Pharmacology, University of California San Diego, San Diego, CA, USA
| | - Janice Jang
- Division of Endocrinology and Metabolism, Department of Medicine and Pharmacology, University of California San Diego, San Diego, CA, USA
| | - Maddox Dinh
- Division of Endocrinology and Metabolism, Department of Medicine and Pharmacology, University of California San Diego, San Diego, CA, USA
| | - Yuliya Skorobogatko
- Division of Endocrinology and Metabolism, Department of Medicine and Pharmacology, University of California San Diego, San Diego, CA, USA
| | - Haipeng Fu
- Division of Endocrinology and Metabolism, Department of Medicine and Pharmacology, University of California San Diego, San Diego, CA, USA
| | - Joseph M. Valentine
- Division of Endocrinology and Metabolism, Department of Medicine and Pharmacology, University of California San Diego, San Diego, CA, USA
| | - Garam An
- Division of Endocrinology and Metabolism, Department of Medicine and Pharmacology, University of California San Diego, San Diego, CA, USA
| | - Wei Ying
- Division of Endocrinology and Metabolism, Department of Medicine and Pharmacology, University of California San Diego, San Diego, CA, USA
| | - Ruth T. Yu
- Gene Expression Laboratory, Salk Institute for Biological Studies, San Diego, CA, USA
| | - Michael Downes
- Gene Expression Laboratory, Salk Institute for Biological Studies, San Diego, CA, USA
| | - Ronald M. Evans
- Gene Expression Laboratory, Salk Institute for Biological Studies, San Diego, CA, USA
| | - Alan R. Saltiel
- Division of Endocrinology and Metabolism, Department of Medicine and Pharmacology, University of California San Diego, San Diego, CA, USA
| |
Collapse
|
20
|
Li Y, Xu Y, Jin C, Qiu J, Jiao X, Pan Z, Guo Y. Salmonella-NLRP3 Inflammasome Crosstalk: Host Defense Activation Versus Bacterial Immune Evasion Strategies. J Inflamm Res 2025; 18:5133-5148. [PMID: 40255664 PMCID: PMC12009050 DOI: 10.2147/jir.s519902] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2025] [Accepted: 04/10/2025] [Indexed: 04/22/2025] Open
Abstract
The innate immune system plays a crucial role in defending against Salmonella infection. Inflammasomes are macromolecular complexes that assemble in response to the recognition of pathogen- or danger-associated molecular patterns. These complexes serve as signaling platforms for the activation of inflammatory Caspases, which subsequently triggers the maturation and secretion of the pro-inflammatory cytokines IL-1β and IL-18. This process also initiates pyroptosis, a highly inflammatory form of programmed cell death characterized by lytic cell lysis. Salmonella are intracellular pathogens that proliferate within epithelial cells and macrophages, posing a significant public health risk in both developed and developing countries. During Salmonella infection, the canonical NLRP3 and NLRC4 inflammasome, as well as non-canonical inflammasome, are activated. Unlike NLRC4 and non-canonical inflammasomes, which play crucial roles during intestinal infection phases, the role of NLRP3 inflammasome in resisting Salmonella infection demonstrates a higher degree of complexity and uncertainty. Nonetheless, the activation of NLRP3 inflammasome, along with the downstream innate and adaptive responses, form a robust host immune barrier against potential pathogens. Therefore, successful pathogens must evolve multiple mechanisms to circumvent or counteract these immune barriers. Here we review and discuss the mechanisms of NLRP3 inflammasome activation triggered by intracellular Salmonella, as well as the multiple strategies employed by Salmonella to avoid or delay NLRP3 inflammasome activation. A deeper understanding of how NLRP3 inflammasomes recognize Salmonella and how pathogens evade NLRP3 activation has the potential to facilitate the development of novel prevention and control measures for Salmonella infection.
Collapse
Affiliation(s)
- Yuxuan Li
- School of Nursing School of Public Health, Yangzhou University, Jiangsu, People’s Republic of China
| | - Ying Xu
- The Department of Economics and Management, Jiangsu College of Tourism, Jiangsu, People’s Republic of China
| | - Cheng Jin
- School of Nursing School of Public Health, Yangzhou University, Jiangsu, People’s Republic of China
| | - Jiayi Qiu
- School of Nursing School of Public Health, Yangzhou University, Jiangsu, People’s Republic of China
| | - Xinan Jiao
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou, Jiangsu, 225009, People’s Republic of China
- Jiangsu Key Laboratory of Zoonosis, Yangzhou University, Yangzhou, Jiangsu, 225009, People’s Republic of China
- Key Laboratory of Prevention and Control of Biological Hazard Factors (Animal Origin) for Agrifood Safety and Quality, Ministry of Agriculture of China, Yangzhou University, Yangzhou, Jiangsu, People’s Republic of China
- Joint International Research Laboratory of Agriculture and Agri-Product Safety of the Ministry of Education, Yangzhou University, Yangzhou, Jiangsu, People’s Republic of China
| | - Zhiming Pan
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou, Jiangsu, 225009, People’s Republic of China
- Jiangsu Key Laboratory of Zoonosis, Yangzhou University, Yangzhou, Jiangsu, 225009, People’s Republic of China
- Key Laboratory of Prevention and Control of Biological Hazard Factors (Animal Origin) for Agrifood Safety and Quality, Ministry of Agriculture of China, Yangzhou University, Yangzhou, Jiangsu, People’s Republic of China
- Joint International Research Laboratory of Agriculture and Agri-Product Safety of the Ministry of Education, Yangzhou University, Yangzhou, Jiangsu, People’s Republic of China
| | - Yaxin Guo
- School of Nursing School of Public Health, Yangzhou University, Jiangsu, People’s Republic of China
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou, Jiangsu, 225009, People’s Republic of China
- Jiangsu Key Laboratory of Zoonosis, Yangzhou University, Yangzhou, Jiangsu, 225009, People’s Republic of China
- Key Laboratory of Prevention and Control of Biological Hazard Factors (Animal Origin) for Agrifood Safety and Quality, Ministry of Agriculture of China, Yangzhou University, Yangzhou, Jiangsu, People’s Republic of China
- Joint International Research Laboratory of Agriculture and Agri-Product Safety of the Ministry of Education, Yangzhou University, Yangzhou, Jiangsu, People’s Republic of China
| |
Collapse
|
21
|
Vallazhath A, Thimmappa PY, Joshi HB, Hebbar KR, Nayak A, Umakanth S, Saoji AA, Manjunath NK, Hadapad BS, Joshi MB. A comprehensive review on the implications of Yogic/Sattvic diet in reducing inflammation in type 2 diabetes. Nutr Diabetes 2025; 15:14. [PMID: 40216734 PMCID: PMC11992243 DOI: 10.1038/s41387-025-00371-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Revised: 03/25/2025] [Accepted: 03/28/2025] [Indexed: 04/14/2025] Open
Abstract
Chronic inflammation in type 2 diabetes (T2D), characterized by constitutively activated immune cells and elevated pro-inflammatory mediators along with hyperglycaemia and increased free fatty acids and branched chain amino acid levels, significantly alters the immuno-metabolic axis. Over the years, dietary intervention has been explored as an effective strategy for managing T2D. Evidence from experimental and clinical studies indicates that various diets, including Mediterranean, Nordic, Palaeolithic and ketogenic diets, increase insulin sensitivity, decrease gluconeogenesis, and adiposity, and exert anti-inflammatory effects, thus preserving immuno-metabolic homeostasis in individuals with T2D. Indian dietary sources are categorized as Sattvic, Rajasic, and Tamasic, depending on their impact on health and behaviour. The Yogic diet, commonly recommended during yoga practice, is predominantly Sattvic, emphasizing plant-based whole foods while limiting processed and high-glycaemic-index items. Yogic diet is also recommended for Mitahara, emphasizing mindful eating, which is attributed to calorie restriction. Adopting a Yogic diet, featuring low-fat vegetarian principles, strongly reduces inflammatory mediator levels. This diet not only ameliorates insulin resistance and maintains a healthy body weight but also regulates immunomodulation, enhances gut microbiome diversity and provides essential phytonutrients, collectively preventing inflammation. Although, preliminary studies show aforementioned beneficial role of Yogic diet in improving diabetes associated metabolic and inflammatory changes, precise cellular and molecular mechanisms are not yet understood. Hence, further studies are warranted to decipher the mechanisms. This review summarizes the multiple roles of Yogic diet and related dietary components in mitigating inflammation and enhancing glycaemic control in T2D.
Collapse
Affiliation(s)
- Anupama Vallazhath
- Department of Ageing Research, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal, 576104, India
| | - Pooja Yedehalli Thimmappa
- Department of Ageing Research, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal, 576104, India
| | - Harshit B Joshi
- Division of Ayurveda, Centre for Integrative Medicine and Research, Manipal Academy of Higher Education, Manipal, 576104, India
| | - Krishna Raghava Hebbar
- Division of Ayurveda, Centre for Integrative Medicine and Research, Manipal Academy of Higher Education, Manipal, 576104, India
| | - Anupama Nayak
- Division of Ayurveda, Centre for Integrative Medicine and Research, Manipal Academy of Higher Education, Manipal, 576104, India
| | | | - Apar Avinash Saoji
- Swami Vivekananda Yoga Anusandhana Samsthana, Bangalore, 560105, Karnataka, India
| | | | - Basavaraj S Hadapad
- Division of Ayurveda, Centre for Integrative Medicine and Research, Manipal Academy of Higher Education, Manipal, 576104, India
| | - Manjunath B Joshi
- Department of Ageing Research, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal, 576104, India.
- Centre for Ayurveda Biology, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal, 576104, India.
| |
Collapse
|
22
|
Muszka Z, Jenei V, Mácsik R, Mezhonova E, Diyab S, Csősz R, Bácsi A, Mázló A, Koncz G. Life-threatening risk factors contribute to the development of diseases with the highest mortality through the induction of regulated necrotic cell death. Cell Death Dis 2025; 16:273. [PMID: 40216765 PMCID: PMC11992264 DOI: 10.1038/s41419-025-07563-7] [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: 06/14/2024] [Revised: 02/17/2025] [Accepted: 03/18/2025] [Indexed: 04/14/2025]
Abstract
Chronic diseases affecting the cardiovascular system, diabetes mellitus, neurodegenerative diseases, and various other organ-specific conditions, involve different underlying pathological processes. However, they share common risk factors that contribute to the development and progression of these diseases, including air pollution, hypertension, obesity, high cholesterol levels, smoking and alcoholism. In this review, we aim to explore the connection between four types of diseases with different etiologies and various risk factors. We highlight that the presence of risk factors induces regulated necrotic cell death, leading to the release of damage-associated molecular patterns (DAMPs), ultimately resulting in sterile inflammation. Therefore, DAMP-mediated inflammation may be the link explaining how risk factors can lead to the development and maintenance of chronic diseases. To explore these processes, we summarize the main cell death pathways activated by the most common life-threatening risk factors, the types of released DAMPs and how these events are associated with the pathophysiology of diseases with the highest mortality. Various risk factors, such as smoking, air pollution, alcoholism, hypertension, obesity, and high cholesterol levels induce regulated necrosis. Subsequently, the release of DAMPs leads to chronic inflammation, which increases the risk of many diseases, including those with the highest mortality rates.
