1
|
Khanna S, Kumar S, Sharma P, Daksh R, Nandakumar K, Shenoy RR. Flavonoids regulating NLRP3 inflammasome: a promising approach in alleviating diabetic peripheral neuropathy. Inflammopharmacology 2025:10.1007/s10787-025-01729-7. [PMID: 40205269 DOI: 10.1007/s10787-025-01729-7] [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: 01/21/2025] [Accepted: 01/25/2025] [Indexed: 04/11/2025]
Abstract
A common and serious side effect of diabetes is diabetic peripheral neuropathy (DPN), which is characterised by gradual nerve damage brought on by oxidative stress, chronic inflammation, and prolonged hyperglycemia. Studies identify NLRP3 inflammasome as a key mediator in the pathogenesis of DPN, connecting neuroinflammation and neuronal damage to metabolic failure. Because of their strong anti-inflammatory and antioxidant qualities, flavonoids, a broad class of naturally occurring polyphenols, have drawn interest as potential treatments for DPN. The various ways that flavonoids affect the NLRP3 inflammasome and their potential as a treatment for DPN are examined in this review. It has been demonstrated that flavonoids prevent NLRP3 activation, which lowers the release of pro-inflammatory cytokines including IL-1β and IL-18 and causes neuroinflammation. Flavonoids work mechanistically by reducing oxidative stress, altering important signalling pathways, and blocking the activities of NF-κB and caspase-1, which are both essential for the activation of the NLRP3 inflammasome. Preclinical research has shown that flavonoids have strong neuroprotective benefits, and few clinical evidence also points to the potential of flavonoids to improve nerve function and lessen neuropathic pain in diabetic patients. The current review emphasises how flavonoids may be used as a treatment strategy to target inflammation in DPN caused by the NLRP3 inflammasome. By targeting important inflammatory pathways, flavonoids provide a new way to slow the progression of this debilitating illness. Further investigation into the mechanisms, clinical translation, and novel drug delivery techniques could enhance the therapeutic efficacy of diabetic peripheral neuropathy.
Collapse
Affiliation(s)
- Saumya Khanna
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, India, 576104
| | - Sachindra Kumar
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, India, 576104
| | - Pratyasha Sharma
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, India, 576104
| | - Rajni Daksh
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, India, 576104
| | - Krishnadas Nandakumar
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, India, 576104
| | - Rekha Raghuveer Shenoy
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, India, 576104.
| |
Collapse
|
2
|
Wong A, Sun Q, Latif II, Karwi QG. Macrophage energy metabolism in cardiometabolic disease. Mol Cell Biochem 2025; 480:1763-1783. [PMID: 39198360 PMCID: PMC11842501 DOI: 10.1007/s11010-024-05099-6] [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: 04/27/2024] [Accepted: 08/19/2024] [Indexed: 09/01/2024]
Abstract
In a rapidly expanding body of literature, the major role of energy metabolism in determining the response and polarization status of macrophages has been examined, and it is currently a very active area of research. The metabolic flux through different metabolic pathways in the macrophage is interconnected and complex and could influence the polarization of macrophages. Earlier studies suggested glucose flux through cytosolic glycolysis is a prerequisite to trigger the pro-inflammatory phenotypes of macrophages while proposing that fatty acid oxidation is essential to support anti-inflammatory responses by macrophages. However, recent studies have shown that this understanding is oversimplified and that the metabolic control of macrophage polarization is highly complex and not fully defined yet. In this review, we systematically reviewed and summarized the literature regarding the role of energy metabolism in controlling macrophage activity and how that might be altered in cardiometabolic diseases, namely heart failure, obesity, and diabetes. We critically appraised the experimental studies and methodologies in the published studies. We also highlighted the challenging concepts in macrophage metabolism and identified several research questions yet to be addressed in future investigations.
Collapse
Affiliation(s)
- Angela Wong
- Division of BioMedical Sciences, Faculty of Medicine, Memorial University of Newfoundland, St. John's, Newfoundland and Labrador, A1B 3V6, Canada
- Cardiovascular Research Centre, University of Alberta, Edmonton, Alberta, Canada
| | - Qiuyu Sun
- Division of BioMedical Sciences, Faculty of Medicine, Memorial University of Newfoundland, St. John's, Newfoundland and Labrador, A1B 3V6, Canada
- Cardiovascular Research Centre, University of Alberta, Edmonton, Alberta, Canada
| | - Ismail I Latif
- Department of Microbiology, College of Medicine, University of Diyala, Baqubaa, Diyala, Iraq
| | - Qutuba G Karwi
- Division of BioMedical Sciences, Faculty of Medicine, Memorial University of Newfoundland, St. John's, Newfoundland and Labrador, A1B 3V6, Canada.
| |
Collapse
|
3
|
Sirajee R, El Khatib S, Dieleman LA, Salla M, Baksh S. ImmunoMet Oncogenesis: A New Concept to Understand the Molecular Drivers of Cancer. J Clin Med 2025; 14:1620. [PMID: 40095546 PMCID: PMC11900543 DOI: 10.3390/jcm14051620] [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: 10/31/2024] [Revised: 02/10/2025] [Accepted: 02/25/2025] [Indexed: 03/19/2025] Open
Abstract
The appearance of cancer progresses through a multistep process that includes genetic, epigenetic, mutational, inflammatory and metabolic disturbances to signaling pathways within an organ. The combined influence of these changes will dictate the growth properties of the cells; the direction of further malignancy depends on the severity of these "disturbances". The molecular mechanisms driving abnormal inflammation and metabolism are beginning to be identified and, in some cases, are quite prominent in pre-condition states of cancer and are significant drivers of the malignant phenotype. As such, utilizing signaling pathways linked to inflammation and metabolism as biomarkers of cancer is an emerging method and includes pathways beyond those well characterized to drive metabolism or inflammation. In this review, we will discuss several emerging elements influencing proliferation, inflammation and metabolism that may play a part as drivers of the cancer phenotype. These include AMPK and leptin (linked to metabolism), NOD2/RIPK2, TAK1 (linked to inflammation), lactate and pyruvate transporters (monocarboxylate transporter [MCT], linked to mitochondrial biogenesis and metabolism) and RASSF1A (linked to proliferation, cell death, cell cycle control, inflammation and epigenetics). We speculate that the aforementioned elements are important drivers of carcinogenesis that should be collectively referenced as being involved in "ImmunoMET Oncogenesis", a new tripartite description of the role of elements in driving cancer. This term would suggest that for a better understanding of cancer, we need to understand how proliferation, inflammation and metabolic pathways are impacted and how they influence classical drivers of malignant transformation in order to drive ImmunoMET oncogenesis and the malignant state.
Collapse
Affiliation(s)
- Reshma Sirajee
- Faculty of Science, 1-001 CCIS, University of Alberta, Edmonton, AB T6G 2E1, Canada;
| | - Sami El Khatib
- Department of Biological & Chemical Sciences, Bekaa Campus, Lebanese International University, West Bekaa, Khiyara 1106, Lebanon; (S.E.K.); (M.S.)
- Center for Applied Mathematics and Bioinformatics (CAMB), Gulf University for Science and Technology, Kuwait City 32093, Kuwait
| | - Levinus A. Dieleman
- Department of Medicine, Faculty of Medicine and Dentistry, University of Alberta, 113 Street 87 Avenue, Edmonton, AB T6G 2E1, Canada;
| | - Mohamed Salla
- Department of Biological & Chemical Sciences, Bekaa Campus, Lebanese International University, West Bekaa, Khiyara 1106, Lebanon; (S.E.K.); (M.S.)
| | - Shairaz Baksh
- Department of Pediatrics, Biochemistry and Division of Experimental Oncology, Faculty of Medicine and Dentistry, University of Alberta, 113 Street 87 Avenue, Edmonton, AB T6G 2E1, Canada
- Women and Children’s Health Research Institute, Edmonton Clinic Health Academy (ECHA), University of Alberta, 4-081 11405 87 Avenue, Edmonton, AB T6G 1C9, Canada
- BioImmuno Designs, 4747 154 Avenue, Edmonton, AB T5Y 0C2, Canada
- Bio-Stream Diagnostics, 2011 94 Street, Edmonton, AB T6H 1N1, Canada
| |
Collapse
|
4
|
Ma J, Wang Y, Xu W, Wang H, Wan Z, Guo J. Macrophage pyroptosis in atherosclerosis: therapeutic potential. Acta Biochim Biophys Sin (Shanghai) 2025. [PMID: 39953798 DOI: 10.3724/abbs.2025004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/17/2025] Open
Abstract
Atherosclerosis (AS) is a chronic inflammatory disease characterized by the accumulation of lipid-rich plaques in arterial walls, leading to cardiovascular events such as myocardial infarction and stroke. Macrophage pyroptosis, a form of programmed cell death driven by the NLRP3 inflammasome and caspase-1 activation, plays a critical role in the progression and destabilization of atherosclerotic plaques. This review explores the molecular mechanisms underlying macrophage pyroptosis and their significant contributions to AS pathogenesis. Recent advancements have highlighted the therapeutic potential of targeting key components of the pyroptotic pathway, including the use of nanotechnology to increase drug delivery specificity. These strategies are promising for reducing inflammation, stabilizing plaques, and mitigating the clinical impact of AS. Future studies should focus on translating these findings into clinical applications to develop effective treatments that can halt or reverse AS progression by modulating macrophage pyroptosis.
Collapse
Affiliation(s)
- Jianying Ma
- Department of Vascular and Endovascular Surgery, the First Affiliated Hospital of Yangtze University, Jingzhou 434000, China
- Department of Pharmacology, School of Medicine, Yangtze University, Jingzhou 434023, China
- Department of Interventional, Jingzhou Hospital Affiliated to Yangtze University, Jingzhou 434020, China
| | - Yixian Wang
- Department of Vascular and Endovascular Surgery, the First Affiliated Hospital of Yangtze University, Jingzhou 434000, China
- Department of Pharmacology, School of Medicine, Yangtze University, Jingzhou 434023, China
| | - Wenna Xu
- Department of Vascular and Endovascular Surgery, the First Affiliated Hospital of Yangtze University, Jingzhou 434000, China
- Department of Pharmacology, School of Medicine, Yangtze University, Jingzhou 434023, China
| | - Hanjing Wang
- Department of Vascular and Endovascular Surgery, the First Affiliated Hospital of Yangtze University, Jingzhou 434000, China
- Department of Pharmacology, School of Medicine, Yangtze University, Jingzhou 434023, China
| | - Zhengdong Wan
- Department of Vascular and Endovascular Surgery, the First Affiliated Hospital of Yangtze University, Jingzhou 434000, China
| | - Jiawei Guo
- Department of Vascular and Endovascular Surgery, the First Affiliated Hospital of Yangtze University, Jingzhou 434000, China
- Department of Pharmacology, School of Medicine, Yangtze University, Jingzhou 434023, China
| |
Collapse
|
5
|
Shan J, Ding J, Li DJ, Wang XQ. The double-edged role of IL-18 in reproductive endocrine and reproductive immune related disorders. Int Immunopharmacol 2025; 147:113859. [PMID: 39755106 DOI: 10.1016/j.intimp.2024.113859] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2024] [Revised: 11/15/2024] [Accepted: 12/11/2024] [Indexed: 01/06/2025]
Abstract
Interleukin (IL)-18 is one of the members of IL-1 family cytokines, it was originally named as interferon gamma (IFN-γ) inducing factor. IL-18 is a pleiotropic immune regulator and has a bidirectional regulatory effect on immunity. It exerts a potent pro-inflammatory effect by inducing the expression of IFN-γ, also has an important anti-inflammatory role. In recent years, IL-18 has received widespread attention and become a research hotspot. Previous studies have described the roles of IL-18 in the pathogenesis of many diseases. However, the biologic activities of IL-18 and its role in the reproductive endocrine and reproductive immune related diseases are still not well understood, such as endometriosis (EMS), recurrent spontaneous abortion (RSA), polycystic ovary syndrome (PCOS), and infertility, which are closely related to inflammation and immunity. Here, we reviewed the research progress of IL-18 in these diseases in the past few years. This article provides an overview of the latest knowledge about the roles of IL-18 in these diseases, with a view to providing new possibilities for the diagnosis and treatment of reproductive endocrine and reproductive immune related disorders.
Collapse
Affiliation(s)
- Jing Shan
- Laboratory for Reproductive Immunology, Key Laboratory of Reproduction Regulation of NPFPC, SIPPR, IRD, Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, The Obstetrics and Gynecology Hospital of Fudan University, Shanghai, China
| | - Jie Ding
- Naval Medical University, Changhai Hospital, Department of Traditional Chinese Medicine, Shanghai, China
| | - Da-Jin Li
- Laboratory for Reproductive Immunology, Key Laboratory of Reproduction Regulation of NPFPC, SIPPR, IRD, Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, The Obstetrics and Gynecology Hospital of Fudan University, Shanghai, China.
| | - Xiao-Qiu Wang
- Laboratory for Reproductive Immunology, Key Laboratory of Reproduction Regulation of NPFPC, SIPPR, IRD, Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, The Obstetrics and Gynecology Hospital of Fudan University, Shanghai, China.
| |
Collapse
|
6
|
Dehesh T, Mosleh-Shirazi MA, Dehesh P. Prevalence and associated factors of anxiety and depression among patients with hypothyroidism in Southern Iran. BMC Psychiatry 2025; 25:54. [PMID: 39833737 PMCID: PMC11748247 DOI: 10.1186/s12888-025-06490-3] [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: 06/04/2024] [Accepted: 01/09/2025] [Indexed: 01/22/2025] Open
Abstract
PURPOSE Depression and anxiety are the main disorders in patients suffering from hypothyroidism. These disorders can lead to increased patient suffering. Since hypothyroidism is one of the most prevalent endocrine diseases, controlling the metabolic variables that increase the severity of anxiety and depression is important. This study aimed to assess the prevalence of anxiety and depression and to identify their associated factors, including metabolic variables, among people with hypothyroidism. PATIENTS AND METHODS We performed a cross-sectional study of 1,600 patients with hypothyroidism in Kerman, the southern part of Iran. The prevalence of depression and anxiety was estimated using the Beck Depression Inventory and the Hamilton Anxiety questionnaires, respectively. First, univariate logistic regression was performed. Factors whose P-values were smaller than 0.2 in univariate logistic regression were included in multiple logistic regression for confounder adjustments. The analysis was performed using SPSS version 20. RESULTS The rates of depression and anxiety were 59% (95% CI: 53.18-62.11) and 63% (95% CI: 58.42-67.22), respectively. Factors found to be independently associated with anxiety were high TSH, high LDL, high TG, high FBS, high TGAb, high TPoAb, high TC, and hypertension. For depression, high TSH, high LDL, high TG, high FBS, high TC, and hypertension were identified. High TGAb and high TPoAb were independently associated with anxiety but not with depression. CONCLUSIONS Study findings revealed that a large proportion of patients with hypothyroidism suffer from depression and anxiety. This study also identified factors associated with these disorders. Controlling some metabolic variables may decrease the prevalence and severity of these disorders, help patients with better treatment, and improve their quality of life.
Collapse
Affiliation(s)
- Tania Dehesh
- Modeling in Health Research Center, Institute for Futures Studies in Health, Kerman University of Medical Sciences, Kerman, Iran
- Department of Biostatistics and Epidemiology, School of Public Health, Kerman University of Medical Sciences, Kerman, Iran
| | - Mohammad Amin Mosleh-Shirazi
- Ionizing and Non-Ionizing Radiation Protection Research Center (INIRPRC), School of Paramedical Sciences, Shiraz University of Medical Sciences, Shiraz, Iran
- Department of Radio-Oncology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Paria Dehesh
- Social Determinants of Health Research Center, Institute for Futures Studies in Health, Kerman University of Medical Sciences, Kerman, Iran.
- Department of Biostatistics and Epidemiology, School of Public Health, Kerman University of Medical Sciences, Kerman, Iran.
| |
Collapse
|
7
|
Yang J, Jiao C, Liu N, Liu W, Wang Y, Pan Y, Kong L, Guo W, Xu Q. Polydatin-Mediated Inhibition of HSP90α Disrupts NLRP3 Complexes and Alleviates Acute Pancreatitis. RESEARCH (WASHINGTON, D.C.) 2024; 7:0551. [PMID: 39691768 PMCID: PMC11651664 DOI: 10.34133/research.0551] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Revised: 11/16/2024] [Accepted: 11/23/2024] [Indexed: 12/19/2024]
Abstract
The NLRP3 inflammasome plays a critical role in various inflammatory conditions. However, despite extensive research in targeted drug development for NLRP3, including MCC950, clinical success remains elusive. Here, we discovered that the activated NLRP3 inflammasome complex (disc-NLRP3) and the activating mutation L351P exhibited resistance to MCC950. Through investigations using the small-molecule compound polydatin, HSP90α was found to stabilize both the resting (cage-NLRP3) and activated state (disc-NLRP3) of NLRP3 complexes, sustaining its activation. Our mechanistic studies revealed that polydatin specifically targets HSP90α, binding to it directly and subsequently interfering with the HSP90α-NLRP3 interaction. This disruption leads to the dissipation of cage-NLRP3, disc-NLRP3 complexes and NLRP3 L351P. Importantly, genetic and pharmacological inactivation of HSP90α effectively reduced NLRP3 inflammasome activation and alleviated cerulein-induced acute pancreatitis. These therapeutic effects highlight the clinical potential of HSP90α inhibition. Our findings demonstrate that HSP90α is crucial for the stability of both the resting and activated states of the NLRP3 inflammasome during its sustained activation, and targeting HSP90α represents a promising therapeutic strategy for diseases driven by the NLRP3 inflammasome.
Collapse
Affiliation(s)
- Jiashu Yang
- State Key Laboratory of Pharmaceutical Biotechnology, Department of Gastroenterology, Nanjing Drum Tower Hospital, School of Life Sciences,
Nanjing University, Nanjing, China
| | - Chenyang Jiao
- State Key Laboratory of Pharmaceutical Biotechnology, Department of Gastroenterology, Nanjing Drum Tower Hospital, School of Life Sciences,
Nanjing University, Nanjing, China
| | - Nannan Liu
- State Key Laboratory of Pharmaceutical Biotechnology, Department of Gastroenterology, Nanjing Drum Tower Hospital, School of Life Sciences,
Nanjing University, Nanjing, China
| | - Wen Liu
- State Key Laboratory of Pharmaceutical Biotechnology, Department of Gastroenterology, Nanjing Drum Tower Hospital, School of Life Sciences,
Nanjing University, Nanjing, China
| | - Yueyao Wang
- School of Pharmacy,
Nanjing University of Chinese Medicine, Nanjing, China
| | - Ying Pan
- State Key Laboratory of Pharmaceutical Biotechnology, Department of Gastroenterology, Nanjing Drum Tower Hospital, School of Life Sciences,
Nanjing University, Nanjing, China
| | - Lingdong Kong
- State Key Laboratory of Pharmaceutical Biotechnology, Department of Gastroenterology, Nanjing Drum Tower Hospital, School of Life Sciences,
Nanjing University, Nanjing, China
| | - Wenjie Guo
- State Key Laboratory of Pharmaceutical Biotechnology, Department of Gastroenterology, Nanjing Drum Tower Hospital, School of Life Sciences,
Nanjing University, Nanjing, China
| | - Qiang Xu
- State Key Laboratory of Pharmaceutical Biotechnology, Department of Gastroenterology, Nanjing Drum Tower Hospital, School of Life Sciences,
Nanjing University, Nanjing, China
| |
Collapse
|
8
|
Xue M, Du R, Zhou Y, Liu Y, Tian Y, Xu Y, Yan J, Song P, Wan L, Xu H, Zhang H, Liang H. Fucoidan Supplementation Relieved Kidney Injury and Modulated Intestinal Homeostasis in Hyperuricemia Mice. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2024; 72:27187-27202. [PMID: 39600107 DOI: 10.1021/acs.jafc.4c07209] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2024]
Abstract
Hyperuricemia is a metabolic disease characterized by an excessively increased level of uric acid (UA) in the blood, with an increasing prevalence and often associated with kidney damage. Gut microbiota and endotoxins of gut origin are key mediators in the gut-kidney axis that can cause renal impairment. The study was to reveal the protective effects of fucoidan on renal injury caused by hyperuricemia. The hyperuricemia model was established in C57BL/6J mice. After 10 weeks of fucoidan supplementation, we found that the levels of serum UA and creatinine were reduced, and the levels of renal tumor necrosis factor α, interleukin-18 (IL-18), IL-6, and interleukin-1β (IL-1β) were also decreased. Fucoidan inhibited the expressions of phosphorylated NF-κB p65, NLRP3, and activated caspase-1 in the kidneys. Fucoidan also regulated the expressions of Bcl-2 family proteins and decreased the activation of caspase-3, thereby exerting antiapoptotic effect. In addition, fucoidan could reduce the expressions of glucose transporter 9 (GLUT9) and urate transporter 1 (URAT1) proteins, thereby promoting the excretion of UA from the kidneys. Moreover, the protective effect of fucoidan on renal injury may be related to maintaining intestinal homeostasis. Fucoidan reduced serum lipopolysaccharide and improved the intestinal mucosal barrier function. Fucoidan decreased the abundances of Blautia, Muribaculaceae, and Dubosiella, and increased the abundances of Lactobacillus. High-dose fucoidan supplementation increased the content of butyric acid and enhanced the expression of ATP binding box transporter G2 (ABCG2) via the AMPK/AKT/CREB pathway in ileum. Conclusion: Fucoidan could protect against hyperuricemia-induced renal injury by inhibiting renal inflammation and apoptosis and modulating intestinal homeostasis in hyperuricemia mice.
Collapse
Affiliation(s)
- Meilan Xue
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Qingdao University, 308 Ningxia Road, Qingdao 266071, P. R. China
| | - Ronghuan Du
- Department of Human Nutrition, College of Public Health, Qingdao University, Qingdao 266071, P. R. China
| | - Yifan Zhou
- School of Traditional Chinese Medicine, Ningxia Medical University, Yinchuan 750004, P. R. China
| | - Yuhan Liu
- School of Biomedical Engineering, Sichuan University, Chengdu 610065, P. R. China
| | - Yingjie Tian
- Department of Human Nutrition, College of Public Health, Qingdao University, Qingdao 266071, P. R. China
| | - Yan Xu
- Department of Human Nutrition, College of Public Health, Qingdao University, Qingdao 266071, P. R. China
| | - Jiayi Yan
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Qingdao University, 308 Ningxia Road, Qingdao 266071, P. R. China
| | - Pengzhao Song
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Qingdao University, 308 Ningxia Road, Qingdao 266071, P. R. China
| | - Lu Wan
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Qingdao University, 308 Ningxia Road, Qingdao 266071, P. R. China
| | - Hongsen Xu
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Qingdao University, 308 Ningxia Road, Qingdao 266071, P. R. China
| | - Huaqi Zhang
- Department of Human Nutrition, College of Public Health, Qingdao University, Qingdao 266071, P. R. China
| | - Hui Liang
- Department of Human Nutrition, College of Public Health, Qingdao University, Qingdao 266071, P. R. China
| |
Collapse
|
9
|
Chen X, Li J, Liu P, Zhou Y, Zhang T, Li L, Shi J, Deng X, Sheng Y, Chen W, Wang D, Hu H. Inflammasome-Independent Mechanism of NLRP3 is Critical for Platelet GPIb-IX Function and Thrombosis. Thromb Haemost 2024; 124:1095-1113. [PMID: 38325399 DOI: 10.1055/a-2263-8372] [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: 02/09/2024]
Abstract
INTRODUCTION Platelets link thrombosis and inflammation, but how platelets handle the endogenous intraplatelet inflammatory machinery is less well understood. NACHT, LRR, and PYD domain-containing protein 3 (NLRP3) is the central component of the interleukin (IL)-1-producing inflammasome. Elucidating the cell type-specific mechanism of NLRP3 in platelets may improve our understanding of thrombotic diseases. METHODS Ferric chloride-induced mesenteric arteriole thrombosis models, tail bleeding models, and microfluidic whole-blood perfusion were used to study thrombosis and hemostasis. Additionally, we utilized aggregometry, flow cytometry, immunoprecipitation, and western blotting to investigate glycoprotein (GP)Ib-IX-mediated platelet function and signaling. RESULTS NLRP3-/- mice exhibited severely impaired thrombosis and hemostasis, whereas apoptosis-associated speck-like protein containing a CARD (ASC)-/-, caspase-1-/-, and Nlrp3 A350V/+ CrePF4 mice did not exhibit such changes. NLRP3-/- platelets exhibited reduced adhesion to injured vessel walls and collagen and impaired von Willebrand factor (vWF)-dependent translocation and rolling behavior. NLRP3 deficiency decreased botrocetin-induced platelet aggregation and the phosphorylation of key signaling molecules in the GPIb-IX pathway. Mechanistically, decreased cAMP/PKA activity led to reduced phosphorylation of NLRP3, thereby enabling the interaction between NLRP3 and filamin A. This interaction accelerated the dissociation of filamin A from GPIbα, which allowed a 14-3-3ζ-dependent increase in GPIb-IX affinity to vWF. Finally, platelet NLRP3 was found to largely regulate thrombotic disease models, such as models of stroke and deep vein thrombosis. CONCLUSION NLRP3 promoted the function of the major platelet adhesion receptor GPIb-IX without involving NLRP3 inflammasome assembly or IL-1β production.
Collapse
Affiliation(s)
- Xiaoyan Chen
- Department of Pathology and Pathophysiology and Bone Marrow Transplantation Center of the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, People's Republic of China
- Institute of Hematology, Zhejiang University & Zhejiang Engineering Laboratory for Stem Cell and Immunotherapy, Hangzhou, People's Republic of China
| | - Jingke Li
- Department of Pathology and Pathophysiology and Bone Marrow Transplantation Center of the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, People's Republic of China
- Institute of Hematology, Zhejiang University & Zhejiang Engineering Laboratory for Stem Cell and Immunotherapy, Hangzhou, People's Republic of China
| | - Pu Liu
- Department of Pathology of the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, People's Republic of China
| | - Yangfan Zhou
- Department of Pathology and Pathophysiology and Bone Marrow Transplantation Center of the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, People's Republic of China
- Institute of Hematology, Zhejiang University & Zhejiang Engineering Laboratory for Stem Cell and Immunotherapy, Hangzhou, People's Republic of China
| | - Tongtong Zhang
- Department of Hepatobiliary and Pancreatic Surgery, The Center for Integrated Oncology and Precision Medicine, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, People's Republic of China
| | - Li Li
- Department of Pathology and Pathophysiology and Bone Marrow Transplantation Center of the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, People's Republic of China
- Institute of Hematology, Zhejiang University & Zhejiang Engineering Laboratory for Stem Cell and Immunotherapy, Hangzhou, People's Republic of China
| | - Jingqi Shi
- Department of Pathology and Pathophysiology and Bone Marrow Transplantation Center of the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, People's Republic of China
- Institute of Hematology, Zhejiang University & Zhejiang Engineering Laboratory for Stem Cell and Immunotherapy, Hangzhou, People's Republic of China
| | - Xin Deng
- Department of Pathology and Pathophysiology and Bone Marrow Transplantation Center of the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, People's Republic of China
- Institute of Hematology, Zhejiang University & Zhejiang Engineering Laboratory for Stem Cell and Immunotherapy, Hangzhou, People's Republic of China
| | - Yilin Sheng
- Department of Pathology and Pathophysiology and Bone Marrow Transplantation Center of the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, People's Republic of China
- Institute of Hematology, Zhejiang University & Zhejiang Engineering Laboratory for Stem Cell and Immunotherapy, Hangzhou, People's Republic of China
| | - Wei Chen
- Department of Cell Biology of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, People's Republic of China
- Department of Cardiology of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, People's Republic of China
| | - Di Wang
- Institute of Immunology, Department of Orthopaedic Surgery of the Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, People's Republic of China
| | - Hu Hu
- Department of Pathology and Pathophysiology and Bone Marrow Transplantation Center of the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, People's Republic of China
- Institute of Hematology, Zhejiang University & Zhejiang Engineering Laboratory for Stem Cell and Immunotherapy, Hangzhou, People's Republic of China
- Key Laboratory of Disease Proteomics of Zhejiang Province, Hangzhou, People's Republic of China
| |
Collapse
|
10
|
Wang Z, Guo L, Yuan C, Zhu C, Li J, Zhong H, Mao P, Li J, Cui L, Dong J, Liu K, Meng X, Zhu G, Wang H. Staphylococcus pseudintermedius induces pyroptosis of canine corneal epithelial cells by activating the ROS-NLRP3 signalling pathway. Virulence 2024; 15:2333271. [PMID: 38515339 PMCID: PMC10984133 DOI: 10.1080/21505594.2024.2333271] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Accepted: 03/16/2024] [Indexed: 03/23/2024] Open
Abstract
Staphylococcus pseudintermedius (S. pseudintermedius) is a common pathogen that causes canine corneal ulcers. However, the pathogenesis remained unclear. In this study, it has been demonstrated that S. pseudintermedius invaded canine corneal epithelial cells (CCECs) intracellularly, mediating oxidative damage and pyroptosis by promoting the accumulation of intracellular reactive oxygen species (ROS) and activating the NLRP3 inflammasome. The canine corneal stroma was infected with S. pseudintermedius to establish the canine corneal ulcer model in vivo. The intracellular infectious model in CCECs was established in vitro to explore the mechanism of the ROS - NLRP3 signalling pathway during the S. pseudintermedius infection by adding NAC or MCC950. Results showed that the expression of NLRP3 and gasdermin D (GSDMD) proteins increased significantly in the infected corneas (p < 0.01). The intracellular infection of S. pseudintermedius was confirmed by transmission electron microscopy and immunofluorescent 3D imaging. Flow cytometry analysis revealed that ROS and pyroptosis rates increased in the experimental group in contrast to the control group (p < 0.01). Furthermore, NAC or MCC950 inhibited activation of the ROS - NLRP3 signalling pathway and pyroptosis rate significantly, by suppressing pro-IL-1β, cleaved-IL-1β, pro-caspase-1, cleaved-caspase-1, NLRP3, GSDMD, GSDMD-N, and HMGB1 proteins. Thus, the research confirmed that oxidative damage and pyroptosis were involved in the process of CCECs infected with S. pseudintermedius intracellularly by the ROS - NLRP3 signalling pathway. The results enrich the understanding of the mechanisms of canine corneal ulcers and facilitate the development of new medicines and prevention measures.