Collapse
Affiliation(s)
- Zsuzsa Muszka
- Department of Immunology, Faculty of Medicine, University of Debrecen, Egyetem square 1, 4032, Debrecen, Hungary
- Doctoral School of Molecular Cell and Immune Biology, University of Debrecen, Egyetem square 1, 4032, Debrecen, Hungary
| | - Viktória Jenei
- Department of Immunology, Faculty of Medicine, University of Debrecen, Egyetem square 1, 4032, Debrecen, Hungary
- Gyula Petrányi Doctoral School of Allergy and Clinical Immunology, University of Debrecen, Egyetem square 1, 4032, Debrecen, Hungary
| | - Rebeka Mácsik
- Department of Immunology, Faculty of Medicine, University of Debrecen, Egyetem square 1, 4032, Debrecen, Hungary
| | - Evgeniya Mezhonova
- Department of Immunology, Faculty of Medicine, University of Debrecen, Egyetem square 1, 4032, Debrecen, Hungary
| | - Silina Diyab
- Department of Immunology, Faculty of Medicine, University of Debrecen, Egyetem square 1, 4032, Debrecen, Hungary
| | - Réka Csősz
- Department of Immunology, Faculty of Medicine, University of Debrecen, Egyetem square 1, 4032, Debrecen, Hungary
| | - Attila Bácsi
- Department of Immunology, Faculty of Medicine, University of Debrecen, Egyetem square 1, 4032, Debrecen, Hungary
| | - Anett Mázló
- Department of Immunology, Faculty of Medicine, University of Debrecen, Egyetem square 1, 4032, Debrecen, Hungary.
| | - Gábor Koncz
- Department of Immunology, Faculty of Medicine, University of Debrecen, Egyetem square 1, 4032, Debrecen, Hungary.
| |
Collapse
|
23
|
Zhang WG, Zheng XR, Yao Y, Sun WJ, Shao BZ. The role of NLRP3 inflammasome in multiple sclerosis: pathogenesis and pharmacological application. Front Immunol 2025; 16:1572140. [PMID: 40242770 PMCID: PMC11999851 DOI: 10.3389/fimmu.2025.1572140] [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: 02/20/2025] [Accepted: 03/21/2025] [Indexed: 04/18/2025] Open
Abstract
Multiple sclerosis (MS) is widely acknowledged as a chronic inflammatory autoimmune disorder characterized by central nervous system (CNS) demyelination and neurodegeneration. The hyperactivation of immune and inflammatory responses is recognized as a pivotal factor contributing to the pathogenesis and progression of MS. Among various immune and inflammatory reactions, researchers have increasingly focused on the inflammasome, a complex of proteins. The initiation and activation of the inflammasome are intricately involved in the onset of MS. Notably, the NLRP3 inflammasome, the most extensively studied member of the inflammasome complex, is closely linked with MS. This review will delve into the roles of the NLRP3 inflammasome in the pathogenesis and progression of MS. Additionally, therapeutic strategies targeting the NLRP3 inflammasome for the treatment of MS, including natural compounds, autophagy regulators, and other small molecular compounds, will be detailed in this review.
Collapse
Affiliation(s)
- Wen-Gang Zhang
- The First Medical Center, General Hospital of the Chinese People’s Liberation Army, Beijing, China
| | - Xiao-Rui Zheng
- Medical Supplies Center, General Hospital of the Chinese People’s Liberation Army, Beijing, China
| | - Yi Yao
- The First Medical Center, General Hospital of the Chinese People’s Liberation Army, Beijing, China
| | - Wei-Jia Sun
- Medical Supplies Center, General Hospital of the Chinese People’s Liberation Army, Beijing, China
| | - Bo-Zong Shao
- The First Medical Center, General Hospital of the Chinese People’s Liberation Army, Beijing, China
| |
Collapse
|
24
|
Miao Z, Zhang X, Xu Y, Liu Y, Yang Q. Unveiling the nexus: pyroptosis and its crucial implications in liver diseases. Mol Cell Biochem 2025; 480:2159-2176. [PMID: 39477911 DOI: 10.1007/s11010-024-05147-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Accepted: 10/22/2024] [Indexed: 04/02/2025]
Abstract
Pyroptosis, a distinctive form of programmed cell death orchestrated by gasdermin proteins, manifests as cellular rupture, accompanied by the release of inflammatory factors. While pyroptosis is integral to anti-infection immunity, its aberrant activation has been implicated in tumorigenesis. The liver, as the body's largest metabolic organ, is rich in various enzymes and governs metabolism. It is also the primary site for protein synthesis. Recent years have witnessed the emergence of pyroptosis as a significant player in the pathogenesis of specific liver diseases, exerting a pivotal role in both physiological and pathological processes. A comprehensive exploration of pyroptosis can unveil its contributions to the development and regression of conditions such as hepatitis, cirrhosis, and hepatocellular carcinoma, offering innovative perspectives for clinical prevention and treatment. This review consolidates current knowledge on key molecules involved in cellular pyroptosis and delineates their roles in liver diseases. Furthermore, we discuss the potential of leveraging pyroptosis as a novel or existing anti-cancer strategy.
Collapse
Affiliation(s)
- Zeyu Miao
- Department of Pathogenobiology, College of Basic Medical Sciences, Jilin University, 126 Xinmin Street, Changchun, 130021, Jilin Province, China
| | - Xiaorong Zhang
- Department of Pathogenobiology, College of Basic Medical Sciences, Jilin University, 126 Xinmin Street, Changchun, 130021, Jilin Province, China
| | - Yang Xu
- Department of Pathogenobiology, College of Basic Medical Sciences, Jilin University, 126 Xinmin Street, Changchun, 130021, Jilin Province, China
| | - Yan Liu
- Department of Pathogenobiology, College of Basic Medical Sciences, Jilin University, 126 Xinmin Street, Changchun, 130021, Jilin Province, China
| | - Qing Yang
- Department of Pathogenobiology, College of Basic Medical Sciences, Jilin University, 126 Xinmin Street, Changchun, 130021, Jilin Province, China.
| |
Collapse
|
25
|
Zhou H, Gizlenci M, Xiao Y, Martin F, Nakamori K, Zicari EM, Sato Y, Tullius SG. Obesity-associated Inflammation and Alloimmunity. Transplantation 2025; 109:588-596. [PMID: 39192462 PMCID: PMC11868468 DOI: 10.1097/tp.0000000000005183] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/29/2024]
Abstract
Obesity is a worldwide health problem with a rapidly rising incidence. In organ transplantation, increasing numbers of patients with obesity accumulate on waiting lists and undergo surgery. Obesity is in general conceptualized as a chronic inflammatory disease, potentially impacting alloimmune response and graft function. Here, we summarize our current understanding of cellular and molecular mechanisms that control obesity-associated adipose tissue inflammation and provide insights into mechanisms affecting transplant outcomes, emphasizing on the beneficial effects of weight loss on alloimmune responses.
Collapse
Affiliation(s)
- Hao Zhou
- Division of Transplant Surgery & Transplant Surgery Research Laboratory, Brigham and Women’s Hospital, Harvard Medical School, Boston, United States
| | - Merih Gizlenci
- Division of Transplant Surgery & Transplant Surgery Research Laboratory, Brigham and Women’s Hospital, Harvard Medical School, Boston, United States
- Department of General, Visceral, Cancer and Transplant Surgery, University Hospital of Cologne, Cologne, Germany
| | - Yao Xiao
- Division of Transplant Surgery & Transplant Surgery Research Laboratory, Brigham and Women’s Hospital, Harvard Medical School, Boston, United States
| | - Friederike Martin
- Division of Transplant Surgery & Transplant Surgery Research Laboratory, Brigham and Women’s Hospital, Harvard Medical School, Boston, United States
- Department of Surgery, CVK/CCM, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Keita Nakamori
- Division of Transplant Surgery & Transplant Surgery Research Laboratory, Brigham and Women’s Hospital, Harvard Medical School, Boston, United States
- Department of Urology, Osaka Medical and Pharmaceutical University, Takatsuki, Osaka, Japan
| | - Elizabeth M. Zicari
- Division of Transplant Surgery & Transplant Surgery Research Laboratory, Brigham and Women’s Hospital, Harvard Medical School, Boston, United States
- Faculté de Pharmacie, Université Paris Cité, Paris, France
| | - Yuko Sato
- Division of Transplant Surgery & Transplant Surgery Research Laboratory, Brigham and Women’s Hospital, Harvard Medical School, Boston, United States
| | - Stefan G. Tullius
- Division of Transplant Surgery & Transplant Surgery Research Laboratory, Brigham and Women’s Hospital, Harvard Medical School, Boston, United States
| |
Collapse
|
26
|
Liu C, Zhou R, Chen B, Yan X, Guo L, Tang Y, Zuo X, Guo X, Yu H, Chen J, Guo Z, Wang F, Xu C. Inflammatory microenvironment-responsive nanomicelles for acute lung injury therapy: ROS-scavenging and macrophage repolarization. Mater Today Bio 2025; 31:101622. [PMID: 40104650 PMCID: PMC11919404 DOI: 10.1016/j.mtbio.2025.101622] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2024] [Revised: 02/09/2025] [Accepted: 02/26/2025] [Indexed: 03/20/2025] Open
Abstract
The pathogenesis of acute lung injury (ALI) is characterized by an uncontrolled inflammatory response, marked by excessive production of reactive oxygen species (ROS) and the infiltration of inflammatory cells, particularly macrophages, which play a pivotal role in disease progression. The synergistic effect of ROS scavenging and macrophage repolarization provides a promising strategy for effective ALI treatment. Herein, we developed a novel type of self-assembling nanomicelles, which were composed of poly-L-glutamic acid (PLG) and 4-Hydroxymethyl phenylboronic acid (PBA). The nanomicelles (PPDex micelles) had a high drug-loading capacity for dexamethasone (Dex) based on boronic ester bonds, which exhibited reversible cleavage under inflammatory conditions characterized by elevated levels of ROS or decreased pH values. These PPDex micelles revealed rapid drug-responsive release behavior in the inflammatory environment, and in vivo studies demonstrated their efficacy in modulating cytokines, inhibiting oxidative stress, and promoting macrophage polarization from the pro-inflammatory M1 phenotype to the anti-inflammatory M2 phenotype, which ultimately suppressed the progression of ALI. Moreover, the PPDex micelles had the effective ability to effectively suppress the NF-кB and ROS/NLRP3 inflammatory pathways. Therefore, this study presented a novel and potent therapeutic strategy for ALI treatment, which could promote the clinical application of polymer nanomicelles in the treatment of ALI.
Collapse
Affiliation(s)
- Chang Liu
- College of Basic Medical Sciences, The Medical Basic Research Innovation Center of Airway Disease in North China, Key Laboratory of Pathobiology, Ministry of Education, Jilin University, Changchun, 130021, China
| | - Rui Zhou
- College of Basic Medical Sciences, The Medical Basic Research Innovation Center of Airway Disease in North China, Key Laboratory of Pathobiology, Ministry of Education, Jilin University, Changchun, 130021, China
| | - Baiqiao Chen
- College of Basic Medical Sciences, The Medical Basic Research Innovation Center of Airway Disease in North China, Key Laboratory of Pathobiology, Ministry of Education, Jilin University, Changchun, 130021, China
| | - Xinran Yan
- College of Basic Medical Sciences, The Medical Basic Research Innovation Center of Airway Disease in North China, Key Laboratory of Pathobiology, Ministry of Education, Jilin University, Changchun, 130021, China
| | - Lei Guo
- College of Basic Medical Sciences, The Medical Basic Research Innovation Center of Airway Disease in North China, Key Laboratory of Pathobiology, Ministry of Education, Jilin University, Changchun, 130021, China
| | - Yixin Tang
- College of Basic Medical Sciences, The Medical Basic Research Innovation Center of Airway Disease in North China, Key Laboratory of Pathobiology, Ministry of Education, Jilin University, Changchun, 130021, China
| | - Xu Zuo
- College of Basic Medical Sciences, The Medical Basic Research Innovation Center of Airway Disease in North China, Key Laboratory of Pathobiology, Ministry of Education, Jilin University, Changchun, 130021, China
| | - Xiaoping Guo
- College of Basic Medical Sciences, The Medical Basic Research Innovation Center of Airway Disease in North China, Key Laboratory of Pathobiology, Ministry of Education, Jilin University, Changchun, 130021, China
| | - Haiyang Yu
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, China
| | - Jie Chen
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, China
| | - Zhaopei Guo
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, China
| | - Fang Wang
- College of Basic Medical Sciences, The Medical Basic Research Innovation Center of Airway Disease in North China, Key Laboratory of Pathobiology, Ministry of Education, Jilin University, Changchun, 130021, China
| | - Caina Xu
- College of Basic Medical Sciences, The Medical Basic Research Innovation Center of Airway Disease in North China, Key Laboratory of Pathobiology, Ministry of Education, Jilin University, Changchun, 130021, China
| |
Collapse
|
27
|
Ježek P. Physiological Fatty Acid-Stimulated Insulin Secretion and Redox Signaling Versus Lipotoxicity. Antioxid Redox Signal 2025; 42:566-622. [PMID: 39834189 DOI: 10.1089/ars.2024.0799] [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] [Indexed: 01/22/2025]
Abstract
Significance: Type 2 diabetes as a world-wide epidemic is characterized by the insulin resistance concomitant to a gradual impairment of β-cell mass and function (prominently declining insulin secretion) with dysregulated fatty acids (FAs) and lipids, all involved in multiple pathological development. Recent Advances: Recently, redox signaling was recognized to be essential for insulin secretion stimulated with glucose (GSIS), branched-chain keto-acids, and FAs. FA-stimulated insulin secretion (FASIS) is a normal physiological event upon postprandial incoming chylomicrons. This contrasts with the frequent lipotoxicity observed in rodents. Critical Issues: Overfeeding causes FASIS to overlap with GSIS providing repeating hyperinsulinemia, initiates prediabetic states by lipotoxic effects and low-grade inflammation. In contrast the protective effects of lipid droplets in human β-cells counteract excessive lipids. Insulin by FASIS allows FATP1 recruitment into adipocyte plasma membranes when postprandial chylomicrons come late at already low glycemia. Future Directions: Impaired states of pancreatic β-cells and peripheral organs at prediabetes and type 2 diabetes should be revealed, including the inter-organ crosstalk by extracellular vesicles. Details of FA/lipid molecular physiology are yet to be uncovered, such as complex phenomena of FA uptake into cells, postabsorptive inactivity of G-protein-coupled receptor 40, carnitine carrier substrate specificity, the role of carnitine-O-acetyltransferase in β-cells, and lipid droplet interactions with mitochondria. Antioxid. Redox Signal. 42, 566-622.
Collapse
Affiliation(s)
- Petr Ježek
- Department of Mitochondrial Physiology, No.75, Institute of Physiology of the Czech Academy of Sciences, Prague, Czech Republic
| |
Collapse
|
28
|
Russo RC, Togbe D, Couillin I, Segueni N, Han L, Quesniaux VFJ, Stoeger T, Ryffel B. Ozone-induced lung injury and inflammation: Pathways and therapeutic targets for pulmonary diseases caused by air pollutants. ENVIRONMENT INTERNATIONAL 2025; 198:109391. [PMID: 40121788 DOI: 10.1016/j.envint.2025.109391] [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: 08/23/2024] [Revised: 03/06/2025] [Accepted: 03/15/2025] [Indexed: 03/25/2025]
Abstract
Exposure to ambient Ozone (O3) air pollution directly causes by its oxidative properties, respiratory epithelial cell injury, and cell death, which promote inflammation and hyperreactivity, posing a significant public health concern. Recent clinical and experimental studies have made strides in elucidating the mechanisms underlying O3-induced epithelial cell injury, inflammation, and airway hyperreactivity, which are discussed herein. The current data suggest that O3-induced oxidative stress is a central event-inducing oxeiptotic cell death pathway. O3-induced epithelial barrier damage and cell death, triggering the release of alarmins and damage-associated molecular patterns (DAMPs), with subsequent endogenous activation of Toll-like receptors (TLRs), DNA sensing pathways, and inflammasomes, activating interleukin-1-Myd88 inflammatory pathway with the production of a range of chemokines and cytokines. This cascade orchestrates lung tissue-resident cell activation in response to O3 in leukocyte and non-leukocyte populations, driving sterile innate immune response. Chronic inflammatory response to O3, by repeated exposures, supports a mixed phenotype combining asthma and emphysema, in which their exacerbation by other particulate pollutants potentially culminates in respiratory failure. We use data from lung single-cell transcriptomics to map genes of O3-damage sensing and signaling pathways to lung cells and thereby highlight potential hotspots of O3 responses. Deeper insights into these pathological pathways might be helpful for the identification of novel therapeutic targets and strategies.
Collapse
Affiliation(s)
- Remo C Russo
- Laboratory of Pulmonary Immunology and Mechanics, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil.
| | - Dieudonnée Togbe
- Laboratory of Immuno-Neuro Modulation, INEM, UMR7355 CNRS and University of Orleans, Orleans, France
| | - Isabelle Couillin
- Laboratory of Immuno-Neuro Modulation, INEM, UMR7355 CNRS and University of Orleans, Orleans, France
| | | | - Lianyong Han
- Institute of Lung Health and Immunity (LHI), Comprehensive Pneumology Center, Helmholtz Zentrum München, German Research Center for Environmental Health, and Member of the German Center of Lung Research (DZL), Germany
| | - Valérie F J Quesniaux
- Laboratory of Immuno-Neuro Modulation, INEM, UMR7355 CNRS and University of Orleans, Orleans, France
| | - Tobias Stoeger
- Institute of Lung Health and Immunity (LHI), Comprehensive Pneumology Center, Helmholtz Zentrum München, German Research Center for Environmental Health, and Member of the German Center of Lung Research (DZL), Germany
| | - Bernhard Ryffel
- Laboratory of Immuno-Neuro Modulation, INEM, UMR7355 CNRS and University of Orleans, Orleans, France; ArtImmune SAS, 13 Avenue Buffon, Orleans, France.
| |
Collapse
|
29
|
Wang T, Chen S, Zhou D, Hong Z. Exploring receptors for pro-resolving and non-pro-resolving mediators as therapeutic targets for sarcopenia. Metabolism 2025; 165:156148. [PMID: 39892864 DOI: 10.1016/j.metabol.2025.156148] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/26/2024] [Revised: 01/01/2025] [Accepted: 01/27/2025] [Indexed: 02/04/2025]
Abstract
Sarcopenia is defined by a reduction in both muscle strength and mass. Sarcopenia may be an inevitable component of the aging process, but it may also be accelerated by comorbidities and metabolic derangements. The underlying mechanisms contributing to these pathological changes remain poorly understood. We propose that chronic inflammation-mediated networks and metabolic defects that exacerbate muscle dysfunction are critical factors in sarcopenia and related diseases. Consequently, utilizing specialized pro-resolving mediators (SPMs) that function through specific G-protein coupled receptors (GPCRs) may offer effective therapeutic options for these disorders. However, challenges such as a limited understanding of SPM/receptor signaling pathways, rapid inactivation of SPMs, and the complexities of SPM synthesis impede their practical application. In this context, stable small-molecule SPM mimetics and receptor agonists present promising alternatives. Moreover, the aged adipose-skeletal axis may contribute to this process. Activating non-SPM GPCRs on adipocytes, immune cells, and muscle cells under conditions of systemic, chronic, low-grade inflammation (SCLGI) could help alleviate inflammation and metabolic dysfunction. Recent preclinical studies indicate that both SPM GPCRs and non-SPM GPCRs can mitigate symptoms of aging-related diseases such as obesity and diabetes, which are driven by chronic inflammation and metabolic disturbances. These findings suggest that targeting these receptors could provide a novel strategy for addressing various chronic inflammatory conditions, including sarcopenia.
Collapse
Affiliation(s)
- Tiantian Wang
- Department of Neurology, Institute of Neurology and Disease, West China Hospital of Sichuan University, Chengdu, Sichuan, China.
| | - Sihan Chen
- West China School of Nursing, Sichuan University, Chengdu, Sichuan, China
| | - Dong Zhou
- Department of Neurology, Institute of Neurology and Disease, West China Hospital of Sichuan University, Chengdu, Sichuan, China
| | - Zhen Hong
- Department of Neurology, Institute of Neurology and Disease, West China Hospital of Sichuan University, Chengdu, Sichuan, China; Institute of Brain Science and Brain-inspired Technology of West China Hospital, Sichuan University, Chengdu, Sichuan, China; Department of Neurology, Chengdu Shangjin Nanfu Hospital, Chengdu, Sichuan, China.
| |
Collapse
|
30
|
Chow SH, Jeon Y, Deo P, Yeung ATY, Hale C, Sridhar S, Abraham G, Nickson J, Olivier FAB, Jiang JH, Ding Y, Han ML, Le Brun AP, Anderson D, Creek D, Tong J, Gabriel K, Li J, Traven A, Dougan G, Shen HH, Naderer T. Staphylococcal toxin PVL ruptures model membranes under acidic conditions through interactions with cardiolipin and phosphatidic acid. PLoS Biol 2025; 23:e3003080. [PMID: 40233125 PMCID: PMC12052211 DOI: 10.1371/journal.pbio.3003080] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2024] [Revised: 05/05/2025] [Accepted: 02/24/2025] [Indexed: 04/17/2025] Open
Abstract
Panton-Valentine leukocidin (PVL) is a pore-forming toxin secreted by Staphylococcus aureus strains that cause severe infections. Bicomponent PVL kills phagocytes depending on cell surface receptors, such as complement 5a receptor 1 (C5aR1). How the PVL-receptor interaction enables assembly of the leukocidin complex, targeting of membranes, and insertion of a pore channel remains incompletely understood. Here, we demonstrate that PVL binds the anionic phospholipids, phosphatidic acid, and cardiolipin, under acidic conditions and targets lipid bilayers that mimic lysosomal and mitochondrial membranes, but not the plasma membrane. The PVL-lipid interaction was sufficient to enable leukocidin complex formation as determined by neutron reflectometry and the rupture of model membranes, independent of protein receptors. In phagocytes, PVL and its C5aR1 receptor were internalized depending on sphingomyelin and cholesterol, which were dispensable for the interaction of the toxin with the plasma membrane. Internalized PVL compromised the integrity of lysosomes and mitochondria before plasma membrane rupture. Preventing the acidification of organelles or the genetic loss of PVL impaired the escape of intracellular S. aureus from macrophages. Together, the findings advance our understanding of how an S. aureus toxin kills host cells and provide key insights into how leukocidins target membranes.