Collapse
Affiliation(s)
- Zhihao Wang
- College of Veterinary Medicine, Yangzhou University, Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, Jiangsu, China
- International Research Laboratory of Prevention and Control of Important Animal Infectious Diseases and Zoonotic Diseases of Jiangsu Higher Education Institutions, Yangzhou University, Yangzhou, China
- Joint International Research Laboratory of Agriculture and Agri-product Safety of the Ministry of Education, Yangzhou, Jiangsu, China
| | - Long Guo
- College of Veterinary Medicine, Yangzhou University, Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, Jiangsu, China
- International Research Laboratory of Prevention and Control of Important Animal Infectious Diseases and Zoonotic Diseases of Jiangsu Higher Education Institutions, Yangzhou University, Yangzhou, China
- Joint International Research Laboratory of Agriculture and Agri-product Safety of the Ministry of Education, Yangzhou, Jiangsu, China
| | - Changning Yuan
- College of Veterinary Medicine, Yangzhou University, Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, Jiangsu, China
- International Research Laboratory of Prevention and Control of Important Animal Infectious Diseases and Zoonotic Diseases of Jiangsu Higher Education Institutions, Yangzhou University, Yangzhou, China
- Joint International Research Laboratory of Agriculture and Agri-product Safety of the Ministry of Education, Yangzhou, Jiangsu, China
| | - Chengcheng Zhu
- College of Veterinary Medicine, Yangzhou University, Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, Jiangsu, China
- International Research Laboratory of Prevention and Control of Important Animal Infectious Diseases and Zoonotic Diseases of Jiangsu Higher Education Institutions, Yangzhou University, Yangzhou, China
- Joint International Research Laboratory of Agriculture and Agri-product Safety of the Ministry of Education, Yangzhou, Jiangsu, China
| | - Jun Li
- College of Veterinary Medicine, Yangzhou University, Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, Jiangsu, China
- International Research Laboratory of Prevention and Control of Important Animal Infectious Diseases and Zoonotic Diseases of Jiangsu Higher Education Institutions, Yangzhou University, Yangzhou, China
- Joint International Research Laboratory of Agriculture and Agri-product Safety of the Ministry of Education, Yangzhou, Jiangsu, China
| | - Haoran Zhong
- National Reference Laboratory for Animal Schistosomiasis, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai, China
| | - Peng Mao
- College of Veterinary Medicine, Yangzhou University, Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, Jiangsu, China
- International Research Laboratory of Prevention and Control of Important Animal Infectious Diseases and Zoonotic Diseases of Jiangsu Higher Education Institutions, Yangzhou University, Yangzhou, China
- Joint International Research Laboratory of Agriculture and Agri-product Safety of the Ministry of Education, Yangzhou, Jiangsu, China
| | - Jianji Li
- College of Veterinary Medicine, Yangzhou University, Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, Jiangsu, China
- International Research Laboratory of Prevention and Control of Important Animal Infectious Diseases and Zoonotic Diseases of Jiangsu Higher Education Institutions, Yangzhou University, Yangzhou, China
- Joint International Research Laboratory of Agriculture and Agri-product Safety of the Ministry of Education, Yangzhou, Jiangsu, China
| | - Luying Cui
- College of Veterinary Medicine, Yangzhou University, Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, Jiangsu, China
- International Research Laboratory of Prevention and Control of Important Animal Infectious Diseases and Zoonotic Diseases of Jiangsu Higher Education Institutions, Yangzhou University, Yangzhou, China
- Joint International Research Laboratory of Agriculture and Agri-product Safety of the Ministry of Education, Yangzhou, Jiangsu, China
| | - Junsheng Dong
- College of Veterinary Medicine, Yangzhou University, Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, Jiangsu, China
- International Research Laboratory of Prevention and Control of Important Animal Infectious Diseases and Zoonotic Diseases of Jiangsu Higher Education Institutions, Yangzhou University, Yangzhou, China
- Joint International Research Laboratory of Agriculture and Agri-product Safety of the Ministry of Education, Yangzhou, Jiangsu, China
| | - Kangjun Liu
- College of Veterinary Medicine, Yangzhou University, Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, Jiangsu, China
- International Research Laboratory of Prevention and Control of Important Animal Infectious Diseases and Zoonotic Diseases of Jiangsu Higher Education Institutions, Yangzhou University, Yangzhou, China
- Joint International Research Laboratory of Agriculture and Agri-product Safety of the Ministry of Education, Yangzhou, Jiangsu, China
| | - Xia Meng
- College of Veterinary Medicine, Yangzhou University, Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, Jiangsu, China
- International Research Laboratory of Prevention and Control of Important Animal Infectious Diseases and Zoonotic Diseases of Jiangsu Higher Education Institutions, Yangzhou University, Yangzhou, China
- Joint International Research Laboratory of Agriculture and Agri-product Safety of the Ministry of Education, Yangzhou, Jiangsu, China
| | - Guoqiang Zhu
- College of Veterinary Medicine, Yangzhou University, Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, Jiangsu, China
- International Research Laboratory of Prevention and Control of Important Animal Infectious Diseases and Zoonotic Diseases of Jiangsu Higher Education Institutions, Yangzhou University, Yangzhou, China
- Joint International Research Laboratory of Agriculture and Agri-product Safety of the Ministry of Education, Yangzhou, Jiangsu, China
| | - Heng Wang
- College of Veterinary Medicine, Yangzhou University, Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, Jiangsu, China
- International Research Laboratory of Prevention and Control of Important Animal Infectious Diseases and Zoonotic Diseases of Jiangsu Higher Education Institutions, Yangzhou University, Yangzhou, China
- Joint International Research Laboratory of Agriculture and Agri-product Safety of the Ministry of Education, Yangzhou, Jiangsu, China
| |
Collapse
|
11
|
Shang D, Zhao S. Molecular mechanisms of obesity predisposes to atopic dermatitis. Front Immunol 2024; 15:1473105. [PMID: 39564133 PMCID: PMC11574713 DOI: 10.3389/fimmu.2024.1473105] [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: 07/30/2024] [Accepted: 10/15/2024] [Indexed: 11/21/2024] Open
Abstract
Obesity is a prevalent metabolic disease that reduces bacterial diversity, colonizes the epidermis with lipophilic bacteria, and increases intestinal pro-inflammatory species, all of which lead to impaired epithelial barriers. Adipose tissue secretes immunomodulatory molecules, such as adipokines, leptin, and adiponectin, which alters the morphology of adipocytes and macrophages as well as modulates T cell differentiation and peripheral Th2-dominated immune responses. Atopic dermatitis (AD) and obesity have similar pathological manifestations, including inflammation as well as insulin and leptin resistance. This review examines the major mechanisms between obesity and AD, which focus on the effect on skin and gut microbiota, immune responses mediated by the toll like receptor (TLR) signaling pathway, and changes in cytokine levels (TNF-a, IL-6, IL-4, and IL13). Moreover, we describe the potential effects of adipokines on AD and finally mechanisms by which PPAR-γ suppresses and regulates type 2 immunity.
Collapse
Affiliation(s)
- Dajin Shang
- School of China Medical University, Shenyang, Liaoning, China
| | - Shengnan Zhao
- School of China Medical University, Shenyang, Liaoning, China
- Department of Dermatology, The First Hospital of China Medical University, Shenyang, China
| |
Collapse
|
12
|
Yu T, Luo L, Xue J, Tang W, Wu X, Yang F. Gut microbiota-NLRP3 inflammasome crosstalk in metabolic dysfunction-associated steatotic liver disease. Clin Res Hepatol Gastroenterol 2024; 48:102458. [PMID: 39233138 DOI: 10.1016/j.clinre.2024.102458] [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: 07/04/2024] [Revised: 08/20/2024] [Accepted: 08/30/2024] [Indexed: 09/06/2024]
Abstract
Nonalcoholic fatty liver disease (NAFLD) is a chronic liver disease associated with metabolic dysfunction, ranging from hepatic steatosis with or without mild inflammation to nonalcoholic steatohepatitis, which can rapidly progress to liver fibrosis and even liver cancer. In 2023, after several rounds of Delphi surveys, a new consensus recommended renaming NAFLD as metabolic dysfunction-associated steatotic liver disease (MASLD). Ninety-nine percent of NAFLD patients meet the new MASLD criteria related to metabolic cardiovascular risk factors under the "multiple parallel hits" of lipotoxicity, insulin resistance (IR), a proinflammatory diet, and an intestinal microbiota disorder, and previous research on NAFLD remains valid. The NLRP3 inflammasome, a well-known member of the pattern recognition receptor (PRR) family, can be activated by danger signals transmitted by pathogen-associated molecular patterns (PAMPs) and damage-associated molecular patterns (DAMPs), as well as cytokines involved in immune and inflammatory responses. The activation of the NLRP3 inflammasome pathway by MASLD triggers the production of the inflammatory cytokines IL-1β and IL-18. In MASLD, while changes in the composition and metabolites of the intestinal microbiota occur, the disrupted intestinal microbiota can also generate the inflammatory cytokines IL-1β and IL-18 by damaging the intestinal barrier, negatively regulating the liver on the gut-liver axis, and further aggravating MASLD. Therefore, modulating the gut-microbiota-liver axis through the NLRP3 inflammasome may emerge as a novel therapeutic approach for MASLD patients. In this article, we review the evidence regarding the functions of the NLRP3 inflammasome and the intestinal microbiota in MASLD, as well as their interactions in this disease.
Collapse
Affiliation(s)
- Tingting Yu
- School of Clinical Medical, Hubei University of Chinese Medicine, Wuhan 430000, PR China
| | - Lei Luo
- Department of Health Management Center, Hubei Provincial Hospital of Traditional Chinese Medicine, Wuhan 430070, PR China
| | - Juan Xue
- Department of Gastroenterology, Hubei Provincial Hospital of Integrated Chinese and Western Medicine, Wuhan 430015, PR China
| | - Wenqian Tang
- Department of Health Management Center, Hubei Provincial Hospital of Traditional Chinese Medicine, Wuhan 430070, PR China
| | - Xiaojie Wu
- School of Clinical Medical, Hubei University of Chinese Medicine, Wuhan 430000, PR China
| | - Fan Yang
- Department of Health Management Center, Hubei Provincial Hospital of Traditional Chinese Medicine, Wuhan 430070, PR China.
| |
Collapse
|
13
|
Xiao F, Yao H, Qian J, Huang J, Xia G. Dexmedetomidine improves mitophagy and pyroptosis through the ALKBH5/FUNDC1 axis during epidural-related maternal fever. Adv Med Sci 2024; 69:272-280. [PMID: 38815927 DOI: 10.1016/j.advms.2024.05.002] [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: 09/28/2023] [Revised: 04/30/2024] [Accepted: 05/27/2024] [Indexed: 06/01/2024]
Abstract
PURPOSE Epidural analgesia has emerged as a commonly used method for relieving labor pain. However, epidural-related maternal fever (ERMF) is characterized by a high occurrence rate and can have detrimental consequences for the well-being of both the mother and the fetus. This study aimed to investigate the functional role and underlying mechanism of dexmedetomidine (DEX) in ERMF. MATERIALS AND METHODS Ropivacaine (ROP)-induced human umbilical vein endothelial cells (HUVECs) were treated with DEX and/or transfected with ALKBH5 or FUNDC1 overexpression plasmid. qPCR and Western blot were adopted for mitophagy and pyroptosis marker protein detection. Autophagosomes were observed through electron microscopy, Caspase-1/PI double-positive cells were determined using flow cytometry. Inflammation-related factors were quantified using ELISA. The N6-methyladenosine (m6A) modification of FUNDC1 mRNA was examined using methylated RNA immunoprecipitation (MeRIP) and the binding between ALKBH5 and FUNDC1 mRNA was confirmed by RNA immunoprecipitation (RIP). RESULTS In ROP-induced HUVECs, there was a significant upregulation in ALKBH5 and FUNDC1, resulting in a notable increase in inflammation, pyroptosis, and mitophagy. The administration of DEX demonstrated the ability to alleviate ROP-induced pyroptosis and promote protective mitophagy. Interestingly, DEX treatment significantly reduced the interaction between ALKBH5 and FUNDC1 mRNA, while simultaneously increasing the m6A level of FUNDC1 mRNA in ROP-treated cells. Moreover, the overexpression of FUNDC1 partially reversed the effects of ALKBH5 overexpression on mitophagy and pyroptosis in HUVECs. CONCLUSIONS DEX can promote mitophagy and inhibit pyroptosis through the ALKBH5/FUNDC1 axis in ERMF, indicating its potential as a therapeutic strategy for clinical ERMF treatment.
Collapse
Affiliation(s)
- Fei Xiao
- Department of Anesthesia, Jiaxing University Affiliated Women and Children Hospital, Jiaxing, Zhejiang Province, PR China
| | - Hanqing Yao
- Department of Anesthesia, Jiaxing University Affiliated Women and Children Hospital, Jiaxing, Zhejiang Province, PR China
| | - Jing Qian
- Department of Anesthesia, Jiaxing University Affiliated Women and Children Hospital, Jiaxing, Zhejiang Province, PR China
| | - Jiayue Huang
- Department of Anesthesia, Jiaxing University Affiliated Women and Children Hospital, Jiaxing, Zhejiang Province, PR China
| | - Guangfa Xia
- Department of Breast Surgery, Jiaxing University Affiliated Women and Children Hospital, Jiaxing, Zhejiang Province, PR China.
| |
Collapse
|
14
|
Russell-Guzmán J, Américo-Da Silva L, Cadagan C, Maturana M, Palomero J, Estrada M, Barrientos G, Buvinic S, Hidalgo C, Llanos P. Activation of the ROS/TXNIP/NLRP3 pathway disrupts insulin-dependent glucose uptake in skeletal muscle of insulin-resistant obese mice. Free Radic Biol Med 2024; 222:187-198. [PMID: 38897422 DOI: 10.1016/j.freeradbiomed.2024.06.011] [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: 04/17/2024] [Revised: 05/31/2024] [Accepted: 06/15/2024] [Indexed: 06/21/2024]
Abstract
Oxidative stress and the activation of the nucleotide-binding domain, leucine-rich-containing family, pyrin domain containing 3 (NLRP3) inflammasome have been linked to insulin resistance in skeletal muscle. In immune cells, the exacerbated generation of reactive oxygen species (ROS) activates the NLRP3 inflammasome, by facilitating the interaction between thioredoxin interacting protein (TXNIP) and NLRP3. However, the precise role of ROS/TXNIP-dependent NLRP3 inflammasome activation in skeletal muscle during obesity-induced insulin resistance remains undefined. Here, we induced insulin resistance in C57BL/6J mice by feeding them for 8 weeks with a high-fat diet (HFD) and explored whether the ROS/TXNIP/NLRP3 pathway was involved in the induction of insulin resistance in skeletal muscle. Skeletal muscle fibers from insulin-resistant mice exhibited increased oxidative stress, as evidenced by elevated malondialdehyde levels, and altered peroxiredoxin 2 dimerization. Additionally, these fibers displayed augmented activation of the NLRP3 inflammasome, accompanied by heightened ROS-dependent proximity between TXNIP and NLRP3, which was abolished by the antioxidant N-acetylcysteine (NAC). Inhibition of the NLRP3 inflammasome with MCC950 or suppressing the ROS/TXNIP/NLRP3 pathway with NAC restored insulin-dependent glucose uptake in muscle fibers from insulin-resistant mice. These findings provide insights into the mechanistic link between oxidative stress, NLRP3 inflammasome activation, and obesity-induced insulin resistance in skeletal muscle.