Collapse
Affiliation(s)
- Seong H. Chow
- Department of Biochemistry and Molecular Biology, Infection Program, Biomedicine Discovery Institute, Monash University, Clayton, Australia
- Centre to Impact AMR, Monash University, Clayton, Australia
| | - Yusun Jeon
- Department of Biochemistry and Molecular Biology, Infection Program, Biomedicine Discovery Institute, Monash University, Clayton, Australia
- Centre to Impact AMR, Monash University, Clayton, Australia
| | - Pankaj Deo
- Department of Biochemistry and Molecular Biology, Infection Program, Biomedicine Discovery Institute, Monash University, Clayton, Australia
- Centre to Impact AMR, Monash University, Clayton, Australia
| | - Amy T. Y. Yeung
- The Wellcome Sanger Institute, Wellcome Trust Genome Campus, Cambridge, United Kingdom
| | - Christine Hale
- The Wellcome Sanger Institute, Wellcome Trust Genome Campus, Cambridge, United Kingdom
- Department of Medicine, Addenbrookes Hospital, Cambridge, United Kingdom
| | - Sushmita Sridhar
- The Wellcome Sanger Institute, Wellcome Trust Genome Campus, Cambridge, United Kingdom
- Department of Medicine, Addenbrookes Hospital, Cambridge, United Kingdom
| | - Gilu Abraham
- Department of Biochemistry and Molecular Biology, Infection Program, Biomedicine Discovery Institute, Monash University, Clayton, Australia
- Centre to Impact AMR, Monash University, Clayton, Australia
| | - Joshua Nickson
- Department of Biochemistry and Molecular Biology, Infection Program, Biomedicine Discovery Institute, Monash University, Clayton, Australia
- Centre to Impact AMR, Monash University, Clayton, Australia
| | - Françios A. B. Olivier
- Department of Biochemistry and Molecular Biology, Infection Program, Biomedicine Discovery Institute, Monash University, Clayton, Australia
- Centre to Impact AMR, Monash University, Clayton, Australia
| | - Jhih-Hang Jiang
- Department of Microbiology, Infection Program, Biomedicine Discovery Institute, Monash University, Clayton, Australia
- Department of Infectious Diseases, Alfred Hospital and Central Clinical School, Monash University, Melbourne, Australia
| | - Yue Ding
- Department of Biochemistry and Molecular Biology, Infection Program, Biomedicine Discovery Institute, Monash University, Clayton, Australia
- Department of Materials Science and Engineering, Faculty of Engineering, Monash University, Clayton, Australia
| | - Mei-Ling Han
- Department of Microbiology, Infection Program, Biomedicine Discovery Institute, Monash University, Clayton, Australia
| | - Anton P. Le Brun
- Australian Centre for Neutron Scattering, Australian Nuclear Science and Technology Organisation, Kirrawee DC, Australia
| | - Dovile Anderson
- Drug Delivery Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Australia
| | - Darren Creek
- Drug Delivery Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Australia
| | - Janette Tong
- Department of Biochemistry and Molecular Biology, Infection Program, Biomedicine Discovery Institute, Monash University, Clayton, Australia
| | - Kip Gabriel
- Department of Biochemistry and Molecular Biology, Infection Program, Biomedicine Discovery Institute, Monash University, Clayton, Australia
| | - Jian Li
- Centre to Impact AMR, Monash University, Clayton, Australia
- Department of Microbiology, Infection Program, Biomedicine Discovery Institute, Monash University, Clayton, Australia
| | - Ana Traven
- Department of Biochemistry and Molecular Biology, Infection Program, Biomedicine Discovery Institute, Monash University, Clayton, Australia
- Centre to Impact AMR, Monash University, Clayton, Australia
| | - Gordon Dougan
- The Wellcome Sanger Institute, Wellcome Trust Genome Campus, Cambridge, United Kingdom
- Department of Medicine, Addenbrookes Hospital, Cambridge, United Kingdom
| | - Hsin-Hui Shen
- Department of Biochemistry and Molecular Biology, Infection Program, Biomedicine Discovery Institute, Monash University, Clayton, Australia
- Department of Materials Science and Engineering, Faculty of Engineering, Monash University, Clayton, Australia
| | - Thomas Naderer
- Department of Biochemistry and Molecular Biology, Infection Program, Biomedicine Discovery Institute, Monash University, Clayton, Australia
- Centre to Impact AMR, Monash University, Clayton, Australia
| |
Collapse
|
31
|
Luong TVT, Yang S, Kim J. Lipotoxicity as a therapeutic target in the type 2 diabetic heart. J Mol Cell Cardiol 2025; 201:105-121. [PMID: 40020774 DOI: 10.1016/j.yjmcc.2025.02.010] [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: 08/28/2024] [Revised: 01/07/2025] [Accepted: 02/24/2025] [Indexed: 03/03/2025]
Abstract
Cardiac lipotoxicity, characterized by excessive lipid accumulation in the cardiac tissue, is a critical contributor to the pathogenesis of diabetic heart. Recent research has highlighted the key mechanisms underlying lipotoxicity, including mitochondrial dysfunction, endoplasmic reticulum stress, inflammation, and cell apoptosis, which ultimately impair the cardiac function. Various therapeutic interventions have been developed to target these pathways, mitigate lipotoxicity, and improve cardiovascular outcomes in diabetic patients. Given the global escalation in the prevalence of diabetes and the urgent demand for effective therapeutic approaches, this review focuses on how targeting cardiac lipotoxicity may be a promising avenue for treating diabetes.
Collapse
Affiliation(s)
- Trang Van T Luong
- Division of Endocrinology and Metabolism, Department of Internal Medicine, College of Medicine, Chung-Ang University, Seoul, Republic of Korea
| | - Seonbu Yang
- Division of Endocrinology and Metabolism, Department of Internal Medicine, College of Medicine, Chung-Ang University, Seoul, Republic of Korea
| | - Jaetaek Kim
- Division of Endocrinology and Metabolism, Department of Internal Medicine, College of Medicine, Chung-Ang University, Seoul, Republic of Korea.
| |
Collapse
|
32
|
Huang X, Chen Q, Su Q, Gong J, Wu L, Xiang L, Li W, Chen J, Zhao H, Huang W, Du S, Ye W. The Mediation Role of Insulin Resistance and Chronic Systemic Inflammation in the Association Between Obesity and NAFLD: Two Cross-Sectional and a Mendelian Randomization Study. Clin Epidemiol 2025; 17:287-302. [PMID: 40160337 PMCID: PMC11952067 DOI: 10.2147/clep.s508514] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2024] [Accepted: 03/11/2025] [Indexed: 04/02/2025] Open
Abstract
Purpose We aimed to identify the association between obesity and nonalcoholic fatty liver disease (NAFLD) and to quantify the mediating effects of insulin resistance (IR) and chronic inflammation through observational studies and Mendelian randomization (MR). Patients and Methods In the current study, three IR-related indicators and three indicators of inflammation were included. The individual and combined mediated effects of IR and inflammation in the association between obesity and NAFLD were investigated in two cross-sectional studies, the Fuqing Cohort from China and the National Health and Nutrition Examination Survey (NHANES). Total, direct, and indirect effects were estimated through direct counterfactual imputation estimation, and the proportion of mediating effects was calculated. We applied a two-step MR to determine the causal mediating role of IR and chronic inflammation in the pathway between obesity and NAFLD by using single nucleotide polymorphisms as instrumental variables to predict obesity, IR, and inflammation genetically. Results In the Fuqing Cohort, all obese phenotypes were associated with an elevated NAFLD risk. Moreover, indicators of IR such as homeostatic model assessment of insulin resistance (HOMA-IR) and indicators of inflammation such as C-reactive protein (CRP) were significantly and positively associated with NAFLD risk. Individuals with obesity had significantly higher levels of IR and inflammation indicators compared to non-obese individuals. The indirect proportions of insulin and HOMA-IR accounted for 50.97-66.72% in the associations between obese phenotypes and NAFLD risk, while the proportions of inflammation indicators were < 14%. Similar results were observed in the NHANES analysis. In the MR analysis, the indirect effects of HOMA-IR and CRP were statistically significant with a greater mediated proportion explained by HOMA-IR than CRP. Conclusion Through two population-based studies and MR, we found the causal mediation roles of IR and inflammation in the association between obesity and NAFLD, in which HOMA-IR and CRP showed stable, significant mediation effects. Furthermore, HOMA-IR showed a higher mediation effect than CRP. We emphasize the vital role of HOMA-IR in NAFLD monitoring.
Collapse
Affiliation(s)
- Xiaoyin Huang
- Department of Epidemiology and Health Statistics, Fujian Medical University, Fuzhou, People’s Republic of China
| | - Qianni Chen
- Department of Ultrasonography, Fuqing City Hospital Affiliated to Fujian Medical University, Fuqing, People’s Republic of China
| | - Qingling Su
- Department of Epidemiology and Health Statistics, Fujian Medical University, Fuzhou, People’s Republic of China
| | - Jiamin Gong
- Department of Epidemiology and Health Statistics, Fujian Medical University, Fuzhou, People’s Republic of China
| | - Liqin Wu
- Department of Ultrasonography, Fuqing City Hospital Affiliated to Fujian Medical University, Fuqing, People’s Republic of China
| | - Liangguang Xiang
- Department of General Surgery, Fuqing City Hospital Affiliated to Fujian Medical University, Fuqing, People’s Republic of China
| | - Wanxin Li
- Department of Epidemiology and Health Statistics, Fujian Medical University, Fuzhou, People’s Republic of China
| | - Jun Chen
- Department of Epidemiology and Health Statistics, Fujian Medical University, Fuzhou, People’s Republic of China
| | - Hongwei Zhao
- Division of Occupational and Environmental Health, University of Utah, Salt Lake City, UT, USA
| | - Wuqing Huang
- Department of Epidemiology and Health Statistics, Fujian Medical University, Fuzhou, People’s Republic of China
| | - Shanshan Du
- Institute of Population Medicine, Fujian Medical University, Fuzhou, People’s Republic of China
| | - Weimin Ye
- Department of Epidemiology and Health Statistics, Fujian Medical University, Fuzhou, People’s Republic of China
- Institute of Population Medicine, Fujian Medical University, Fuzhou, People’s Republic of China
- Department of Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
33
|
Shen DM, Byth KF, Bertheloot D, Braams S, Bradley S, Dean D, Dekker C, El-Kattan AF, Franchi L, Glick GD, Ghosh S, Hinniger A, Katz JD, Kitanovic A, Lu X, Olhava EJ, Opipari AW, Sanchez B, Seidel HM, Stunden J, Stutz A, Telling A, Venkatraman S, Winkler DG, Roush WR. Discovery of DFV890, a Potent Sulfonimidamide-Containing NLRP3 Inflammasome Inhibitor. J Med Chem 2025; 68:5529-5550. [PMID: 40036600 DOI: 10.1021/acs.jmedchem.4c02759] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/06/2025]
Abstract
The discovery of DFV890 ((R)-1), a potent and selective NLRP3 antagonist, is described. Replacement of the sulfonyl urea core from the first-generation NLRP3 antagonist CRID3 with a sulfonimidamide core afforded a novel and potent series of NLRP3 antagonists. The (R)-enantiomers of the sulfonimidamide series were found to be consistently more potent than structurally related sulfonyl ureas. Replacement of the furan unit of CRID3 with a 5-substituted thiazole unit led to DFV890 ((R)-1), which potently inhibited IL-1β production in THP-1 cells and in primary human cells, blocked multiple downstream effectors of NLRP3 activation, and substantially improved PK properties and significantly lowered the predicted human dose compared to that for CRID3. DFV890 ((R)-1) was also effective in an air pouch model of gout.
Collapse
Affiliation(s)
- Dong-Ming Shen
- IFM Therapeutics, Boston, Massachusetts 02116, United States
| | - Kate F Byth
- IFM Therapeutics, Boston, Massachusetts 02116, United States
| | | | | | - Sarah Bradley
- IFM Therapeutics, Boston, Massachusetts 02116, United States
| | - Dennis Dean
- IFM Therapeutics, Boston, Massachusetts 02116, United States
| | - Carien Dekker
- Novartis Biomedical Research, Basel CH-4002, Switzerland
| | | | - Luigi Franchi
- IFM Therapeutics, Boston, Massachusetts 02116, United States
| | - Gary D Glick
- IFM Therapeutics, Boston, Massachusetts 02116, United States
| | - Shomir Ghosh
- IFM Therapeutics, Boston, Massachusetts 02116, United States
| | | | - Jason D Katz
- IFM Therapeutics, Boston, Massachusetts 02116, United States
| | | | - Xiaokang Lu
- IFM Therapeutics, Ann Arbor, Michigan 48108, United States
| | - Edward J Olhava
- IFM Therapeutics, Boston, Massachusetts 02116, United States
| | | | - Brian Sanchez
- IFM Therapeutics, Ann Arbor, Michigan 48108, United States
| | - H Martin Seidel
- IFM Therapeutics, Boston, Massachusetts 02116, United States
| | | | | | - Alissa Telling
- IFM Therapeutics, Ann Arbor, Michigan 48108, United States
| | | | - David G Winkler
- IFM Therapeutics, Boston, Massachusetts 02116, United States
| | - William R Roush
- IFM Therapeutics, Boston, Massachusetts 02116, United States
| |
Collapse
|
34
|
Wu X, Chen H, Tian Y, Wang H, Hou H, Hu Q, Wang C. Amelioration of obesity-associated disorders using solanesol with the mitigation of NLRP3 inflammasome activation and macrophage inflammation in adipose tissue. Food Funct 2025; 16:1903-1918. [PMID: 39935386 DOI: 10.1039/d4fo05586a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/13/2025]
Abstract
Obesity and obesity-related metabolic diseases are causally linked to inflammatory activation. Proinflammatory macrophage infiltration and NOD-like receptor pyrin domain containing 3 (NLRP3) inflammasome activation contribute to chronic inflammation and insulin resistance. Alleviating inflammatory responses is a reliable method to restore insulin sensitivity and reduce the severity of metabolic syndrome. Solanesol, rich in anti-inflammatory foods (potato, tomato, eggplant, chili peppers), has demonstrated anti-inflammatory properties, but whether it plays a beneficial role in obesity-induced chronic inflammation remains poorly understood. In this study, we investigated the effects of solanesol on the NLRP3 inflammasome and inflammatory responses both in vitro and in high-fat diet (HFD)-fed mice. We found that oral administration of solanesol reduced weight gain, insulin resistance, and inflammation in epididymal white adipose tissue (eWAT) in both HFD-fed obese mice and mice concurrently treated with a HFD. This effect was involved with reducing macrophage inflammation and inactivating the NLRP3 inflammasome by reducing the K+ efflux and reactive oxygen species (ROS) production in macrophages. Solanesol also reprogrammed the phenotype of inflammatory macrophages. Taken together, our study suggests that solanesol may be a promising candidate for treating obesity and obesity-related metabolic diseases.
Collapse
Affiliation(s)
- Xiaqing Wu
- The Center for Biomedical Research, NHC Key Laboratory of Respiratory Diseases, Department of Respiratory and Critical Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Sciences and Technology, China.
- China National Tobacco Quality Supervision & Test Center, Key Laboratory of Tobacco Biological Effects, Zhengzhou, China.
- Beijing Life Science Academy, Beijing, China
| | - Huan Chen
- China National Tobacco Quality Supervision & Test Center, Key Laboratory of Tobacco Biological Effects, Zhengzhou, China.
- Beijing Life Science Academy, Beijing, China
| | - Yushan Tian
- China National Tobacco Quality Supervision & Test Center, Key Laboratory of Tobacco Biological Effects, Zhengzhou, China.
- Beijing Life Science Academy, Beijing, China
| | - Hongjuan Wang
- China National Tobacco Quality Supervision & Test Center, Key Laboratory of Tobacco Biological Effects, Zhengzhou, China.
- Beijing Life Science Academy, Beijing, China
| | - Hongwei Hou
- China National Tobacco Quality Supervision & Test Center, Key Laboratory of Tobacco Biological Effects, Zhengzhou, China.
- Beijing Life Science Academy, Beijing, China
| | - Qingyuan Hu
- China National Tobacco Quality Supervision & Test Center, Key Laboratory of Tobacco Biological Effects, Zhengzhou, China.
- Beijing Life Science Academy, Beijing, China
| | - Congyi Wang
- The Center for Biomedical Research, NHC Key Laboratory of Respiratory Diseases, Department of Respiratory and Critical Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Sciences and Technology, China.
| |
Collapse
|
35
|
Wang H, Ma L, Su W, Liu Y, Xie N, Liu J. NLRP3 inflammasome in health and disease (Review). Int J Mol Med 2025; 55:48. [PMID: 39930811 PMCID: PMC11781521 DOI: 10.3892/ijmm.2025.5489] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2024] [Accepted: 12/16/2024] [Indexed: 02/13/2025] Open
Abstract
Activation of inflammasomes is the activation of inflammation‑related caspase mediated by the assembly signal of multi‑protein complex and the maturity of inflammatory factors, such as IL‑1β and IL‑18. Among them, the Nod‑like receptor family pyrin domain containing 3 (NLRP3) inflammasome is the most thoroughly studied type of inflammatory corpuscle at present, which is involved in the occurrence and development of numerous human diseases. Therefore, targeting the NLRP3 inflammasome has become the focus of drug development for related diseases. In this paper, the research progress of the NLRP3 inflammasome in recent years is summarized, including the activation and regulation of NLRP3 and its association with diseases. A deep understanding of the regulatory mechanism of NLRP3 will be helpful to the discovery of new drug targets and the development of therapeutic drugs.
Collapse
Affiliation(s)
- Haoran Wang
- Department of Orthopaedics, Tongji Hospital Affiliated to Tongji University, Shanghai 200065, P.R. China
| | - Li Ma
- Department of Oncology, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210011, P.R. China
| | - Weiran Su
- Department of Internal Medicine, Jiading District Central Hospital, Shanghai 201800, P.R. China
| | - Yangruoyu Liu
- Department of Orthopaedics, Tongji Hospital Affiliated to Tongji University, Shanghai 200065, P.R. China
| | - Ning Xie
- Department of Orthopaedics, Tongji Hospital Affiliated to Tongji University, Shanghai 200065, P.R. China
| | - Jun Liu
- Department of Orthopaedics, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210011, P.R. China
| |
Collapse
|
36
|
Kielbowski K, Bratborska AW, Bakinowska E, Pawlik A. Sirtuins as therapeutic targets in diabetes. Expert Opin Ther Targets 2025; 29:117-135. [PMID: 40116767 DOI: 10.1080/14728222.2025.2482563] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Revised: 03/01/2025] [Accepted: 03/18/2025] [Indexed: 03/23/2025]
Abstract
INTRODUCTION Sirtuins (SIRTs) are NAD+-dependent deacetylases that mediate post-translational modifications of proteins. Seven members of the SIRT family have been identified in mammals. Importantly, SIRTs interact with numerous metabolic and inflammatory pathways. Thus, researchers have investigated their role in metabolic and inflammatory disorders. AREAS COVERED In this review, we comprehensively discuss the involvement of SIRTs in the processes of pancreatic β-cell dysfunction, glucose tolerance, insulin secretion, lipid metabolism, and adipocyte functions. In addition, we describe the current evidence regarding modulation of the expression and activity of SIRTs in diabetes, diabetic complications, and obesity. EXPERT OPINION The development of specific SIRT activators and inhibitors that exhibit high selectivity toward specific SIRT isoforms remains a major challenge. This involves the need to elucidate the physiological pathways involving SIRTs, as well as their important role in the development of metabolic disorders. Molecular modeling techniques will be helpful to develop new compounds that modulate the activity of SIRTs, which may contribute to the preparation of new drugs that selectively target specific SIRTs. SIRTs hold promise as potential targets in metabolic disease, but there is much to learn about specific modulators and the final answers will await clinical trials.
Collapse
Affiliation(s)
- Kajetan Kielbowski
- Department of Physiology, Pomeranian Medical University, Szczecin, Poland
| | | | - Estera Bakinowska
- Department of Physiology, Pomeranian Medical University, Szczecin, Poland
| | - Andrzej Pawlik
- Department of Physiology, Pomeranian Medical University, Szczecin, Poland
| |
Collapse
|
37
|
Esmaeili Z, Shavali Gilani P, Khosravani M, Motamedi M, Maleknejad S, Adabi M, Sadighara P. Nanotechnology-driven EGCG: bridging antioxidant and therapeutic roles in metabolic and cancer pathways. Nanomedicine (Lond) 2025; 20:621-636. [PMID: 39924937 PMCID: PMC11881875 DOI: 10.1080/17435889.2025.2462521] [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: 10/28/2024] [Accepted: 01/31/2025] [Indexed: 02/11/2025] Open
Abstract
Epigallocatechin-3-gallate (EGCG), the primary polyphenol in green tea, is renowned for its potent antioxidant properties. EGCG interacts with various cellular targets, inhibiting cancer cell proliferation through apoptosis and cell cycle arrest induction, while also modulating metabolic pathways. Studies have demonstrated its potential in addressing cancer development, obesity, and diabetes. Given the rising prevalence of metabolic diseases and cancers, EGCG is increasingly recognized as a promising therapeutic agent. This review provides a comprehensive overview of the latest findings on the effects of both free and nano-encapsulated EGCG on mechanisms involved in the management and prevention of hyperlipidemia, diabetes, and gastrointestinal (GI) cancers. The review highlights EGCG role in modulating key signaling pathways, enhancing bioavailability through nano-formulations, and its potential applications in clinical settings.
Collapse
Affiliation(s)
- Zahra Esmaeili
- Department of Medical Nanotechnology, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Parisa Shavali Gilani
- Department of Environmental Health Engineering, Division of Food Safety and Hygiene, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Masood Khosravani
- Department of Medical Nanotechnology, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Maral Motamedi
- Department of Medical Nanotechnology, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Shokofeh Maleknejad
- Department of Food Hygiene and Quality Control, Faculty of Veterinary Medicine, Shahid Chamran University of Ahvaz, Ahvaz, Iran
| | - Mahdi Adabi
- Department of Medical Nanotechnology, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
- Food Microbiology Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Parisa Sadighara
- Department of Environmental Health Engineering, Division of Food Safety and Hygiene, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
38
|
Ji Q, Meng Y, Han X, Yi C, Chen X, Zhan Y. Bioinformatic Insights and XGBoost Identify Shared Genetics in Chronic Obstructive Pulmonary Disease and Type 2 Diabetes. THE CLINICAL RESPIRATORY JOURNAL 2025; 19:e70057. [PMID: 40045538 PMCID: PMC11882755 DOI: 10.1111/crj.70057] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Revised: 08/15/2024] [Accepted: 01/30/2025] [Indexed: 03/09/2025]
Abstract
BACKGROUND The correlation between chronic obstructive pulmonary disease (COPD) and Type 2 diabetes mellitus (T2DM) has long been recognized, but their shared molecular underpinnings remain elusive. This study aims to uncover common genetic markers and pathways in COPD and T2DM, providing insights into their molecular crosstalk. METHODS Utilizing the Gene Expression Omnibus (GEO) database, we analyzed gene expression datasets from six COPD and five T2DM studies. A multifaceted bioinformatics approach, encompassing the limma R package, unified matrix analysis, and weighted gene co-expression network analysis (WGCNA), was deployed to identify differentially expressed genes (DEGs) and hub genes. Functional enrichment and protein-protein interaction (PPI) analyses were conducted, followed by cross-species validation in Mus musculus models. Machine learning techniques, including random forest and LASSO regression, were applied for further validation, culminating in the development of a prognostic model using XGBoost. RESULTS Our analysis revealed shared DEGs such as KIF1C, CSTA, GMNN, and PHGDH in both COPD and T2DM. Cross-species comparison identified common genes including PON1 and CD14, exhibiting varying expression patterns. The random forest and LASSO regression identified six critical genes, with our XGBoost model demonstrating significant predictive accuracy (AUC = 0.996 for COPD). CONCLUSIONS This study identifies key genetic markers shared between COPD and T2DM, providing new insights into their molecular pathways. Our XGBoost model exhibited high predictive accuracy for COPD, highlighting the potential utility of these markers. These findings offer promising biomarkers for early detection and enhance our understanding of the diseases' interplay. Further validation in larger cohorts is recommended.
Collapse
Affiliation(s)
- Qianqian Ji
- Department of Epidemiology, School of Public Health (Shenzhen)Sun Yat‐Sen UniversityShenzhenGuangdongChina
| | - Yaxian Meng
- Department of Epidemiology, School of Public Health (Shenzhen)Sun Yat‐Sen UniversityShenzhenGuangdongChina
| | - Xiaojie Han
- Department of Chronic Disease ControlGuangming Center for Disease Control and PreventionShenzhenGuangdongChina
| | - Chao Yi
- Department of Chronic Disease ControlGuangming Center for Disease Control and PreventionShenzhenGuangdongChina
| | - Xiaoliang Chen
- Department of Chronic Disease ControlGuangming Center for Disease Control and PreventionShenzhenGuangdongChina
| | - Yiqiang Zhan
- Department of Epidemiology, School of Public Health (Shenzhen)Sun Yat‐Sen UniversityShenzhenGuangdongChina
- Guangdong Engineering Technology Research Center of Nutrition TransformationSun Yat‐sen UniversityShenzhenGuangdongChina
- Institute of Environmental MedicineKarolinska InstitutetStockholmSweden
| |
Collapse
|
39
|
Radmehr E, Yazdanpanah N, Rezaei N. Non-coding RNAs affecting NLRP3 inflammasome pathway in diabetic cardiomyopathy: a comprehensive review of potential therapeutic options. J Transl Med 2025; 23:249. [PMID: 40022088 PMCID: PMC11871836 DOI: 10.1186/s12967-025-06269-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2025] [Accepted: 02/15/2025] [Indexed: 03/03/2025] Open
Abstract
Cardiomyopathies are a heterogeneous group of disorders that can lead to fulminant heart failure and sudden cardiac death. In recent years, the prevalence of all types of cardiomyopathies has shown an upward trend globally. Up to 40% of patients with cardiomyopathy-related heart failure have diabetes mellitus (DM). With the fast global spread of DM, the prevalence of DCM is increasing accordingly and it remains the leading cause of morbidity and mortality in chronic diabetic patients. NLRP3 inflammasome significantly contributes to the development and pathological progression of DCM. Targeting the inflammasome or any of the mediators along its activation pathway provides new potential therapeutic targets for developing specialized drugs to treat DCM.In this comprehensive review, we sought to introduce and summarize the non-coding RNAs with potential therapeutic effects targeting NLRP3 inflammasome signaling in DCM. We hope this general overview can aid future research in developing new therapies for DCM.