Collapse
Affiliation(s)
- Javier Russell-Guzmán
- Institute for Research in Dental Sciences, Faculty of Dentistry, Universidad de Chile, Santiago, 8380544, Chile; Pedagogy in Physical Education, Faculty of Education, Universidad Autónoma de Chile, Santiago, 8910123, Chile
| | - Luan Américo-Da Silva
- Institute for Research in Dental Sciences, Faculty of Dentistry, Universidad de Chile, Santiago, 8380544, Chile
| | - Cynthia Cadagan
- Institute for Research in Dental Sciences, Faculty of Dentistry, Universidad de Chile, Santiago, 8380544, Chile
| | - Martín Maturana
- Institute for Research in Dental Sciences, Faculty of Dentistry, Universidad de Chile, Santiago, 8380544, Chile
| | - Jesús Palomero
- Department of Physiology and Pharmacology, Faculty of Medicine, Campus Miguel de Unamuno, Universidad de Salamanca, Salamanca, 37007, Spain
| | - Manuel Estrada
- Institute of Biomedical Sciences, Facultad de Medicina, Universidad de Chile, Santiago, 8380000, Chile
| | - Genaro Barrientos
- Institute of Biomedical Sciences, Facultad de Medicina, Universidad de Chile, Santiago, 8380000, Chile; Center for Exercise, Metabolism and Cancer, Facultad de Medicina, Universidad de Chile, Santiago, 8380453, Chile
| | - Sonja Buvinic
- Institute for Research in Dental Sciences, Faculty of Dentistry, Universidad de Chile, Santiago, 8380544, Chile; Center for Exercise, Metabolism and Cancer, Facultad de Medicina, Universidad de Chile, Santiago, 8380453, Chile
| | - Cecilia Hidalgo
- Institute of Biomedical Sciences, Facultad de Medicina, Universidad de Chile, Santiago, 8380000, Chile; Center for Exercise, Metabolism and Cancer, Facultad de Medicina, Universidad de Chile, Santiago, 8380453, Chile; Department of Neurosciences and Biomedical Neuroscience Institute, Universidad de Chile, Santiago, 8380453, Chile
| | - Paola Llanos
- Institute for Research in Dental Sciences, Faculty of Dentistry, Universidad de Chile, Santiago, 8380544, Chile; Center for Exercise, Metabolism and Cancer, Facultad de Medicina, Universidad de Chile, Santiago, 8380453, Chile.
| |
Collapse
|
15
|
Zhong H, Liu T, Shang Y, Huang C, Pan S. Breaking the vicious cycle: Targeting the NLRP3 inflammasome for treating sepsis-associated encephalopathy. Biomed Pharmacother 2024; 177:117042. [PMID: 39004064 DOI: 10.1016/j.biopha.2024.117042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Revised: 06/21/2024] [Accepted: 06/25/2024] [Indexed: 07/16/2024] Open
Abstract
Sepsis-associated encephalopathy (SAE) is a collection of clinical syndromes resulting from sepsis and characterized by widespread brain dysfunction. The high prevalence of SAE has adverse outcomes on the clinical management and prognosis of sepsis patients. However, currently, there are no effective treatments to ameliorate SAE. The pathogenesis of SAE is complex, including neuroinflammation and microglia activation, destruction of the blood-brain barrier (BBB), neurotransmitter dysfunction, cerebral metabolism and mitochondrial impairment, accumulation of amyloid beta and tauopathy, complement activation, among others. Furthermore, these mechanisms intertwine with each other, further complicating the comprehension of SAE. Among them, neuroinflammation mediated by hyperactivated microglia is considered the primary etiology of SAE. This instigates a detrimental cycle wherein BBB permeability escalates, facilitating direct damage to the central nervous system (CNS) by various neurotoxic substances. Activation of the NLRP3 inflammasome, situated within microglia, can be triggered by diverse danger signals, leading to cell pyroptosis, apoptosis, and tauopathy. These complex processes intricately regulate the onset and progression of neuroinflammation. In this review, we focus on elucidating the inhibitory regulatory mechanism of the NLRP3 inflammasome in microglia, which ultimately manifests as suppression of the inflammatory response. Our ultimate objective is to augment comprehension regarding the role of microglial NLRP3 inflammasome as we explore potential targets for therapeutic interventions against SAE.
Collapse
Affiliation(s)
- Hui Zhong
- Wuhan Jinyintan Hospital, Tongji Medical College of Huazhong University of Science and Technology, Wuhan Institute of Virology and Wuhan Jinyintan Hospital, Chinese Academy of Sciences, ,; Hubei Clinical Research Center for Infectious Diseases, ,; Wuhan Research Center for Communicable Disease Diagnosis and Treatment, Chinese Academy of Medical Sciences, ,; Joint Laboratory of Infectious Diseases and Health, Wuhan Institute of Virology and Wuhan Jinyintan Hospital, Chinese Academy of Sciences,
| | - Tianshu Liu
- Department of Ultrasound Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology,
| | - You Shang
- Department of Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology,
| | - Chaolin Huang
- Wuhan Jinyintan Hospital, Tongji Medical College of Huazhong University of Science and Technology, Wuhan Institute of Virology and Wuhan Jinyintan Hospital, Chinese Academy of Sciences, ,; Hubei Clinical Research Center for Infectious Diseases, ,; Wuhan Research Center for Communicable Disease Diagnosis and Treatment, Chinese Academy of Medical Sciences, ,; Joint Laboratory of Infectious Diseases and Health, Wuhan Institute of Virology and Wuhan Jinyintan Hospital, Chinese Academy of Sciences, ,.
| | - Shangwen Pan
- Department of Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, ,.
| |
Collapse
|
16
|
Chen Y, Zhao Y, Lu H, Zhang W, Gai Y, Niu G, Meng X, Lv H, Qian X, Ding X, Chen J. Protective effect of short-chain fructo-oligosaccharides from chicory on alcohol-induced injury in GES-1 cells via Keap1/Nrf2 and NLRP3 inflammasome signaling pathways. Front Nutr 2024; 11:1374579. [PMID: 38807640 PMCID: PMC11132183 DOI: 10.3389/fnut.2024.1374579] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Accepted: 04/09/2024] [Indexed: 05/30/2024] Open
Abstract
Numerous studies have demonstrated that polysaccharides derived from chicory possess the ability to regulate host signaling and modify mucosal damage. Yet, the effect and mechanism of short-chain fructo-oligosaccharides (scFOS) on gastric mucosa remain unclear. Hence, the protective effect of three scFOS (1-Kestose, Nystose, and 1F-Fructofuranosylnystose) against ethanol-induced injury in gastric epithelial (GES-1) cells, and the underlying molecular mechanism involved was investigated in this study. Treatment with 7% ethanol decreased the cell viability of GES-1 cells, resulting in oxidative stress and inflammation. However, pretreatment with scFOS exhibited significant improvements in cell viability, and mitigated oxidative stress and inflammation. scFOS markedly elevated the protein expression of Nrf2, HO-1, SOD1 and SOD2, while suppressing the expression of Keap1. scFOS pretreatment could also maintain mitochondrial membrane potential balance and reduce apoptosis. In addition, scFOS was observed to reduce the protein level of NLRP3, Caspase-1 and ASC. In conclusion, scFOS served a preventive function in mitigating oxidative stress and inflammation in ethanol-exposed GES-1 cells through modulation of the Keap1/Nrf2 and NLRP3 inflammasome signaling pathways. Collectively, the results indicated that scFOS could significantly mitigate ethanol-induced gastric cell damage, suggesting its potential for safeguarding gastrointestinal health.
Collapse
Affiliation(s)
- Yan Chen
- Jiangsu Key Laboratory for the Research and Utilization of Plant Resources, Institute of Botany, Jiangsu Province and Chinese Academy of Sciences, Nanjing, China
| | - Yanan Zhao
- Jiangsu Key Laboratory for the Research and Utilization of Plant Resources, Institute of Botany, Jiangsu Province and Chinese Academy of Sciences, Nanjing, China
| | - Hao Lu
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China
| | - Weichen Zhang
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China
| | - Yanan Gai
- Jiangsu Key Laboratory for the Research and Utilization of Plant Resources, Institute of Botany, Jiangsu Province and Chinese Academy of Sciences, Nanjing, China
| | - Guanting Niu
- Jiangsu Key Laboratory for the Research and Utilization of Plant Resources, Institute of Botany, Jiangsu Province and Chinese Academy of Sciences, Nanjing, China
| | - Xiuhua Meng
- Jiangsu Key Laboratory for the Research and Utilization of Plant Resources, Institute of Botany, Jiangsu Province and Chinese Academy of Sciences, Nanjing, China
| | - Han Lv
- Jiangsu Key Laboratory for the Research and Utilization of Plant Resources, Institute of Botany, Jiangsu Province and Chinese Academy of Sciences, Nanjing, China
| | - Xiaoguo Qian
- Fengning PingAn High-Tech Industrial Co., Ltd, Chengde, China
| | - Xiaoqin Ding
- Jiangsu Key Laboratory for the Research and Utilization of Plant Resources, Institute of Botany, Jiangsu Province and Chinese Academy of Sciences, Nanjing, China
| | - Jian Chen
- Jiangsu Key Laboratory for the Research and Utilization of Plant Resources, Institute of Botany, Jiangsu Province and Chinese Academy of Sciences, Nanjing, China
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China
| |
Collapse
|
17
|
Li N, Ma Y, Li C, Sun M, Qi F. Dexmedetomidine alleviates sevoflurane-induced neuroinflammation and neurocognitive disorders by suppressing the P2X4R/NLRP3 pathway in aged mice. Int J Neurosci 2024; 134:511-521. [PMID: 36066545 DOI: 10.1080/00207454.2022.2121921] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Revised: 08/25/2022] [Accepted: 09/01/2022] [Indexed: 10/14/2022]
Abstract
PURPOSE Microglia-mediated inflammation is associated with perioperative neurocognitive disorders (PNDs) caused by sevoflurane. Dexmedetomidine has been reported to protect against sevoflurane-induced cognitive impairment. In this study, we investigated the effects and underlying mechanisms of dexmedetomidine on sevoflurane-induced microglial neuroinflammation and PNDs. METHODS Wild-type and purinergic ionotropic 4 receptor (P2X4R) overexpressing C57/BL6 mice were intraperitoneally injected with 20 μg/kg dexmedetomidine or an equal volume of normal saline 2 h prior to sevoflurane exposure. The Morris water maze (MWM) test was performed to assess cognitive function. Immunofluorescence staining was employed to detect microglial activation. The expression levels of proinflammatory cytokines were measured by real-time quantitative PCR (qRT-PCR) and enzyme-linked immunosorbent assay (ELISA). The protein levels of P2X4R and NOD-like receptor protein 3 (NLRP3) were detected by Western Blotting. RESULTS Sevoflurane increased the number of microglia, upregulated the levels of proinflammatory cytokines, elevated the protein levels of P2X4R and NLRP3 in the hippocampus and induced cognitive decline, while pretreatment with dexmedetomidine downregulated the protein levels of P2X4R and NLRP3, alleviated sevoflurane-induced microglial neuroinflammation and improved cognitive dysfunction. Moreover, overexpression of P2X4R weakened the neuroprotective effect of dexmedetomidine. CONCLUSIONS Dexmedetomidine protected against sevoflurane-induced neuroinflammation and neurocognitive disorders by suppressing the P2X4R/NLRP3 pathway.
Collapse
Affiliation(s)
- Ning Li
- Department of Anesthesiology, Qilu Hospital of Shandong University, Jinan, China
- Department of Anesthesiology, The Second Hospital of Shandong University, Jinan, China
| | - Yufeng Ma
- Department of Anesthesiology, The Second Hospital of Shandong University, Jinan, China
| | - Chuangang Li
- Department of Anesthesiology, The Second Hospital of Shandong University, Jinan, China
| | - Manyi Sun
- Department of Anesthesiology, The Second Hospital of Shandong University, Jinan, China
| | - Feng Qi
- Department of Anesthesiology, Qilu Hospital of Shandong University, Jinan, China
| |
Collapse
|
18
|
Lou D, Fang Q, He Y, Ma R, Wang X, Li H, Qi M. Oxymatrine alleviates high-fat diet/streptozotocin-induced non-alcoholic fatty liver disease in C57BL/6 J mice by modulating oxidative stress, inflammation and fibrosis. Biomed Pharmacother 2024; 174:116491. [PMID: 38537582 DOI: 10.1016/j.biopha.2024.116491] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Revised: 03/12/2024] [Accepted: 03/19/2024] [Indexed: 05/01/2024] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) represents a complex complication of type 2 diabetes mellitus (T2DM). Oxymatrine (OMT) is an alkaloid extracted from Sophora flavescens with broad pharmacological effects. However, there is currently a lack of research on OMT in the field of NAFLD. The present study aimed to explore the effects and underlying mechanisms of oxymatrine in treating T2DM with NAFLD. The T2DM mice model was induced by high-fat diet (HFD) combined with streptozotocin (STZ) injection in male C57BL/6 J mice. Animals were randomly divided into four groups (n = 8): Control group, DC group, OMT-L group (45 mg/kg i.g.), and OMT-H group (90 mg/kg, i.g.). The drug was administered once a day for 8 weeks. In addition, HepG2 hepatocytes were incubated with palmitic acid (PA) to establish a fatty liver cell model. Treated with OMT, the body weight and fasting blood glucose (FBG) of DC mice were reduced and the liver organ coefficient was significantly optimized. Meanwhile, OMT markedly enhanced the activities of key antioxidant enzymes, including superoxide dismutase (SOD), catalase (CAT), and glutathione peroxidase (GSH-Px), and also reduced malondialdehyde (MDA) levels. These biochemical alterations were accompanied by noticeable improvements in liver histopathology. Furthermore, OMT down-regulated the expression of NOD-like receptor protein 3 (NLRP3), interleukin-1β (IL-1β), transforming growth factor-β1 (TGF-β1) and collagen I significantly, highlighting its potential in modulating inflammatory and fibrotic pathways. In conclusion, OMT improved liver impairment effectively in diabetic mice by suppressing oxidative stress, inflammation and fibrosis. These results suggest that OMT may represent a novel therapy for NAFLD with diabetes.
Collapse
Affiliation(s)
- Di Lou
- Institution of Pharmacology, College of Pharmaceutical Sciences, Zhejiang University of Technology, Hangzhou, Zhejiang 310014, China
| | - Qing Fang
- Institution of Pharmacology, College of Pharmaceutical Sciences, Zhejiang University of Technology, Hangzhou, Zhejiang 310014, China
| | - Yinghao He
- Institution of Pharmacology, College of Pharmaceutical Sciences, Zhejiang University of Technology, Hangzhou, Zhejiang 310014, China
| | - Ruyu Ma
- Institution of Pharmacology, College of Pharmaceutical Sciences, Zhejiang University of Technology, Hangzhou, Zhejiang 310014, China
| | - Xinyan Wang
- Institution of Pharmacology, College of Pharmaceutical Sciences, Zhejiang University of Technology, Hangzhou, Zhejiang 310014, China
| | - Hanbing Li
- Institution of Pharmacology, College of Pharmaceutical Sciences, Zhejiang University of Technology, Hangzhou, Zhejiang 310014, China.
| | - Minyou Qi
- Institution of Pharmacology, College of Pharmaceutical Sciences, Zhejiang University of Technology, Hangzhou, Zhejiang 310014, China.
| |
Collapse
|
19
|
Bakhshi A, Khani M, Alipour Parsa S, Khaheshi I, Namazi MH, Mazouri A, Bidram P, Safi M, Vakili H, Eslami V, Saadat H, Heidari L, Sohrabifar N. Investigating the expression level of miR-17-3p, miR-101-3p, miR-335-3p, and miR-296-3p in the peripheral blood of patients with acute myocardial infarction. Mol Cell Biochem 2024; 479:859-868. [PMID: 37222878 DOI: 10.1007/s11010-023-04766-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Accepted: 05/09/2023] [Indexed: 05/25/2023]
Abstract
The role of inflammation has been proven in acute myocardial infarction (AMI) pathogenesis. Due to the effect of NLRP3 gene expression in the inflammation process of MI, we aimed to explore the expression changes and diagnostic power of four inflammation-related miRNAs including miR-17-3p, miR-101-3p, miR-335-3p, miR-296-3p and their potential target, NLRP3, in ST-segment elevation myocardial infarction (STEMI), and non-STEMI (NSTEMI) patients as two major classes of AMI. The expression level of these genes were evaluated in 300 participants equally divided into three groups of STEMI, NSTEMI, and control using quantitative real-time PCR. The expression level of NLRP3 was upregulated in STEMI and NSTEMI patients compared to control subjects. Besides, the expression levels of miR-17-3p, miR-101-3p, and miR-296-3p were significantly downregulated in STEMI and NSTEMI patients compared to controls. The increased expression of NLRP3 had a very strong inverse correlation with miR-17-3p in patients with STEMI and with miR-101-3p in the STEMI and NSTEMI patients. ROC curve analysis showed that the expression level of miR-17-3p had the highest diagnostic power for discrimination between STEMI patients and controls. Remarkably, the combination of all markers resulted in a higher AUC. In summary, there is a significant association between the expression levels of miR-17-3p, miR-101-3p, miR-335-3p, miR-296-3p, and NLRP3 and the incidence of AMI. Although the miR-17-3p expression level has the highest diagnostic power to distinguish between STEMI patients and control subjects, the combination of these miRNAs and NLRP3 could serve as a novel potential diagnostic biomarker of STEMI.