Collapse
Affiliation(s)
- Elahe Radmehr
- Colorectal Research Center, Imam Khomeini Hospital Complex, Tehran University of Medical Sciences (TUMS), Tehran, Iran
- Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Niloufar Yazdanpanah
- Research Center for Immunodeficiencies, Children's Medical Center Hospital, Tehran University of Medical Sciences, Dr. Qarib St, Keshavarz Blvd, Tehran, 14194, Iran
- Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), Tehran, Iran
- School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Nima Rezaei
- Research Center for Immunodeficiencies, Children's Medical Center Hospital, Tehran University of Medical Sciences, Dr. Qarib St, Keshavarz Blvd, Tehran, 14194, Iran.
- Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), Tehran, Iran.
- Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
40
|
Penna C, Pagliaro P. Endothelial Dysfunction: Redox Imbalance, NLRP3 Inflammasome, and Inflammatory Responses in Cardiovascular Diseases. Antioxidants (Basel) 2025; 14:256. [PMID: 40227195 PMCID: PMC11939635 DOI: 10.3390/antiox14030256] [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: 01/26/2025] [Revised: 02/17/2025] [Accepted: 02/20/2025] [Indexed: 04/15/2025] Open
Abstract
Endothelial dysfunction (ED) is characterized by an imbalance between vasodilatory and vasoconstrictive factors, leading to impaired vascular tone, thrombosis, and inflammation. These processes are critical in the development of cardiovascular diseases (CVDs) such as atherosclerosis, hypertension and ischemia/reperfusion injury (IRI). Reduced nitric oxide (NO) production and increased oxidative stress are key contributors to ED. Aging further exacerbates ED through mitochondrial dysfunction and increased oxidative/nitrosative stress, heightening CVD risk. Antioxidant systems like superoxide-dismutase (SOD), glutathione-peroxidase (GPx), and thioredoxin/thioredoxin-reductase (Trx/TXNRD) pathways protect against oxidative stress. However, their reduced activity promotes ED, atherosclerosis, and vulnerability to IRI. Metabolic syndrome, comprising insulin resistance, obesity, and hypertension, is often accompanied by ED. Specifically, hyperglycemia worsens endothelial damage by promoting oxidative stress and inflammation. Obesity leads to chronic inflammation and changes in perivascular adipose tissue, while hypertension is associated with an increase in oxidative stress. The NLRP3 inflammasome plays a significant role in ED, being triggered by factors such as reactive oxygen and nitrogen species, ischemia, and high glucose, which contribute to inflammation, endothelial injury, and exacerbation of IRI. Treatments, such as N-acetyl-L-cysteine, SGLT2 or NLRP3 inhibitors, show promise in improving endothelial function. Yet the complexity of ED suggests that multi-targeted therapies addressing oxidative stress, inflammation, and metabolic disturbances are essential for managing CVDs associated with metabolic syndrome.
Collapse
Affiliation(s)
- Claudia Penna
- Department of Clinical and Biological Sciences, University of Turin, 10043 Orbassano, Italy;
- National Institute for Cardiovascular Research (INRC), 40126 Bologna, Italy
| | - Pasquale Pagliaro
- Department of Clinical and Biological Sciences, University of Turin, 10043 Orbassano, Italy;
- National Institute for Cardiovascular Research (INRC), 40126 Bologna, Italy
| |
Collapse
|
41
|
Zhao J, Li X, Liang C, Yan Y. Can Exercise-Mediated Adipose Browning Provide an Alternative Explanation for the Obesity Paradox? Int J Mol Sci 2025; 26:1790. [PMID: 40076419 PMCID: PMC11898606 DOI: 10.3390/ijms26051790] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2024] [Revised: 02/06/2025] [Accepted: 02/14/2025] [Indexed: 03/14/2025] Open
Abstract
Overweight patients with cardiovascular disease (CVD) tend to survive longer than normal-weight patients, a phenomenon known as the "obesity paradox". The phenotypic characteristics of adipose distribution in these patients (who survive longer) often reveal a larger proportion of subcutaneous white adipose tissue (scWAT), suggesting that the presence of scWAT is negatively associated with all-cause mortality and that scWAT appears to provide protective benefits in patients facing unhealthy states. Exercise-mediated browning is a crucial aspect of the benign remodeling process of adipose tissue (AT). Reduced accumulation, reduced inflammation, and associated adipokine secretion are directly related to the reduction in CVD mortality. This paper summarized the pathogenetic factors associated with AT accumulation in patients with CVD and analyzed the possible role and pathway of exercise-mediated adipose browning in reducing the risk of CVD and CVD-related mortality. It is suggested that exercise-mediated browning may provide a new perspective on the "obesity paradox"; that is, overweight CVD patients who have more scWAT may gain greater cardiovascular health benefits through exercise.
Collapse
Affiliation(s)
- Jiani Zhao
- Department of Sport Biochemistry, School of Sport Science, Beijing Sport University (BSU), Beijing 100084, China; (J.Z.); (X.L.)
| | - Xuehan Li
- Department of Sport Biochemistry, School of Sport Science, Beijing Sport University (BSU), Beijing 100084, China; (J.Z.); (X.L.)
| | - Chunyu Liang
- School of Physical Education, Guangxi University (GXU), Nanning 530004, China
| | - Yi Yan
- Department of Sport Biochemistry, School of Sport Science, Beijing Sport University (BSU), Beijing 100084, China; (J.Z.); (X.L.)
- Laboratory of Sports Stress and Adaptation of General Administration of Sport, Beijing Sport University (BSU), Beijing 100084, China
- Exercise and Physical Fitness, Beijing Sport University (BSU), Beijing 100084, China
| |
Collapse
|
42
|
Huemer MT, Spagnuolo MC, Maalmi H, Wagner R, Bönhof GJ, Heier M, Koenig W, Rathmann W, Prystupa K, Nano J, Ziegler D, Peters A, Roden M, Thorand B, Herder C. Phenotype-based clusters, inflammation and cardiometabolic complications in older people before the diagnosis of type 2 diabetes: KORA F4/FF4 cohort study. Cardiovasc Diabetol 2025; 24:83. [PMID: 39972466 PMCID: PMC11841139 DOI: 10.1186/s12933-025-02617-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/09/2024] [Accepted: 01/27/2025] [Indexed: 02/21/2025] Open
Abstract
BACKGROUND Using a data-driven approach, six clusters with different risk profiles and burden of complications were recently identified in middle-aged people before the diagnosis of type 2 diabetes (T2D). We aimed to investigate whether these clusters could be generalised to older people and if subclinical inflammation was related to their cardiometabolic risk profiles. METHODS We assigned 843 participants of the KORA F4 study aged 61-82 years without T2D to the six previously defined phenotype-based clusters. Based on 73 biomarkers of subclinical inflammation, we derived an inflammation-related score ("inflammatory load") using principal component analysis to assess subclinical inflammation. Risk factors, inflammatory load as well as prevalence and incidence of (pre)diabetes-related complications were compared between the clusters using pairwise comparisons and regression analyses. RESULTS Clusters 1 and 2 had the lowest cardiometabolic risk, whereas clusters 5 and 6 the highest. T2D risk was highest in clusters 3, 4, 5, and 6 compared with the low-risk cluster 2 (age- and sex-adjusted ORs between 3.6 and 34.0). In cross-sectional analyses, there were significant between-cluster differences in chronic kidney disease (CKD), distal sensorimotor polyneuropathy (DSPN) and cardiovascular disease (all p < 0.045). In prospective analyses (mean follow-up time 6.5-8.3 years), clusters differed significantly in CKD and DSPN incidence, but not in incident CVD or all-cause mortality. The inflammatory load was highest in the high-risk cluster 5 and lowest in cluster 2. Adjustment for the inflammatory load had only a minor impact on the aforementioned differences in outcomes between clusters. CONCLUSIONS Our findings extend the knowledge about the previously identified six phenotype-based clusters in older people without T2D. Differences between clusters were more pronounced for T2D risk than for prevalent or incident (pre)diabetes-related complications and absent for mortality. The high cardiometabolic risk corresponded to the high inflammatory load in cluster 5 but not to the lower inflammatory load of high-risk clusters 3 and 6.
Collapse
Affiliation(s)
- Marie-Theres Huemer
- Institute of Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Neuherberg, Germany
| | - Maria C Spagnuolo
- Institute for Clinical Diabetology, German Diabetes Center (DDZ), Leibniz Center for Diabetes Research at Heinrich Heine Universität, Auf'm Hennekamp 65, 40225, Düsseldorf, Germany
- German Center for Diabetes Research (DZD), Partner Düsseldorf, Neuherberg, Germany
| | - Haifa Maalmi
- Institute for Clinical Diabetology, German Diabetes Center (DDZ), Leibniz Center for Diabetes Research at Heinrich Heine Universität, Auf'm Hennekamp 65, 40225, Düsseldorf, Germany
- German Center for Diabetes Research (DZD), Partner Düsseldorf, Neuherberg, Germany
| | - Robert Wagner
- Institute for Clinical Diabetology, German Diabetes Center (DDZ), Leibniz Center for Diabetes Research at Heinrich Heine Universität, Auf'm Hennekamp 65, 40225, Düsseldorf, Germany
- German Center for Diabetes Research (DZD), Partner Düsseldorf, Neuherberg, Germany
- Department of Endocrinology and Diabetology, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine Universität, Düsseldorf, Germany
| | - Gidon J Bönhof
- Institute for Clinical Diabetology, German Diabetes Center (DDZ), Leibniz Center for Diabetes Research at Heinrich Heine Universität, Auf'm Hennekamp 65, 40225, Düsseldorf, Germany
- German Center for Diabetes Research (DZD), Partner Düsseldorf, Neuherberg, Germany
- Department of Endocrinology and Diabetology, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine Universität, Düsseldorf, Germany
| | - Margit Heier
- Institute of Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Neuherberg, Germany
| | - Wolfgang Koenig
- School of Medicine and Health, German Heart Centre, TUM University Hospital, Technical University of Munich, Munich, Germany
- German Centre for Cardiovascular Research (DZHK), Partner Site Munich Heart Alliance, Munich, Germany
- Institute of Epidemiology and Medical Biometry, University of Ulm, Ulm, Germany
| | - Wolfgang Rathmann
- German Center for Diabetes Research (DZD), Partner Düsseldorf, Neuherberg, Germany
- Institute for Biometrics and Epidemiology, German Diabetes Center (DDZ), Leibniz Center for Diabetes Research at Heinrich Heine Universität, Düsseldorf, Germany
| | - Katsiaryna Prystupa
- Institute for Clinical Diabetology, German Diabetes Center (DDZ), Leibniz Center for Diabetes Research at Heinrich Heine Universität, Auf'm Hennekamp 65, 40225, Düsseldorf, Germany
- German Center for Diabetes Research (DZD), Partner Düsseldorf, Neuherberg, Germany
| | - Jana Nano
- Institute of Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Neuherberg, Germany
| | - Dan Ziegler
- Institute for Clinical Diabetology, German Diabetes Center (DDZ), Leibniz Center for Diabetes Research at Heinrich Heine Universität, Auf'm Hennekamp 65, 40225, Düsseldorf, Germany
| | - Annette Peters
- Institute of Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Neuherberg, Germany
- German Centre for Cardiovascular Research (DZHK), Partner Site Munich Heart Alliance, Munich, Germany
- German Center for Diabetes Research (DZD), Partner München-Neuherberg, Neuherberg, Germany
- Institute for Medical Information Processing, Biometry and Epidemiology (IBE), Faculty of Medicine, Pettenkofer School of Public Health, LMU Munich, Munich, Germany
| | - Michael Roden
- Institute for Clinical Diabetology, German Diabetes Center (DDZ), Leibniz Center for Diabetes Research at Heinrich Heine Universität, Auf'm Hennekamp 65, 40225, Düsseldorf, Germany
- German Center for Diabetes Research (DZD), Partner Düsseldorf, Neuherberg, Germany
- Department of Endocrinology and Diabetology, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine Universität, Düsseldorf, Germany
| | - Barbara Thorand
- Institute of Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Neuherberg, Germany
- German Center for Diabetes Research (DZD), Partner München-Neuherberg, Neuherberg, Germany
- Institute for Medical Information Processing, Biometry and Epidemiology (IBE), Faculty of Medicine, Pettenkofer School of Public Health, LMU Munich, Munich, Germany
| | - Christian Herder
- Institute for Clinical Diabetology, German Diabetes Center (DDZ), Leibniz Center for Diabetes Research at Heinrich Heine Universität, Auf'm Hennekamp 65, 40225, Düsseldorf, Germany.