Collapse
Affiliation(s)
- Alireza Bakhshi
- Cardiovascular Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohammad Khani
- Cardiovascular Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Saeed Alipour Parsa
- Cardiovascular Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Isa Khaheshi
- Cardiovascular Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohammad Hassan Namazi
- Cardiovascular Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Arash Mazouri
- Cardiovascular Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Peyman Bidram
- Cardiovascular Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Morteza Safi
- Cardiovascular Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Hossein Vakili
- Cardiovascular Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Vahid Eslami
- Cardiovascular Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Habib Saadat
- Cardiovascular Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Laleh Heidari
- Medical Genetic Department, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Nasim Sohrabifar
- Cardiovascular Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
20
|
Wang C, Chen Q, Chen S, Fan L, Gan Z, Zhao M, Shi L, Bin P, Yang G, Zhou X, Ren W. Serine synthesis sustains macrophage IL-1β production via NAD +-dependent protein acetylation. Mol Cell 2024; 84:744-759.e6. [PMID: 38266638 DOI: 10.1016/j.molcel.2024.01.002] [Citation(s) in RCA: 16] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Revised: 10/10/2023] [Accepted: 01/03/2024] [Indexed: 01/26/2024]
Abstract
Serine metabolism is involved in the fate decisions of immune cells; however, whether and how de novo serine synthesis shapes innate immune cell function remain unknown. Here, we first demonstrated that inflammatory macrophages have high expression of phosphoglycerate dehydrogenase (PHGDH, the rate-limiting enzyme of de novo serine synthesis) via nuclear factor κB signaling. Notably, the pharmacological inhibition or genetic modulation of PHGDH limits macrophage interleukin (IL)-1β production through NAD+ accumulation and subsequent NAD+-dependent SIRT1 and SIRT3 expression and activity. Mechanistically, PHGDH not only sustains IL-1β expression through H3K9/27 acetylation-mediated transcriptional activation of Toll-like receptor 4 but also supports IL-1β maturation via NLRP3-K21/22/24/ASC-K21/22/24 acetylation-mediated activation of the NLRP3 inflammasome. Moreover, mice with myeloid-specific depletion of Phgdh show alleviated inflammatory responses in lipopolysaccharide-induced systemic inflammation. This study reveals a network by which a metabolic enzyme, involved in de novo serine synthesis, mediates post-translational modifications and epigenetic regulation to orchestrate IL-1β production, providing a potential inflammatory disease target.
Collapse
Affiliation(s)
- Chuanlong Wang
- State Key Laboratory of Swine and Poultry Breeding Industry, Guangdong Laboratory of Lingnan Modern Agriculture, College of Animal Science, South China Agricultural University, Guangzhou 510642, China
| | - Qingyi Chen
- State Key Laboratory of Swine and Poultry Breeding Industry, Guangdong Laboratory of Lingnan Modern Agriculture, College of Animal Science, South China Agricultural University, Guangzhou 510642, China
| | - Siyuan Chen
- State Key Laboratory of Swine and Poultry Breeding Industry, Guangdong Laboratory of Lingnan Modern Agriculture, College of Animal Science, South China Agricultural University, Guangzhou 510642, China
| | - Lijuan Fan
- State Key Laboratory of Swine and Poultry Breeding Industry, Guangdong Laboratory of Lingnan Modern Agriculture, College of Animal Science, South China Agricultural University, Guangzhou 510642, China
| | - Zhending Gan
- State Key Laboratory of Swine and Poultry Breeding Industry, Guangdong Laboratory of Lingnan Modern Agriculture, College of Animal Science, South China Agricultural University, Guangzhou 510642, China
| | - Muyang Zhao
- State Key Laboratory of Swine and Poultry Breeding Industry, Guangdong Laboratory of Lingnan Modern Agriculture, College of Animal Science, South China Agricultural University, Guangzhou 510642, China
| | - Lexuan Shi
- State Key Laboratory of Swine and Poultry Breeding Industry, Guangdong Laboratory of Lingnan Modern Agriculture, College of Animal Science, South China Agricultural University, Guangzhou 510642, China
| | - Peng Bin
- State Key Laboratory of Swine and Poultry Breeding Industry, Guangdong Laboratory of Lingnan Modern Agriculture, College of Animal Science, South China Agricultural University, Guangzhou 510642, China
| | - Guan Yang
- Department of Infectious Diseases and Public Health, City University of Hong Kong, Kowloon, Hong Kong SAR 999077, China
| | - Xihong Zhou
- Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha 410125, China
| | - Wenkai Ren
- State Key Laboratory of Swine and Poultry Breeding Industry, Guangdong Laboratory of Lingnan Modern Agriculture, College of Animal Science, South China Agricultural University, Guangzhou 510642, China.
| |
Collapse
|
21
|
Sima Y, Wang X, Zhang L. Interaction of eosinophilic and neutrophilic inflammation in patients with chronic rhinosinusitis. Curr Opin Allergy Clin Immunol 2024; 24:25-31. [PMID: 37966141 DOI: 10.1097/aci.0000000000000956] [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: 11/16/2023]
Abstract
PURPOSE OF REVIEW In the past year, the endotype of chronic rhinosinusitis (CRS) has been studied from a new perspective. Eosinophilic and neutrophilic inflammation are not independent processes in the pathogenesis of CRS. In this review, we will focus on recent research on mixed eosinophilic-neutrophilic inflammation in CRS and discuss the mechanism and potential treatments. RECENT FINDINGS Traditionally, patients with eosinophilic CRS (ECRS) present with severe clinical manifestations, comorbidities, and a higher recurrence rate. Recent studies have found that approximately 40% of patients with ECRS present with neutrophilic infiltration, while patients with predominantly eosinophilic infiltration along with neutrophilic inflammation present with more complex inflammation, clinical manifestations and exhibit refractory characteristics. SUMMARY The complex inflammatory profile and refractory clinical characteristics of mixed eosinophilic-neutrophilic inflammation in CRS are current challenges for clinicians. We summarize the features of eosinophilic and neutrophilic inflammation and current studies on the mechanisms of mixed eosinophilic-neutrophilic inflammation and suggest potentially effective therapeutic methods. We hope that this review will help with determining precise treatment options for patients.
Collapse
Affiliation(s)
- Yutong Sima
- Department of Otolaryngology Head and Neck Surgery, Beijing Tongren Hospital, Capital Medical University
- Beijing Laboratory of Allergic Diseases, Beijing Municipal Education Commission and Beijing Key Laboratory of Nasal Diseases, Beijing Institute of Otolaryngology
| | - Xiangdong Wang
- Department of Otolaryngology Head and Neck Surgery, Beijing Tongren Hospital, Capital Medical University
- Beijing Laboratory of Allergic Diseases, Beijing Municipal Education Commission and Beijing Key Laboratory of Nasal Diseases, Beijing Institute of Otolaryngology
- Department of Allergy, Beijing Tongren Hospital, Capital Medical University
| | - Luo Zhang
- Department of Otolaryngology Head and Neck Surgery, Beijing Tongren Hospital, Capital Medical University
- Beijing Laboratory of Allergic Diseases, Beijing Municipal Education Commission and Beijing Key Laboratory of Nasal Diseases, Beijing Institute of Otolaryngology
- Department of Allergy, Beijing Tongren Hospital, Capital Medical University
- Research Unit of Diagnosis and Treatment of Chronic Nasal Diseases, Chinese Academy of Medical Sciences, Beijing, China
| |
Collapse
|
22
|
Liu P, Zhang Z, Chen H, Chen Q. Pyroptosis: Mechanisms and links with diabetic cardiomyopathy. Ageing Res Rev 2024; 94:102182. [PMID: 38182080 DOI: 10.1016/j.arr.2023.102182] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Revised: 12/27/2023] [Accepted: 12/30/2023] [Indexed: 01/07/2024]
Abstract
Diabetes mellitus (DM) is a chronic metabolic disease characterized by hyperglycaemia that seriously affects human health. Diabetic cardiomyopathy (DCM) is a major cardiovascular complication and one of the main causes of death in patients with DM. Although DCM attracts great attention, and new therapeutic methods are continuously developed, there is a lack of effective treatment strategies. Therefore, exploring and targeting new signalling pathways related to the evolution of DCM becomes a hotspot and difficulty in the prevention and treatment of DCM. Pyroptosis is a newly discovered regulated cell death that is heavily dependent on the formation of plasma membrane pores by members of the gasdermin protein family and is reported to be involved in the occurrence, development, and pathogenesis of DCM. In this review, we focus on the molecular mechanisms of pyroptosis, its involvement in the relevant signalling pathways of DCM, and potential pyroptosis-targeting therapeutic strategies for the treatment of DCM. Our review provides new insights into the use of pyroptosis as a useful tool for the prevention and treatment of DCM and clarifies future research directions.
Collapse
Affiliation(s)
- Pan Liu
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu 610072, Sichuan Province, PR China
| | - Zhengdong Zhang
- School of Clinical Medicine, Chengdu Medical College, Chengdu 610500, Sichuan Province, PR China; Department of Orthopedics, The First Affiliated Hospital of Chengdu Medical College, Chengdu 610500, Sichuan Province, PR China
| | - Huizhen Chen
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu 610072, Sichuan Province, PR China
| | - Qiu Chen
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu 610072, Sichuan Province, PR China.
| |
Collapse
|
23
|
Wang Y, Wang J, Tao SY, Liang Z, Xie R, Liu NN, Deng R, Zhang Y, Deng D, Jiang G. Mitochondrial damage-associated molecular patterns: A new insight into metabolic inflammation in type 2 diabetes mellitus. Diabetes Metab Res Rev 2024; 40:e3733. [PMID: 37823338 DOI: 10.1002/dmrr.3733] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Revised: 07/18/2023] [Accepted: 09/08/2023] [Indexed: 10/13/2023]
Abstract
The pathogenesis of diabetes is accompanied by increased levels of inflammatory factors, also known as "metabolic inflammation", which runs through the whole process of the occurrence and development of the disease. Mitochondria, as the key site of glucose and lipid metabolism, is often accompanied by mitochondrial function damage in type 2 diabetes mellitus (T2DM). Damaged mitochondria release pro-inflammatory factors through damage-related molecular patterns that activate inflammation pathways and reactions to oxidative stress, further aggravate metabolic disorders, and form a vicious circle. Currently, the pathogenesis of diabetes is still unclear, and clinical treatment focuses primarily on symptomatic intervention of the internal environment of disorders of glucose and lipid metabolism with limited clinical efficacy. The proinflammatory effect of mitochondrial damage-associated molecular pattern (mtDAMP) in T2DM provides a new research direction for exploring the pathogenesis and intervention targets of T2DM. Therefore, this review covers the most recent findings on the molecular mechanism and related signalling cascades of inflammation caused by mtDAMP in T2DM and discusses its pathogenic role of it in the pathological process of T2DM to search potential intervention targets.
Collapse
Affiliation(s)
- Yan Wang
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
- Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| | - Jingwu Wang
- Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| | - Si-Yu Tao
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | | | - Rong Xie
- Xinjiang Medical University, Urumqi, China
| | - Nan-Nan Liu
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Ruxue Deng
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Yuelin Zhang
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Deqiang Deng
- Department of Endocrinology, Urumqi Hospital of Traditional Chinese Medicine, Urumqi, China
| | - Guangjian Jiang
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| |
Collapse
|
24
|
Bae WJ, Shin D, Piao JJ, Kim S, Choi YS, Park BH, Jung HJ, Sorkhi S, Chawla S, Cheon CW, Kang DU, Choi JT, Park SH, Kim SW, Rajasekaran MR. Extracorporeal Shockwave Therapy Alleviates Inflammatory Pain by Down-Regulating NLRP3 Inflammasome in Experimental Chronic Prostatitis and Chronic Pelvic Pain Syndrome. World J Mens Health 2024; 42:157-167. [PMID: 37382279 PMCID: PMC10782125 DOI: 10.5534/wjmh.220241] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2022] [Revised: 01/15/2023] [Accepted: 02/06/2023] [Indexed: 06/30/2023] Open
Abstract
PURPOSE To evaluate the anti-inflammatory and antioxidative effects of extracorporeal shockwave therapy (ESWT) on prostatitis and explore the mechanism of alleviating pain. MATERIALS AND METHODS For in vitro testing, RWPE-1 cells were randomly divided into 5 groups: (1) RWPE-1 group (normal control), (2) LPS group (lipopolysaccharide inducing inflammation), (3) 0.1ESWT group (treated by 0.1 mJ/mm² energy level), (4) 0.2ESWT group (treated by 0.2 mJ/mm² energy level), and (5) 0.3ESWT group (treated by 0.3 mJ/mm² energy level). After ESWT was administered, cells and supernatant were collected for ELISA and western blot. For in vivo testing, Sprague-Dawley male rats were randomly divided into 3 groups: (1) normal group, (2) prostatitis group, and (3) ESWT group (n=12 for each). Prostatitis was induced by 17 beta-estradiol and dihydrotestosterone (DHT) administration. Four weeks after ESWT, the pain index was assessed for all groups and prostate tissues were collected for immunohistochemistry, immunofluorescence, apoptosis analysis and, western blot. RESULTS Our in vitro studies showed that the optimal energy flux density of ESWT was 0.2 mJ/mm². In vivo, ESWT ameliorated discomfort in rats with prostatitis and inflammation symptoms were improved. Compared to normal rats, overexpressed NLRP3 inflammasomes triggered apoptosis in rats with prostatitis and this was improved by ESWT. TLR4-NFκB pathway was overactive after experimental prostatitis, compared to normal and ESWT groups, and prostatitis induced alterations in BAX/BAK pathway were inhibited by ESWT. CONCLUSIONS ESWT improved CP/CPPS by reducing NLRP3 inflammasome and ameliorated apoptosis via inhibiting BAX/BAK pathway in a rat model. TLR4 may play a key role in bonding NLRP3 inflammasome and BAX/BAK pathways. ESWT might be a promising approach for the treatment of CP/CPPS.
Collapse
Affiliation(s)
- Woong Jin Bae
- Department of Urology, College of Medicine, The Catholic University of Korea, Seoul, Korea
- Catholic Integrative Medicine Research Institute, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Dongho Shin
- Department of Urology, College of Medicine, The Catholic University of Korea, Seoul, Korea
- Catholic Integrative Medicine Research Institute, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Jun Jie Piao
- Department of Urology, College of Medicine, The Catholic University of Korea, Seoul, Korea
- Catholic Integrative Medicine Research Institute, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Soomin Kim
- Department of Urology, College of Medicine, The Catholic University of Korea, Seoul, Korea
- Catholic Integrative Medicine Research Institute, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Yong Sun Choi
- Department of Urology, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Bong Hee Park
- Department of Urology, Uijeongbu St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Uijeongbu, Korea
| | - Hyun Jin Jung
- Department of Urology, Daegu Catholic University School of Medicine, Daegu, Korea
| | - Samuel Sorkhi
- Department of Urology, University of California San Diego Medical Center, San Diego, CA, USA
| | - Saager Chawla
- Department of Urology, University of California San Diego Medical Center, San Diego, CA, USA
| | | | | | | | - Sang-Hyuck Park
- Institute of Cannabis Research, Colorado State University-Pueblo, Pueblo, CO, USA
| | - Sae Woong Kim
- Department of Urology, College of Medicine, The Catholic University of Korea, Seoul, Korea
- Catholic Integrative Medicine Research Institute, College of Medicine, The Catholic University of Korea, Seoul, Korea
- Green Medicine Co., Ltd, Busan, Korea.
| | - Mahadevan Raj Rajasekaran
- Department of Urology, University of California San Diego Medical Center, San Diego, CA, USA
- San Diego Veterans Affairs Health Care System, San Diego, CA, USA.
| |
Collapse
|
25
|
Han J, Jia D, Yao H, Xu C, Huan Z, Jin H, Ge X. GRP78 improves the therapeutic effect of mesenchymal stem cells on hemorrhagic shock-induced liver injury: Involvement of the NF-кB and HO-1/Nrf-2 pathways. FASEB J 2024; 38:e23334. [PMID: 38050647 DOI: 10.1096/fj.202301456rrr] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Revised: 11/08/2023] [Accepted: 11/13/2023] [Indexed: 12/06/2023]
Abstract
Mesenchymal stem cells (MSCs) are a popular cell source for repairing the liver. Improving the survival rate and colonization time of MSCs may significantly improve the therapeutic outcomes of MSCs. Studies showed that 78-kDa glucose-regulated protein (GRP78) expression improves cell viability and migration. This study aims to examine whether GRP78 overexpression improves the efficacy of rat bone marrow-derived MSCs (rBMSCs) in HS-induced liver damage. Bone marrow was isolated from the femurs and tibias of rats. rBMSCs were transfected with a GFP-labeled GRP78 expression vector. Flow cytometry, transwell invasion assay, scratch assay immunoblotting, TUNEL assay, MTT assay, and ELISA were carried out. The results showed that GRP78 overexpression enhanced the migration and invasion of rBMSCs. Moreover, GRP78-overexpressing rBMSCs relieved liver damage, repressed liver oxidative stress, and inhibited apoptosis. We found that overexpression of GRP78 in rBMSCs inhibited activation of the NLRP3 inflammasome, significantly decreased the levels of inflammatory factors, and decreased the expression of CD68. Notably, GRP78 overexpression activated the Nrf-2/HO-1 pathway and inhibited the NF-κB pathway. High expression of GRP78 efficiently enhanced the effect of rBMSC therapy. GRP78 may be a potential target to improve the therapeutic efficacy of BMSCs.
Collapse
Affiliation(s)
- Jiahui Han
- Department of Critical Care Medicine, Wuxi 9th People's Hospital Affiliated to Soochow University, Wuxi, People's Republic of China
| | - Di Jia
- Department of Critical Care Medicine, Wuxi 9th People's Hospital Affiliated to Soochow University, Wuxi, People's Republic of China
| | - Hao Yao
- Department of Critical Care Medicine, Wuxi 9th People's Hospital Affiliated to Soochow University, Wuxi, People's Republic of China
| | - Ce Xu
- Department of Critical Care Medicine, Wuxi 9th People's Hospital Affiliated to Soochow University, Wuxi, People's Republic of China
| | - Zhirong Huan
- Department of Critical Care Medicine, Wuxi 9th People's Hospital Affiliated to Soochow University, Wuxi, People's Republic of China
| | - Hongdou Jin
- Department of General Surgery, Wuxi 9th People's Hospital Affiliated to Soochow University, Wuxi, People's Republic of China
| | - Xin Ge
- Department of Critical Care Medicine, Wuxi 9th People's Hospital Affiliated to Soochow University, Wuxi, People's Republic of China
- Orthopedic Institution of Wuxi City, Wuxi, People's Republic of China
| |
Collapse
|
26
|
Li X, Jin Y, Ding X, Zhu T, Wei C, Yao L. Long-term exercise training inhibits inflammation by suppressing hippocampal NLRP3 in APP/PS1 mice. SPORTS MEDICINE AND HEALTH SCIENCE 2023; 5:329-335. [PMID: 38314041 PMCID: PMC10831383 DOI: 10.1016/j.smhs.2023.09.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Revised: 09/03/2023] [Accepted: 09/21/2023] [Indexed: 02/06/2024] Open
Abstract
Behavioral experiments have demonstrated that long-term physical exercise can be beneficial for learning and memory dysfunction caused by neuroinflammation in Alzheimer's disease (AD). However, the molecular mechanism remains poorly understood due to a lack of sufficient pertinent biochemical evidence. We investigated the potential effect of long-term physical exercise on cognition and hippocampal gene and protein expression changes in a transgenic AD mouse model. Following twenty weeks of treadmill exercise, transgenic AD mice showed improvement in cognitive functions and downregulation of Nod-like receptor protein 3 (NLRP3) (p < 0.01), interleukin-1beta (IL-1β) (p < 0.05), and amyloid-β1-42 (Aβ1-42) (p < 0.05) expression levels. In addition, we observed significant reductions of microglial activation and hippocampal neuronal damage in the exercised AD mice (p < 0.01), which might be a result of the downregulation of NLRP3-mediated signaling and neuro-inflammatory responses. As neuronal damage due to inflammation might be a likely cause of AD-associated cognitive dysfunction. Our results suggested that the anti-inflammatory effects of exercise training involved downregulating the expression of key inflammatory factors and might play an important role in protecting hippocampal neurons against damage during the course of AD.
Collapse
Affiliation(s)
- Xue Li
- School of Sports Medicine and Health, Chengdu Sport University, Chengdu, Sichuan, China
| | - Yu Jin
- School of Sports Medicine and Health, Chengdu Sport University, Chengdu, Sichuan, China
| | - Xianyi Ding
- School of Sports Medicine and Health, Chengdu Sport University, Chengdu, Sichuan, China
| | - Tongyang Zhu
- School of Sports Medicine and Health, Chengdu Sport University, Chengdu, Sichuan, China
| | - Changling Wei
- School of Sports Medicine and Health, Chengdu Sport University, Chengdu, Sichuan, China
| | - Li Yao
- School of Sports Medicine and Health, Chengdu Sport University, Chengdu, Sichuan, China
| |
Collapse
|
27
|
Li A, Guan L, Su W, Zhao N, Song X, Wang J, Tang X, Li W, Jiao X. TXNIP inhibition in the treatment of type 2 diabetes mellitus: design, synthesis, and biological evaluation of quinazoline derivatives. J Enzyme Inhib Med Chem 2023; 38:2166937. [PMID: 36651294 PMCID: PMC9858527 DOI: 10.1080/14756366.2023.2166937] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
Thioredoxin interacting protein (TXNIP) is a potential drug target for type 2 diabetes mellitus (T2DM) treatment. A series of quinazoline derivatives were designed, synthesised, and evaluated to inhibit TXNIP expression and protect from palmitate (PA)-induced β cell injury. In vitro cell viability assay showed that compounds D-2 and C-1 could effectively protect β cell from PA-induced apoptosis, and subsequent results showed that these two compounds decreased TXNIP expression by accelerating its protein degradation. Mechanistically, compounds D-2 and C-1 reduced intracellular reactive oxygen species (ROS) production and modulated TXNIP-NLRP3 inflammasome signalling, and thus alleviating oxidative stress injury and inflammatory response under PA insult. Besides, these two compounds were predicted to possess better drug-likeness properties using SwissADME. The present study showed that compounds D-2 and C-1, especially compound D-2, were potent pancreatic β cell protective agents to inhibit TXNIP expression and might serve as promising lead candidates for the treatment of T2DM.
Collapse
Affiliation(s)
- Aiyun Li
- Key Laboratory of Cellular Physiology (Shanxi Medical University), Ministry of Education, and the Department of Physiology, Shanxi Medical University, Taiyuan, PR China
| | - Li Guan
- College of Pharmacy, Xi’an Medical University, Xi’an, PR China
| | - Wanzhen Su
- Key Laboratory of Cellular Physiology (Shanxi Medical University), Ministry of Education, and the Department of Physiology, Shanxi Medical University, Taiyuan, PR China
| | - Ning Zhao
- College of Pharmacy, Xi’an Medical University, Xi’an, PR China
| | - Xuwen Song
- College of Pharmacy, Xi’an Medical University, Xi’an, PR China
| | - Jin Wang
- Key Laboratory of Cellular Physiology (Shanxi Medical University), Ministry of Education, and the Department of Physiology, Shanxi Medical University, Taiyuan, PR China
| | - Xiaoxiao Tang
- College of Pharmacy, Xi’an Medical University, Xi’an, PR China
| | - Weize Li
- College of Pharmacy, Xi’an Medical University, Xi’an, PR China,CONTACT Xiangying Jiao Key Laboratory of Cellular Physiology (Shanxi Medical University), Ministry of Education, and the Department of Physiology, Shanxi Medical University, Taiyuan030001, PR China
| | - Xiangying Jiao
- Key Laboratory of Cellular Physiology (Shanxi Medical University), Ministry of Education, and the Department of Physiology, Shanxi Medical University, Taiyuan, PR China,Weize Li College of Pharmacy, Xi’an Medical University, Xi’an710021, PR China
| |
Collapse
|
28
|
Xie D, Guo H, Li M, Jia L, Zhang H, Liang D, Wu N, Yang Z, Tian Y. Splenic monocytes mediate inflammatory response and exacerbate myocardial ischemia/reperfusion injury in a mitochondrial cell-free DNA-TLR9-NLRP3-dependent fashion. Basic Res Cardiol 2023; 118:44. [PMID: 37814087 DOI: 10.1007/s00395-023-01014-0] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Revised: 09/22/2023] [Accepted: 09/23/2023] [Indexed: 10/11/2023]
Abstract
The spleen contributes importantly to myocardial ischemia/reperfusion (MI/R) injury. Nucleotide-binding oligomerization domain-like receptor family pyrin domain containing 3 (NLRP3) recruits inflammasomes, initiating inflammatory responses and mediating tissue injury. We hypothesize that myocardial cell-free DNA (cfDNA) activates the splenic NLRP3 inflammasome during early reperfusion, increases systemic inflammatory response, and exacerbates myocardial infarct. Mice were subjected to 40 min of ischemia followed by 0, 1, 5, or 15 min, or 24 h of reperfusion. Splenic leukocyte adoptive transfer was performed by injecting isolated splenocytes to mice with splenectomy performed prior to left coronary artery occlusion. CY-09 (4 mg/kg) was administered 5 min before reperfusion. During post-ischemic reperfusion, splenic protein levels of NLRP3, cleaved caspase-1, and interleukin-1β (IL-1β) were significantly elevated and peaked (2.1 ± 0.2-, 3.4 ± 0.4-, and 3.2 ± 0.2-fold increase respectively, p < 0.05) within 5 min of reperfusion. In myocardial tissue, NLRP3 was not upregulated until 24 h after reperfusion. Suppression by CY09, a specific NLRP3 inflammasome inhibitor, or deficiency of NLRP3 significantly reduced myocardial infarct size (17.3% ± 4.2% and 33.2% ± 1.8% decrease respectively, p < 0.01). Adoptive transfer of NLRP3-/- splenocytes to WT mice significantly decreased infarct size compared to transfer of WT splenocytes (19.1% ± 2.8% decrease, p < 0.0001). NLRP3 was mainly activated at 5 min after reperfusion in CD11b+ and LY6G- splenocytes, which significantly increased during reperfusion (24.8% ± 0.7% vs.14.3% ± 0.6%, p < 0.0001). The circulating cfDNA level significantly increased in patients undergoing cardiopulmonary bypass (CPB) (43.3 ± 5.3 ng/mL, compared to pre-CPB 23.8 ± 3.5 ng/mL, p < 0.01). Mitochondrial cfDNA (mt-cfDNA) contributed to NLRP3 activation in macrophages (2.1 ± 0.2-fold increase, p < 0.01), which was inhibited by a Toll-like receptor 9(TLR9) inhibitor. The NLRP3 inflammasome in splenic monocytes is activated and mediates the inflammatory response shortly after reperfusion onset, exacerbating MI/R injury in mt-cfDNA/TLR9-dependent fashion. The schema reveals splenic NLRP3 mediates the inflammatory response in macrophages and exacerbates MI/R in a mitochondrial cfDNA/ TLR9-dependent fashion.
Collapse
Affiliation(s)
- Dina Xie
- Department of Cardiovascular Surgery, Tianjin Medical University General Hospital, Tianjin, 300052, China
| | - Hanliang Guo
- Department of Cardiovascular Surgery, Tianjin Medical University General Hospital, Tianjin, 300052, China
| | - Mingbiao Li
- Department of Cardiovascular Surgery, Tianjin Medical University General Hospital, Tianjin, 300052, China
| | - Liqun Jia
- Department of Cardiovascular Surgery, Tianjin Medical University General Hospital, Tianjin, 300052, China
| | - Hao Zhang
- Department of Cardiovascular Surgery, Tianjin Medical University General Hospital, Tianjin, 300052, China
| | - Degang Liang
- Department of Cardiovascular Surgery, Tianjin Medical University General Hospital, Tianjin, 300052, China
| | - Naishi Wu
- Department of Cardiovascular Surgery, Tianjin Medical University General Hospital, Tianjin, 300052, China
| | - Zequan Yang
- Department of Surgery, University of Virginia, Charlottesville, VA, USA
| | - Yikui Tian
- Department of Cardiovascular Surgery, Tianjin Medical University General Hospital, Tianjin, 300052, China.
- Tianjin Key Laboratory of Lung Cancer Metastasis and Tumor Microenvironment, Tianjin Lung Cancer Institute, Tianjin Medical University General Hospital, Tianjin, 300052, China.
| |
Collapse
|
29
|
Treasure K, Harris J, Williamson G. Exploring the anti-inflammatory activity of sulforaphane. Immunol Cell Biol 2023; 101:805-828. [PMID: 37650498 DOI: 10.1111/imcb.12686] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Revised: 07/24/2023] [Accepted: 08/10/2023] [Indexed: 09/01/2023]
Abstract
Dysregulation of innate immune responses can result in chronic inflammatory conditions. Glucocorticoids, the current frontline therapy, are effective immunosuppressive drugs but come with a trade-off of cumulative and serious side effects. Therefore, alternative drug options with improved safety profiles are urgently needed. Sulforaphane, a phytochemical derived from plants of the brassica family, is a potent inducer of phase II detoxification enzymes via nuclear factor-erythroid factor 2-related factor 2 (NRF2) signaling. Moreover, a growing body of evidence suggests additional diverse anti-inflammatory properties of sulforaphane through interactions with mediators of key signaling pathways and inflammatory cytokines. Multiple studies support a role for sulforaphane as a negative regulator of nuclear factor kappa-light chain enhancer of activated B cells (NF-κB) activation and subsequent cytokine release, inflammasome activation and direct regulation of the activity of macrophage migration inhibitory factor. Significantly, studies have also highlighted potential steroid-sparing activity for sulforaphane, suggesting that it may have potential as an adjunctive therapy for some inflammatory conditions. This review discusses published research on sulforaphane, including proposed mechanisms of action, and poses questions for future studies that might help progress our understanding of the potential clinical applications of this intriguing molecule.