- German Center for Diabetes Research (DZD), Partner Düsseldorf, Neuherberg, Germany.
- Department of Endocrinology and Diabetology, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine Universität, Düsseldorf, Germany.
| |
Collapse
|
43
|
Wu T, Dai Z, Luo Y, Yu Q, Zhang Y, Bao X, Li R, Zhang Y, Hao J, Shen Q, Xue Y. Refined highland barley ameliorates obesity-associated insulin resistance in high-fat diet-fed mice by targeting the gut microbiota and liver transcriptomics. Eur J Nutr 2025; 64:96. [PMID: 39964534 DOI: 10.1007/s00394-025-03614-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Accepted: 02/06/2025] [Indexed: 03/19/2025]
Abstract
PURPOSE It is generally believed that refined grains lack nutritional value compared to whole grains. The objective of this study was to investigate whether refined highland barley (RHB) holds the potential to combat obesity-associated insulin resistance. METHODS Thirty-two male 6-week-old C57BL/6J mice were randomly divided into four groups fed with a normal chow diet, a high-fat diet (HFD), a 30% RHB supplemented HFD diet, and a 30% whole-grain highland barley (WGHB) supplemented HFD diet. We examined the anti-obesity and anti-insulin resistance effects of RHB and compared them with WGHB in mice. RESULTS RHB intervention effectively improved obesity and insulin resistance, enhanced the intestinal mucosal barrier, and reduced inflammation. Moreover, it promoted the abundance of beneficial gut bacteria such as Akkermansia, Bifidobacterium, Lachnospiraceae_NK4A136_group, Lachnospiraceae_UCG-001, Alloprevotella, and increased the production of short-chain fatty acids (SCFAs) in faeces. Additionally, RHB intervention modulated liver gene transcription, downregulating inflammatory genes like IRF3/7, STAT1/2, NLRP3, and TLR2. CONCLUSIONS RHB could effectively alleviate obesity-related insulin resistance by targeting gut microbiota and liver transcriptomics, and its beneficial impacts are comparable to those of WGHB.
Collapse
Affiliation(s)
- Tong Wu
- College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, 100083, China
- National Center of Technology Innovation in Food Industry, China Agricultural University, Beijing, 100083, China
| | - Zijian Dai
- College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, 100083, China
- National Center of Technology Innovation in Food Industry, China Agricultural University, Beijing, 100083, China
| | - Yingting Luo
- College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, 100083, China
| | - Qinye Yu
- College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, 100083, China
| | - Yiyun Zhang
- College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, 100083, China
- National Center of Technology Innovation in Food Industry, China Agricultural University, Beijing, 100083, China
| | - Xin Bao
- College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, 100083, China
- National Center of Technology Innovation in Food Industry, China Agricultural University, Beijing, 100083, China
| | - Rong Li
- College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, 100083, China
- National Center of Technology Innovation in Food Industry, China Agricultural University, Beijing, 100083, China
| | - Yuhong Zhang
- Tibet Academy of Agriculture and Animal Sciences, Lhasa, 850002, China
| | - Jing Hao
- Qinghai Tianyoude Technology Investment Management Group Co., Ltd, Qinghai, 810500, China
- Qinghai Engineering Technology Research Institute for Comprehensive Utilization of Highland Barley Resources, Qinghai, 810016, China
| | - Qun Shen
- College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, 100083, China
- National Center of Technology Innovation in Food Industry, China Agricultural University, Beijing, 100083, China
| | - Yong Xue
- College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, 100083, China.
- National Center of Technology Innovation in Food Industry, China Agricultural University, Beijing, 100083, China.
| |
Collapse
|
44
|
Zhang CY, Liu S, Sui YX, Yang M. Nucleotide-binding domain, leucine-rich repeat, and pyrin domain-containing protein 3 inflammasome: From action mechanism to therapeutic target in clinical trials. World J Gastrointest Oncol 2025; 17:100094. [PMID: 39958558 PMCID: PMC11756006 DOI: 10.4251/wjgo.v17.i2.100094] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Revised: 10/23/2024] [Accepted: 11/05/2024] [Indexed: 01/18/2025] Open
Abstract
The nucleotide-binding domain, leucine-rich repeat, and pyrin domain-containing protein 3 (NLRP3) inflammasome is a critical modulator in inflammatory disease. Activation and mutation of NLRP3 can cause severe inflammation in diseases such as chronic infantile neurologic cutaneous and articular syndrome, Muckle-Wells syndrome, and familial cold autoinflammatory syndrome 1. To date, a great effort has been made to decode the underlying mechanisms of NLRP3 activation. The priming and activation of NLRP3 drive the maturation and release of active interleukin (IL)-18 and IL-1β to cause inflammation and pyroptosis, which can significantly trigger many diseases including inflammatory diseases, immune disorders, metabolic diseases, and neurodegenerative diseases. The investigation of NLRP3 as a therapeutic target for disease treatment is a hot topic in both preclinical studies and clinical trials. Developing potent NLRP3 inhibitors and downstream IL-1 inhibitors attracts wide-spectrum attention in both research and pharmaceutical fields. In this minireview, we first updated the molecular mechanisms involved in NLRP3 inflammasome activation and the associated downstream signaling pathways. We then reviewed the molecular and cellular pathways of NLRP3 in many diseases, including obesity, diabetes, and other metabolic diseases. In addition, we briefly reviewed the roles of NLRP3 in cancer growth and relative immune checkpoint therapy. Finally, clinical trials with treatments targeting NLRP3 and its downstream signaling pathways were summarized.
Collapse
Affiliation(s)
- Chun-Ye Zhang
- Bond Life Sciences Center, University of Missouri, Columbia, MO 65212, United States
| | - Shuai Liu
- The First Affiliated Hospital, Zhejiang University, Hangzhou 310006, Zhejiang Province, China
| | - Yu-Xiang Sui
- School of Life Science, Shanxi Normal University, Linfen 041004, Shanxi Province, China
| | - Ming Yang
- Department of Surgery, University of Connecticut, School of Medicine, Farmington, CT 06030, United States
| |
Collapse
|
45
|
D’Amico RC, Nagashima S, Carstens LB, Bertoldi KDG, Mataruco S, Honório D’Agostini JC, Hlatchuk EC, da Silva SB, de Noronha L, Baena CP. COVID-19 Induces Greater NLRP3 Inflammasome Activation in Obese Patients than Other Chronic Illnesses: A Case-Control Study. Int J Mol Sci 2025; 26:1541. [PMID: 40004007 PMCID: PMC11855377 DOI: 10.3390/ijms26041541] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2024] [Revised: 02/02/2025] [Accepted: 02/05/2025] [Indexed: 02/27/2025] Open
Abstract
Obesity has been identified as an independent risk factor for severe COVID-19 unfavorable outcomes. Several factors, such as increased ACE2 receptor expression and chronic inflammation, can contribute to this relationship, yet the activation of the NLRP3 inflammasome pathway is also a key element. Our primary goal was to determine whether chronic NLRP3 inflammasome activation in people with obesity is different in critical COVID-19 and in critical chronic conditions. A retrospective analysis was conducted using clinical data and post-mortem lung tissue samples from 14 COVID-19 patients with obesity (group A) and 9 patients with obesity who died from non-COVID-19 causes (group B). Immunohistochemical analysis assessed twelve markers related to the NLRP3 inflammasome pathway. Group A showed a significantly higher expression of ASC (p = 0.0387) and CASP-1 (p = 0.0142). No significant differences were found for IL-8, TNF-α, NF-kB, NLRP3, IL-1β, and gasdermin-D. Group B had higher levels of IL-6 (p < 0.0001), IL-18 (p = 0.002), CASP-9 (p < 0.0001), and HIF (p = 0.0327). We concluded that COVID-19 activates the NLRP3 inflammasome pathway, possibly leading to pyroptotic cell death mediated by caspase-1. In contrast, people with obesity without COVID-19, despite exhibiting some markers of the NLRP3 inflammasome, are more likely to experience necroptosis mediated by caspase-9.
Collapse
Affiliation(s)
- Raíssa Campos D’Amico
- Post-Graduate Program in Health Sciences (PPGCS), Pontifícia Universidade Católica do Paraná, Curitiba 80215-901, Paraná, Brazil; (R.C.D.); (S.N.); (L.B.C.); (K.d.G.B.); (C.P.B.)
- School of Medicine, Pontifícia Universidade Católica do Paraná, Curitiba 80215-901, Paraná, Brazil; (S.M.); (S.B.d.S.)
| | - Seigo Nagashima
- Post-Graduate Program in Health Sciences (PPGCS), Pontifícia Universidade Católica do Paraná, Curitiba 80215-901, Paraná, Brazil; (R.C.D.); (S.N.); (L.B.C.); (K.d.G.B.); (C.P.B.)
| | - Lucas Baena Carstens
- Post-Graduate Program in Health Sciences (PPGCS), Pontifícia Universidade Católica do Paraná, Curitiba 80215-901, Paraná, Brazil; (R.C.D.); (S.N.); (L.B.C.); (K.d.G.B.); (C.P.B.)
- School of Medicine, Pontifícia Universidade Católica do Paraná, Curitiba 80215-901, Paraná, Brazil; (S.M.); (S.B.d.S.)
| | - Karina de Guadalupe Bertoldi
- Post-Graduate Program in Health Sciences (PPGCS), Pontifícia Universidade Católica do Paraná, Curitiba 80215-901, Paraná, Brazil; (R.C.D.); (S.N.); (L.B.C.); (K.d.G.B.); (C.P.B.)
| | - Sabrina Mataruco
- School of Medicine, Pontifícia Universidade Católica do Paraná, Curitiba 80215-901, Paraná, Brazil; (S.M.); (S.B.d.S.)
| | | | - Elisa Carolina Hlatchuk
- School of Medicine, Universidade Federal do Paraná, Curitiba 80060-240, Paraná, Brazil; (J.C.H.D.); (E.C.H.)
| | - Sofia Brunoro da Silva
- School of Medicine, Pontifícia Universidade Católica do Paraná, Curitiba 80215-901, Paraná, Brazil; (S.M.); (S.B.d.S.)
| | - Lucia de Noronha
- Post-Graduate Program in Health Sciences (PPGCS), Pontifícia Universidade Católica do Paraná, Curitiba 80215-901, Paraná, Brazil; (R.C.D.); (S.N.); (L.B.C.); (K.d.G.B.); (C.P.B.)
- School of Medicine, Pontifícia Universidade Católica do Paraná, Curitiba 80215-901, Paraná, Brazil; (S.M.); (S.B.d.S.)
- School of Medicine, Universidade Federal do Paraná, Curitiba 80060-240, Paraná, Brazil; (J.C.H.D.); (E.C.H.)
| | - Cristina Pellegrino Baena
- Post-Graduate Program in Health Sciences (PPGCS), Pontifícia Universidade Católica do Paraná, Curitiba 80215-901, Paraná, Brazil; (R.C.D.); (S.N.); (L.B.C.); (K.d.G.B.); (C.P.B.)
- School of Medicine, Pontifícia Universidade Católica do Paraná, Curitiba 80215-901, Paraná, Brazil; (S.M.); (S.B.d.S.)
| |
Collapse
|
46
|
Zhang Y, Li M, Liu H, Fan Y, Liu HH. The application of procyanidins in diabetes and its complications: a review of preclinical studies. Front Pharmacol 2025; 16:1532246. [PMID: 39995417 PMCID: PMC11847907 DOI: 10.3389/fphar.2025.1532246] [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: 11/21/2024] [Accepted: 01/14/2025] [Indexed: 02/26/2025] Open
Abstract
Diabetes mellitus (DM) and its various complications, including diabetic nephropathy, retinopathy, neuropathy, cardiovascular disease, and ulcers, pose significant challenges to global health. This review investigates the potential of procyanidins (PCs), a natural polyphenolic compound, in preventing and managing diabetes and its complications. PCs, recognized for their strong antioxidant, anti-inflammatory, and anti-hyperglycemic properties, play a crucial role in reducing oxidative stress and enhancing endothelial function, which are essential for managing diabetic complications. This review elucidates the molecular mechanisms by which PCs improve insulin sensitivity and endothelial health, thereby providing protection against the various complications of diabetes. The comprehensive analysis underscores the promising therapeutic role of PCs in diabetes care, indicating the need for further clinical studies to confirm and leverage their potential in comprehensive diabetes management strategies.
Collapse
Affiliation(s)
- Yongchuang Zhang
- Department of Rehabilitation Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Mengna Li
- Institute of Pain Medicine and Special Environmental Medicine, Nantong University, Nantong, China
| | - Haoyuan Liu
- Rehabilitation Department, Henan Provincial Hospital of Traditional Chinese Medicine, Zhengzhou, China
| | - Yongfu Fan
- School of Rehabilitation Medicine, Henan University of Chinese Medicine, Zhengzhou, China
| | - Huan Huan Liu
- International institute for Traditional Chinese Medicine, Guanzhou University of Chinese Medicine, Guangzhou, China
| |
Collapse
|
47
|
Rahman MA, Datta S, Lakkakula H, Koka S, Boini KM. Acid Sphingomyelinase and Ceramide Signaling Pathway Mediates Nicotine-Induced NLRP3 Inflammasome Activation and Podocyte Injury. Biomedicines 2025; 13:416. [PMID: 40002829 PMCID: PMC11852453 DOI: 10.3390/biomedicines13020416] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2024] [Revised: 01/23/2025] [Accepted: 02/07/2025] [Indexed: 02/27/2025] Open
Abstract
Background: Recent studies have shown that Nlrp3 inflammasome activation is importantly involved in podocyte dysfunction induced by nicotine. The present study was designed to test whether acid sphingomyelinase (Asm) and ceramide signaling play a role in mediating nicotine-induced Nlrp3 inflammasome activation and subsequent podocyte damage. Methods and Results: Nicotine treatment significantly increased the Asm expression and ceramide production compared to control cells. However, prior treatment with amitriptyline, an Asm inhibitor significantly attenuated the nicotine-induced Asm expression and ceramide production. Confocal microscopic and biochemical analyses showed that nicotine treatment increased the colocalization of NLRP3 with Asc, Nlrp3 vs. caspase-1, IL-1β production, caspase-1 activity, and desmin expression in podocytes compared to control cells. Pretreatment with amitriptyline abolished the nicotine-induced colocalization of NLRP3 with Asc, Nlrp3 with caspase-1, IL-1β production, caspase-1 activity and desmin expression. Immunofluorescence analyses showed that nicotine treatment significantly decreased the podocin expression compared to control cells. However, prior treatment with amitriptyline attenuated the nicotine-induced podocin reduction. In addition, nicotine treatment significantly increased the cell permeability, O2 production, and apoptosis compared to control cells. However, prior treatment with amitriptyline significantly attenuated the nicotine-induced cell permeability, O2 production and apoptosis in podocytes. Conclusions: Asm is one of the important mediators of nicotine-induced inflammasome activation and podocyte injury. Asm may be a therapeutic target for the treatment or prevention of glomerulosclerosis associated with smoking.
Collapse
Affiliation(s)
- Mohammad Atiqur Rahman
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, 4349 Martin Luther King Blvd, Houston, TX 77204, USA (H.L.)
| | - Sayantap Datta
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, 4349 Martin Luther King Blvd, Houston, TX 77204, USA (H.L.)
| | - Harini Lakkakula
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, 4349 Martin Luther King Blvd, Houston, TX 77204, USA (H.L.)
- Novi High School, Novi, MI 48375, USA
| | - Saisudha Koka
- Department of Pharmaceutical Sciences, Irma Lerma Rangel College of Pharmacy, Texas A & M University, Kingsville, TX 78363, USA
| | - Krishna M. Boini
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, 4349 Martin Luther King Blvd, Houston, TX 77204, USA (H.L.)
| |
Collapse
|
48
|
Fernandes DC, Silva-de-França F, Pohl PC, Eto SF, Sardinha LR, Lambris JD, Tambourgi DV. Cp40-mediated complement C3 inhibition dampens inflammasome activation and inflammatory mediators storm induced by Bitis arietans venom. Int Immunopharmacol 2025; 147:113701. [PMID: 39809101 DOI: 10.1016/j.intimp.2024.113701] [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: 10/01/2024] [Revised: 11/19/2024] [Accepted: 11/20/2024] [Indexed: 01/16/2025]
Abstract
The complement system plays a crucial role in various pathophysiological conditions, including snake envenomation. In this study, we investigated the effects of Bitis arietans venom on the complement system using an ex vivo human whole blood model. Our findings demonstrate that B. arietans venom was able to activate the complement system, leading to a significant increase in the production of anaphylatoxins (C3a/C3a-desArg, C5a/C5a-desArg) and the soluble Terminal Complement Complex (sTCC). Inhibition of the C3 component by Cp40, a C3-C3b inhibitor, resulted in the reduction of C3a/C3a-desArg, C5a/C5a-desArg, and sTCC levels to baseline in venom-stimulated samples. Furthermore, treatment with Cp40 promoted a substantial decrease in the production of pro-inflammatory mediators, such as Prostaglandin E2 (PGE2), IL-8/CXCL8, MCP-1/CCL2, and MIG/CXCL9. To further elucidate the molecular mechanisms, we utilized the THP-1 cell line differentiated into M0 macrophages. Incubation of these macrophages with human plasma, from the human whole blood treated with B. arietans venom, resulted in the expression of the NLRP3 inflammasome and the production of IL-8 and IL-1β. Importantly, Cp40 was able to diminish the production of these cytokines, as well as the levels of ASC and caspase-1 proteins. In conclusion, our results indicate that the inhibition of the complement by Cp40 at C3/C3b level can modulate the inflammatory response and inflammasome activation induced by B. arietans venom. These findings suggest that complement inhibition may be a promising therapeutic approach for managing the inflammatory complications associated with this snake envenomation.
Collapse
Affiliation(s)
- Dayanne Carla Fernandes
- Immunochemistry Laboratory, Butantan Institute, São Paulo, SP, Brazil; Center of Toxins, Cell Signaling and Immune Response (CeTICS), CEPID, FAPESP, Brazil
| | - Felipe Silva-de-França
- Immunochemistry Laboratory, Butantan Institute, São Paulo, SP, Brazil; Center of Toxins, Cell Signaling and Immune Response (CeTICS), CEPID, FAPESP, Brazil
| | | | | | | | - John D Lambris
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, United States
| | - Denise V Tambourgi
- Immunochemistry Laboratory, Butantan Institute, São Paulo, SP, Brazil; Center of Toxins, Cell Signaling and Immune Response (CeTICS), CEPID, FAPESP, Brazil.
| |
Collapse
|
49
|
Parrotta ME, Colangeli L, Scipione V, Vitale C, Sbraccia P, Guglielmi V. Time Restricted Eating: A Valuable Alternative to Calorie Restriction for Addressing Obesity? Curr Obes Rep 2025; 14:17. [PMID: 39899119 PMCID: PMC11790783 DOI: 10.1007/s13679-025-00609-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 01/20/2025] [Indexed: 02/04/2025]
Abstract
PURPOSE OF REVIEW In this review, we summarize the molecular effects of time-restricted eating (TRE) and its possible role in appetite regulation. We also discuss the potential clinical benefits of TRE in obesity. RECENT FINDINGS TRE is an emerging dietary approach consisting in limiting food intake to a specific window of time each day. The rationale behind this strategy is to restore the circadian misalignment, commonly seen in obesity. Preclinical studies have shown that restricting food intake only during the active phase of the day can positively influence several cellular functions including senescence, mitochondrial activity, inflammation, autophagy and nutrients' sensing pathways. Furthermore, TRE may play a role by modulating appetite and satiety hormones, though further research is needed to clarify its exact mechanisms. Clinical trials involving patients with obesity or type 2 diabetes suggest that TRE can be effective for weight loss, but its broader effects on improving other clinical outcomes, such as cardiovascular risk factors, remain less certain. The epidemic proportions of obesity cause urgency to find dietary, pharmacological and surgical interventions that can be effective in the medium and long term. According to its molecular effects, TRE can be an interesting alternative to caloric restriction in the treatment of obesity, but the considerable variability across clinical trials regarding population, intervention, and follow-up duration makes it difficult to reach definitive conclusions.
Collapse
Affiliation(s)
| | - Luca Colangeli
- Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy
- Internal Medicine Unit - Obesity Center, University Hospital Policlinico Tor Vergata, Rome, Italy
| | - Valeria Scipione
- Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy
| | - Carolina Vitale
- Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy
| | - Paolo Sbraccia
- Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy
- Internal Medicine Unit - Obesity Center, University Hospital Policlinico Tor Vergata, Rome, Italy
| | - Valeria Guglielmi
- Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy.
- Internal Medicine Unit - Obesity Center, University Hospital Policlinico Tor Vergata, Rome, Italy.
| |
Collapse
|
50
|
Song M, Bai Y, Song F. High-fat diet and neuroinflammation: The role of mitochondria. Pharmacol Res 2025; 212:107615. [PMID: 39842474 DOI: 10.1016/j.phrs.2025.107615] [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: 08/15/2024] [Revised: 12/28/2024] [Accepted: 01/17/2025] [Indexed: 01/24/2025]
Abstract
In recent years, increasing evidence has supported that high-fat diet (HFD) can induce the chronic, low-grade neuroinflammation in the brain, which is closely associated with the impairment of cognitive function. As the key organelles responsible for energy metabolism in the cell, mitochondria are believed to involved in the pathogenesis of a variety of neurological disorders. This review summarizes the current progress in the field of the relationship between HFD exposure and neurodegenerative diseases, and outline the major routines of HFD induced neuroinflammation and its pathological significance in the pathogenesis of neurodegenerative diseases. Furthermore, the article highlights the pivotal role of mitochondrial dysfunction in driving the neuroinflammation in the setting of HFD. Danger-associated molecular patterns (DAMPs) from damaged mitochondria can activate innate immune signaling pathways, while mitochondrial dysfunction itself can lead to metabolic remodeling of inflammatory cells, thus inducing neuroinflammation. More importantly, mitochondrial damage, neuroinflammation, and insulin resistance caused by HFD form a mutually reinforcing vicious cycle, ultimately leading to the death of neurons and promoting the progression of neurodegenerative diseases. Thus, in-depth elucidation of the role and underlying mechanisms of mitochondrial dysfunction in HFD-induced metabolic disorders may not only expand our understanding of the mechanistic linkages between HFD and etiology of neurodegenerative diseases, but also help develop the specific strategies for the prevention and treatment of neurodegenerative diseases.
Collapse
Affiliation(s)
- Mingxue Song
- Department of Toxicology, School of Public Health, Cheeloo College of Medicine, Shandong University, 44 Wenhuaxi Road, Jinan, Shandong 250012, China.
| | - Yao Bai
- NHC Key Laboratory of Food Safety Risk Assessment, China National Center for Food Safety Risk Assessment, Beijing 100021, China.
| | - Fuyong Song
- Department of Toxicology, School of Public Health, Cheeloo College of Medicine, Shandong University, 44 Wenhuaxi Road, Jinan, Shandong 250012, China.
| |
Collapse
|