Collapse
Affiliation(s)
- Katie Treasure
- Department of Nutrition, Dietetics and Food, School of Clinical Sciences at Monash Health, Faculty of Medicine, Nursing and Health Sciences, Monash University, Clayton, VIC, Australia
- Victorian Heart Hospital, Monash University, Clayton, VIC, Australia
| | - James Harris
- Biomedical Manufacturing, Commonwealth Scientific and Industrial Research Organisation (CSIRO), Clayton, VIC, Australia
- Centre for Inflammatory Diseases, School of Clinical Sciences at Monash Health, Faculty of Medicine, Nursing and Health Sciences, Monash University, Clayton, VIC, Australia
| | - Gary Williamson
- Department of Nutrition, Dietetics and Food, School of Clinical Sciences at Monash Health, Faculty of Medicine, Nursing and Health Sciences, Monash University, Clayton, VIC, Australia
- Victorian Heart Hospital, Monash University, Clayton, VIC, Australia
| |
Collapse
|
30
|
Qin Y, Zhao W. Posttranslational modifications of NLRP3 and their regulatory roles in inflammasome activation. Eur J Immunol 2023; 53:e2350382. [PMID: 37382218 DOI: 10.1002/eji.202350382] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2023] [Revised: 05/09/2023] [Accepted: 06/19/2023] [Indexed: 06/30/2023]
Abstract
The NACHT, LRR, and PYD domains-containing protein 3 (NLRP3) inflammasome is a multimolecular complex that plays a fundamental role in inflammation. Optimal activation of NLRP3 inflammasome is crucial for host defense against pathogens and the maintenance of immune homeostasis. Aberrant NLRP3 inflammasome activity has been implicated in various inflammatory diseases. Posttranslational modifications (PTMs) of NLRP3, a key inflammasome sensor, play critical roles in directing inflammasome activation and controlling the severity of inflammation and inflammatory diseases, such as arthritis, peritonitis, inflammatory bowel disease, atherosclerosis, and Parkinson's disease. Various NLRP3 PTMs, including phosphorylation, ubiquitination, and SUMOylation, could direct inflammasome activation and control inflammation severity by affecting the protein stability, ATPase activity, subcellular localization, and oligomerization of NLRP3 as well as the association between NLRP3 and other inflammasome components. Here, we provide an overview of the PTMs of NLRP3 and their roles in controlling inflammation and summarize potential anti-inflammatory drugs targeting NLRP3 PTMs.
Collapse
Affiliation(s)
- Ying Qin
- Department of Pathogenic Biology and Key Laboratory of Infection and Immunity of Shandong Province, School of Basic Medical Science, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Wei Zhao
- Department of Pathogenic Biology and Key Laboratory of Infection and Immunity of Shandong Province, School of Basic Medical Science, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| |
Collapse
|
31
|
Chen Y, Miao C, Zhao Y, Yang L, Wang R, Shen D, Ren N, Zhang Q. Inflammasomes in human reproductive diseases. Mol Hum Reprod 2023; 29:gaad035. [PMID: 37788097 DOI: 10.1093/molehr/gaad035] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2023] [Revised: 09/20/2023] [Indexed: 10/05/2023] Open
Abstract
Inflammasomes are multi-protein complexes localized within immune and non-immune cells that induce caspase activation, proinflammatory cytokine secretion, and ultimately pyroptosis-a type of cell death. Inflammasomes are involved in a variety of human diseases, especially acute or chronic inflammatory diseases. In this review, we focused on the strong correlation between the NLRP3 inflammasome and various reproductive diseases, including ovarian aging or premature ovarian insufficiency, PCOS, endometriosis, recurrent spontaneous abortion, preterm labor, pre-eclampsia, and male subfertility, as well as the multifaceted role of NLRP3 in the pathogenesis and treatment of these diseases. In addition, we provide an overview of the structure and amplification of inflammasomes. This comprehensive review demonstrates the vital role of NLRP3 inflammasome activation in human reproductive diseases together with the underlying mechanisms, offers new insights for mechanistic studies of reproduction, and provides promising possibilities for the development of drugs targeting the NLRP3 inflammasome for the treatment of reproductive disorders in the future.
Collapse
Affiliation(s)
- Yun Chen
- Department of TCM Gynecology, Hangzhou TCM Hospital affiliated to Zhejiang Chinese Medical University, Hangzhou, China
| | - Chenyun Miao
- Department of TCM Gynecology, Hangzhou TCM Hospital affiliated to Zhejiang Chinese Medical University, Hangzhou, China
| | - Ying Zhao
- Department of TCM Gynecology, Hangzhou TCM Hospital affiliated to Zhejiang Chinese Medical University, Hangzhou, China
| | - Liuqing Yang
- Department of TCM Gynecology, Hangzhou TCM Hospital affiliated to Zhejiang Chinese Medical University, Hangzhou, China
| | - Ruye Wang
- Department of TCM Gynecology, Hangzhou TCM Hospital affiliated to Zhejiang Chinese Medical University, Hangzhou, China
| | - Dan Shen
- Department of TCM Gynecology, Hangzhou TCM Hospital affiliated to Zhejiang Chinese Medical University, Hangzhou, China
| | - Ning Ren
- Department of TCM Gynecology, Hangzhou TCM Hospital affiliated to Zhejiang Chinese Medical University, Hangzhou, China
| | - Qin Zhang
- Department of TCM Gynecology, Hangzhou TCM Hospital affiliated to Zhejiang Chinese Medical University, Hangzhou, China
| |
Collapse
|
32
|
Lamouroux A, Tournier M, Iaculli D, Caufriez A, Rusiecka OM, Martin C, Bes V, Carpio LE, Girardin Y, Loris R, Tabernilla A, Molica F, Gozalbes R, Mayán MD, Vinken M, Kwak BR, Ballet S. Structure-Based Design and Synthesis of Stapled 10Panx1 Analogues for Use in Cardiovascular Inflammatory Diseases. J Med Chem 2023; 66:13086-13102. [PMID: 37703077 PMCID: PMC10544015 DOI: 10.1021/acs.jmedchem.3c01116] [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: 06/20/2023] [Indexed: 09/14/2023]
Abstract
Following a rational design, a series of macrocyclic ("stapled") peptidomimetics of 10Panx1, the most established peptide inhibitor of Pannexin1 (Panx1) channels, were developed and synthesized. Two macrocyclic analogues SBL-PX1-42 and SBL-PX1-44 outperformed the linear native peptide. During in vitro adenosine triphosphate (ATP) release and Yo-Pro-1 uptake assays in a Panx1-expressing tumor cell line, both compounds were revealed to be promising bidirectional inhibitors of Panx1 channel function, able to induce a two-fold inhibition, as compared to the native 10Panx1 sequence. The introduction of triazole-based cross-links within the peptide backbones increased helical content and enhanced in vitro proteolytic stability in human plasma (>30-fold longer half-lives, compared to 10Panx1). In adhesion assays, a "double-stapled" peptide, SBL-PX1-206 inhibited ATP release from endothelial cells, thereby efficiently reducing THP-1 monocyte adhesion to a TNF-α-activated endothelial monolayer and making it a promising candidate for future in vivo investigations in animal models of cardiovascular inflammatory disease.
Collapse
Affiliation(s)
- Arthur Lamouroux
- Research
Group of Organic Chemistry, Departments of Chemistry and Bioengineering
Sciences, Vrije Universiteit Brussel, Pleinlaan 2, B-1050 Brussels, Belgium
| | - Malaury Tournier
- Department
of Pathology and Immunology and Geneva Center for Inflammation Research,
Faculty of Medicine, University of Geneva, Rue Michel-Servet 1, CH-1211 Geneva, Switzerland
| | - Debora Iaculli
- Research
Group of Organic Chemistry, Departments of Chemistry and Bioengineering
Sciences, Vrije Universiteit Brussel, Pleinlaan 2, B-1050 Brussels, Belgium
| | - Anne Caufriez
- Research
Group of Organic Chemistry, Departments of Chemistry and Bioengineering
Sciences, Vrije Universiteit Brussel, Pleinlaan 2, B-1050 Brussels, Belgium
- Research
Unit of In Vitro Toxicology and Dermato-Cosmetology, Department of
Pharmaceutical Sciences, Vrije Universiteit
Brussel, Laarbeeklaan
103, 1090 Brussels, Belgium
| | - Olga M. Rusiecka
- Department
of Pathology and Immunology and Geneva Center for Inflammation Research,
Faculty of Medicine, University of Geneva, Rue Michel-Servet 1, CH-1211 Geneva, Switzerland
| | - Charlotte Martin
- Research
Group of Organic Chemistry, Departments of Chemistry and Bioengineering
Sciences, Vrije Universiteit Brussel, Pleinlaan 2, B-1050 Brussels, Belgium
| | - Viviane Bes
- Department
of Pathology and Immunology and Geneva Center for Inflammation Research,
Faculty of Medicine, University of Geneva, Rue Michel-Servet 1, CH-1211 Geneva, Switzerland
| | - Laureano E. Carpio
- ProtoQSAR
SL, Centro Europeo de Empresas Innovadoras, Parque Tecnológico de Valencia, Avda. Benjamin Franklin 12, 46980 Paterna, Spain
| | - Yana Girardin
- Structural
Biology Brussels, Department of Biotechnology, Vrije Universiteit Brussel, Pleinlaan 2, B-1050 Brussels, Belgium
- Centre for
Structural Biology, VIB, Pleinlaan 2, 1050 Brussels, Belgium
| | - Remy Loris
- Structural
Biology Brussels, Department of Biotechnology, Vrije Universiteit Brussel, Pleinlaan 2, B-1050 Brussels, Belgium
- Centre for
Structural Biology, VIB, Pleinlaan 2, 1050 Brussels, Belgium
| | - Andrés Tabernilla
- Research
Unit of In Vitro Toxicology and Dermato-Cosmetology, Department of
Pharmaceutical Sciences, Vrije Universiteit
Brussel, Laarbeeklaan
103, 1090 Brussels, Belgium
| | - Filippo Molica
- Department
of Pathology and Immunology and Geneva Center for Inflammation Research,
Faculty of Medicine, University of Geneva, Rue Michel-Servet 1, CH-1211 Geneva, Switzerland
| | - Rafael Gozalbes
- ProtoQSAR
SL, Centro Europeo de Empresas Innovadoras, Parque Tecnológico de Valencia, Avda. Benjamin Franklin 12, 46980 Paterna, Spain
- MolDrug
AI Systems SL, c/Olimpia
Arozena 45, 46018 Valencia, Spain
| | - María D. Mayán
- CellCOM
Research Group, Instituto de Investigación Biomédica
de A Coruña, Servizo Galego de Saúde, Universidade da Coruña, 15071 A Coruña, Spain
| | - Mathieu Vinken
- Research
Unit of In Vitro Toxicology and Dermato-Cosmetology, Department of
Pharmaceutical Sciences, Vrije Universiteit
Brussel, Laarbeeklaan
103, 1090 Brussels, Belgium
| | - Brenda R. Kwak
- Department
of Pathology and Immunology and Geneva Center for Inflammation Research,
Faculty of Medicine, University of Geneva, Rue Michel-Servet 1, CH-1211 Geneva, Switzerland
| | - Steven Ballet
- Research
Group of Organic Chemistry, Departments of Chemistry and Bioengineering
Sciences, Vrije Universiteit Brussel, Pleinlaan 2, B-1050 Brussels, Belgium
| |
Collapse
|
33
|
Yu W, Jiang W, Wu W, Wang G, Zhao D, Yan C, Lin P. Combining idebenone and rosuvastatin prevents atherosclerosis by suppressing oxidative stress and NLRP3 inflammasome activation. Eur J Pharmacol 2023; 955:175911. [PMID: 37451421 DOI: 10.1016/j.ejphar.2023.175911] [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: 02/12/2023] [Revised: 06/28/2023] [Accepted: 07/11/2023] [Indexed: 07/18/2023]
Abstract
Atherosclerosis is a progressive inflammatory disease activated by excessive oxidized low-density lipoprotein (ox-LDL). Statins are the first-line choice to reduce the risk of cardiovascular disease. However, statin-associated side effects prompt dose reduction or discontinuation. Idebenone could protect against atherosclerosis by scavenging reactive oxygen species (ROS). Although both idebenone and statins have certain efficacy, neither of them can achieve a completely satisfactory effect. Here, we aim to investigate the anti-atherosclerotic effect of the combination of idebenone and statins. Apolipoprotein E knockout (ApoE-/-) mice were given idebenone (400 mg/kg/d), rosuvastatin (10 mg/kg/d) or a combination of idebenone and rosuvastatin. Histological and immunohistochemical staining were used to analyze the size and composition of the plaque. In vivo and in vitro experiments were conducted to explore the possible mechanism. Idebenone and rosuvastatin both reduced plaque burden and increased the stability of atherosclerotic plaques in the ApoE-/- mice. Mice receiving the combination therapy had even reduced and more stable atherosclerotic plaques than mice treated with idebenone or rosuvastatin alone. NLRP3 and IL-1β were further downregulated in mice receiving combination therapy compared with mice treated with monotherapy. The combination treatment also markedly reduced oxidative stress and cell apoptosis in vivo and in vitro. In conclusion, our data demonstrate that the combination of idebenone and rosuvastatin works synergistically to inhibit atherosclerosis, and that the use of both substances together is more effective than using either substance alone. From a therapeutic point, combining idebenone and rosuvastatin appears to be a promising strategy to further prevent atherosclerosis.
Collapse
Affiliation(s)
- Wenfei Yu
- Department of Neurology, Qilu Hospital of Shandong University, Jinan, 250012, Shandong Province, China; University of Health and Rehabilitation Sciences, No. 17, Shandong Road, Shinan District, Qingdao City, Shandong Province, China
| | - Wei Jiang
- Department of Neurology, The Third Affiliated Hospital of Sun Yat-Sen University, No. 600 Tianhe Road, Guangzhou, 510630, Guangdong Province, China
| | - Wenjing Wu
- Department of Neurology, Qilu Hospital of Shandong University, Jinan, 250012, Shandong Province, China
| | - Guangyu Wang
- Department of Neurology, Qilu Hospital of Shandong University, Jinan, 250012, Shandong Province, China
| | - Dandan Zhao
- Department of Neurology, Qilu Hospital of Shandong University, Jinan, 250012, Shandong Province, China
| | - Chuanzhu Yan
- Department of Neurology, Qilu Hospital of Shandong University, Jinan, 250012, Shandong Province, China.
| | - Pengfei Lin
- Department of Neurology, Qilu Hospital of Shandong University, Jinan, 250012, Shandong Province, China.
| |
Collapse
|
34
|
Su WJ, Li JM, Zhang T, Cao ZY, Hu T, Zhong SY, Xu ZY, Gong H, Jiang CL. Microglial NLRP3 inflammasome activation mediates diabetes-induced depression-like behavior via triggering neuroinflammation. Prog Neuropsychopharmacol Biol Psychiatry 2023; 126:110796. [PMID: 37209992 DOI: 10.1016/j.pnpbp.2023.110796] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Revised: 05/02/2023] [Accepted: 05/15/2023] [Indexed: 05/22/2023]
Abstract
BACKGROUND Abundant evidence suggests that the prevalence and risk of depression in people with diabetes is high. However, the pathogenesis of diabetes-related depression remains unclear. Since neuroinflammation is associated with the pathophysiology of diabetic complications and depression, this study aims to elucidate the neuroimmune mechanism of diabetes-related depression. METHODS Male C57BL/6 mice were injected with streptozotocin to establish a diabetes model. After screening, diabetic mice were treated with the NLRP3 inhibitor MCC950. Then, metabolic indicators and depression-like behaviors were evaluated in these mice, as well as their central and peripheral inflammation. To explore the mechanism of high glucose-induced microglial NLRP3 inflammasome activation, we performed in vitro studies focusing on its canonical upstream signal I (TLR4/MyD88/NF-κB) and signal II (ROS/PKR/P2X7R/TXNIP). RESULTS Diabetic mice exhibited depression-like behaviors and activation of NLRP3 inflammasome in hippocampus. In vitro high-glucose (50 mM) environment primed microglial NLRP3 inflammasome by promoting NF-κB phosphorylation in a TLR4/MyD88-independent manner. Subsequently, high glucose activated the NLRP3 inflammasome via enhancing intracellular ROS accumulation, upregulating P2X7R, as well as promoting PKR phosphorylation and TXNIP expression, thereby facilitating the production and secretion of IL-1β. Inhibition of NLRP3 with MCC950 significantly restored hyperglycemia-induced depression-like behavior and reversed the increase in IL-1β levels in the hippocampus and serum. CONCLUSION The activation of NLRP3 inflammasome, probably mainly in hippocampal microglia, mediates the development of depression-like behaviors in STZ-induced diabetic mice. Targeting the microglial inflammasome is a feasible strategy for the treatment of diabetes-related depression.
Collapse
Affiliation(s)
- Wen-Jun Su
- Department of Stress Medicine, Faculty of Psychology, Naval Medical University, Shanghai 200433, China.
| | - Jia-Mei Li
- Department of Stress Medicine, Faculty of Psychology, Naval Medical University, Shanghai 200433, China; The 971st Hospital of PLA Navy, Qingdao 266072, China
| | - Ting Zhang
- Department of Stress Medicine, Faculty of Psychology, Naval Medical University, Shanghai 200433, China
| | - Zhi-Yong Cao
- Department of Stress Medicine, Faculty of Psychology, Naval Medical University, Shanghai 200433, China; Department of Psychiatry and Sleep Disorder, The 904th Hospital of PLA, Changzhou 213004, China
| | - Ting Hu
- Department of Stress Medicine, Faculty of Psychology, Naval Medical University, Shanghai 200433, China
| | - Shi-Yang Zhong
- Department of Stress Medicine, Faculty of Psychology, Naval Medical University, Shanghai 200433, China
| | - Zhang-Yang Xu
- Department of Stress Medicine, Faculty of Psychology, Naval Medical University, Shanghai 200433, China; The Battalion 3 of Cadet Brigade, School of Basic Medicine, Naval Medical University, Shanghai 200433, China
| | - Hong Gong
- Department of Developmental Neuropsychology, Faculty of Medical Psychology, Army Medical University (Third Military Medical University), Chongqing 400038, China
| | - Chun-Lei Jiang
- Department of Stress Medicine, Faculty of Psychology, Naval Medical University, Shanghai 200433, China.
| |
Collapse
|
35
|
Lu X, Kong X, Wu H, Hao J, Li S, Gu Z, Zeng X, Shen Y, Wang S, Chen J, Fei X, Sun Y, Li X, Jiang L, Yang F, Wang J, Cai Z. UBE2M-mediated neddylation of TRIM21 regulates obesity-induced inflammation and metabolic disorders. Cell Metab 2023; 35:1390-1405.e8. [PMID: 37343564 DOI: 10.1016/j.cmet.2023.05.011] [Citation(s) in RCA: 37] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/18/2022] [Revised: 03/22/2023] [Accepted: 05/30/2023] [Indexed: 06/23/2023]
Abstract
Inflammation is closely associated with obesity and related metabolic disorders. However, its origin during obesity is largely unknown. Here, we report that ubiquitin-conjugating enzyme E2M (UBE2M) is critical to obesity-related inflammation induced by macrophages. In mice with UBE2M-deficient macrophages, obesity, insulin resistance, and hepatic steatosis induced by a high-fat diet are greatly alleviated, an effect related to the decreased proinflammatory activity of macrophages due to reduced IL-1β production. Mechanistically, UBE2M deficiency inhibits the neddylation of E3 ubiquitin ligase TRIM21 on K129/134, leading to reduced recruitment and ubiquitination-mediated degradation of E3 ubiquitin ligase VHL. Subsequently, VHL reduces HIF-1α-induced IL-1β production by degrading HIF-1α. Targeting macrophage TRIM21 with Trim21 antisense oligonucleotide-loaded red blood cell extracellular vesicles effectively inhibits obesity-induced inflammation and related metabolic disorders. Thus, our results demonstrate that macrophage UBE2M is essential for obesity-induced inflammation and that TRIM21 is a proof-of-concept target for treating obesity and associated metabolic diseases.
Collapse
Affiliation(s)
- Xinliang Lu
- Institute of Immunology and Department of Orthopaedics of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310058, China; Institute of Immunology and Bone Marrow Transplantation Center of the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Xianghui Kong
- Institute of Immunology and Department of Orthopaedics of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Hao Wu
- Gastroenterology, Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, Wenzhou 325000, China
| | - Jiayue Hao
- Institute of Immunology and Department of Orthopaedics of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Sirui Li
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Zichun Gu
- Department of Plastic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou 310016, China
| | - Xianchang Zeng
- Institute of Immunology and Department of Orthopaedics of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Yingying Shen
- Laboratory of Cancer Biology, Key Lab of Biotherapy in Zhejiang, Cancer Center of Zhejiang University, Sir Run Run Shaw Hospital, Medical School of Zhejiang University, Hangzhou 310020, China
| | - Shibo Wang
- Institute of Immunology and Department of Orthopaedics of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Jiming Chen
- Institute of Immunology and Department of Orthopaedics of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Xuefeng Fei
- Department of Laboratory Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Yi Sun
- Cancer Institute of the Second Affiliated Hospital, Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou 310029, China
| | - Xu Li
- School of Life Science, Westlake University, Hangzhou 310024, China
| | - Lingling Jiang
- Assisted Reproduction Unit, Department of Obstetrics and Gynecology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou 310016, China
| | - Fei Yang
- Chronic Disease Research Institute, The Children's Hospital, National Clinical Research Center for Child Health, School of Public Health, School of Medicine, Zhejiang University, Hangzhou 310058, China.
| | - Jianli Wang
- Institute of Immunology and Bone Marrow Transplantation Center of the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310058, China.
| | - Zhijian Cai
- Institute of Immunology and Department of Orthopaedics of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310058, China.
| |
Collapse
|
36
|
Wang T, Xu H, Guo Y, Guo Y, Guan H, Wang D. Perfluorodecanoic acid promotes high-fat diet-triggered adiposity and hepatic lipid accumulation by modulating the NLRP3/caspase-1 pathway in male C57BL/6J mice. Food Chem Toxicol 2023; 178:113943. [PMID: 37451596 DOI: 10.1016/j.fct.2023.113943] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Revised: 06/07/2023] [Accepted: 07/11/2023] [Indexed: 07/18/2023]
Abstract
Perfluorodecanoic acid (PFDA), a chemical contaminant, may casue became obesity, which makes it a public health concern. In this study, we investigated the effects of PFDA on adiposity development and hepatic lipid accumulation in mice fed with a high-fat diet (HFD). Animals were assigned to two diet treatments (low-fat and high-fat); and PFDA was administered through drinking water for 12 weeks. The contaminant promoted body weight gain and adiposity in HFD-fed mice. Moreover, HFD-fed mice exposed to PFDA had impaired glucose metabolism, inflammation and hepatic lipid accumulation compared to mice fed HFD alone. PFDA activated the expression of hepatic NLRP3 and caspase-1, and induced that of SREBP-1c expression in the liver of HFD-fed mice. PFDA exposure in HFD-fed mice significantly inhibited hepatic AMPK expression than animals fed HFD without PFDA exposure. Furthermore, MCC950, an NLRP3 inhibitor, suppressed the upregulation of NLRP3 and caspase-1 expression, and inhibited the expression of SREBP-1c and the accumulation of hepatic lipid in mice exposed to PFDA. Thus, PFDA may enhance HFD-induced adiposity and hepatic lipid accumulation through the NLRP3/caspase-1 pathway. This contaminant may be a key risk factor for obesity development in individuals consuming high-fat foods, particularly Western diet.
Collapse
Affiliation(s)
- Taotao Wang
- Department of Clinical Nutrition, Affiliated Hospital of Jiangsu University, 212000, Zhenjiang, China
| | - Hong Xu
- School of Grain Science and Technology, Jiangsu University of Science and Technology, 212100, Zhenjiang, China
| | - Yu Guo
- School of Grain Science and Technology, Jiangsu University of Science and Technology, 212100, Zhenjiang, China
| | - Yuanxin Guo
- School of Grain Science and Technology, Jiangsu University of Science and Technology, 212100, Zhenjiang, China
| | - Huanan Guan
- School of Grain Science and Technology, Jiangsu University of Science and Technology, 212100, Zhenjiang, China.
| | - Dongxu Wang
- School of Grain Science and Technology, Jiangsu University of Science and Technology, 212100, Zhenjiang, China.
| |
Collapse
|
37
|
Gastaldi S, Rocca C, Gianquinto E, Granieri MC, Boscaro V, Blua F, Rolando B, Marini E, Gallicchio M, De Bartolo A, Romeo N, Mazza R, Fedele F, Pagliaro P, Penna C, Spyrakis F, Bertinaria M, Angelone T. Discovery of a novel 1,3,4-oxadiazol-2-one-based NLRP3 inhibitor as a pharmacological agent to mitigate cardiac and metabolic complications in an experimental model of diet-induced metaflammation. Eur J Med Chem 2023; 257:115542. [PMID: 37290185 DOI: 10.1016/j.ejmech.2023.115542] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Revised: 05/30/2023] [Accepted: 06/01/2023] [Indexed: 06/10/2023]
Abstract
Inspired by the recent advancements in understanding the binding mode of sulfonylurea-based NLRP3 inhibitors to the NLRP3 sensor protein, we developed new NLRP3 inhibitors by replacing the central sulfonylurea moiety with different heterocycles. Computational studies evidenced that some of the designed compounds were able to maintain important interaction within the NACHT domain of the target protein similarly to the most active sulfonylurea-based NLRP3 inhibitors. Among the studied compounds, the 1,3,4-oxadiazol-2-one derivative 5 (INF200) showed the most promising results being able to prevent NLRP3-dependent pyroptosis triggered by LPS/ATP and LPS/MSU by 66.3 ± 6.6% and 61.6 ± 11.5% and to reduce IL-1β release (35.5 ± 8.8% μM) at 10 μM in human macrophages. The selected compound INF200 (20 mg/kg/day) was then tested in an in vivo rat model of high-fat diet (HFD)-induced metaflammation to evaluate its beneficial cardiometabolic effects. INF200 significantly counteracted HFD-dependent "anthropometric" changes, improved glucose and lipid profiles, and attenuated systemic inflammation and biomarkers of cardiac dysfunction (particularly BNP). Hemodynamic evaluation on Langendorff model indicate that INF200 limited myocardial damage-dependent ischemia/reperfusion injury (IRI) by improving post-ischemic systolic recovery and attenuating cardiac contracture, infarct size, and LDH release, thus reversing the exacerbation of obesity-associated damage. Mechanistically, in post-ischemic hearts, IFN200 reduced IRI-dependent NLRP3 activation, inflammation, and oxidative stress. These results highlight the potential of the novel NLRP3 inhibitor, INF200, and its ability to reverse the unfavorable cardio-metabolic dysfunction associated with obesity.
Collapse
Affiliation(s)
- Simone Gastaldi
- Department of Drug Science and Technology, University of Turin, 10125, Turin, Italy
| | - Carmine Rocca
- Cellular and Molecular Cardiovascular Pathophysiology Laboratory, Department of Biology, E. and E.S. (DiBEST), University of Calabria, 87036, Rende, Italy
| | - Eleonora Gianquinto
- Department of Drug Science and Technology, University of Turin, 10125, Turin, Italy
| | - Maria Concetta Granieri
- Cellular and Molecular Cardiovascular Pathophysiology Laboratory, Department of Biology, E. and E.S. (DiBEST), University of Calabria, 87036, Rende, Italy
| | - Valentina Boscaro
- Department of Drug Science and Technology, University of Turin, 10125, Turin, Italy
| | - Federica Blua
- Department of Drug Science and Technology, University of Turin, 10125, Turin, Italy
| | - Barbara Rolando
- Department of Drug Science and Technology, University of Turin, 10125, Turin, Italy
| | - Elisabetta Marini
- Department of Drug Science and Technology, University of Turin, 10125, Turin, Italy
| | | | - Anna De Bartolo
- Cellular and Molecular Cardiovascular Pathophysiology Laboratory, Department of Biology, E. and E.S. (DiBEST), University of Calabria, 87036, Rende, Italy
| | - Naomi Romeo
- Cellular and Molecular Cardiovascular Pathophysiology Laboratory, Department of Biology, E. and E.S. (DiBEST), University of Calabria, 87036, Rende, Italy
| | - Rosa Mazza
- Cellular and Molecular Cardiovascular Pathophysiology Laboratory, Department of Biology, E. and E.S. (DiBEST), University of Calabria, 87036, Rende, Italy
| | - Francesco Fedele
- Department of Clinical, Internal, Anesthesiology and Cardiovascular Sciences, Sapienza University of Rome, Viale del Policlinico, 155, 00161, Rome, Italy; National Institute for Cardiovascular Research (INRC), Bologna, Italy
| | - Pasquale Pagliaro
- Department of Clinical and Biological Sciences, University of Turin, Turin, Italy; National Institute for Cardiovascular Research (INRC), Bologna, Italy
| | - Claudia Penna
- Department of Clinical and Biological Sciences, University of Turin, Turin, Italy; National Institute for Cardiovascular Research (INRC), Bologna, Italy.
| | - Francesca Spyrakis
- Department of Drug Science and Technology, University of Turin, 10125, Turin, Italy.
| | - Massimo Bertinaria
- Department of Drug Science and Technology, University of Turin, 10125, Turin, Italy.
| | - Tommaso Angelone
- Cellular and Molecular Cardiovascular Pathophysiology Laboratory, Department of Biology, E. and E.S. (DiBEST), University of Calabria, 87036, Rende, Italy; National Institute for Cardiovascular Research (INRC), Bologna, Italy
| |
Collapse
|
38
|
Tu XK, Chen PP, Chen JY, Ding YH, Chen Q, Shi SS. GLP-1R knockdown abrogates the protective effects of liraglutide on ischaemic stroke via inhibition of M2 polarisation and activation of NLRP3 inflammasome by reducing Nrf2 activation. Neuropharmacology 2023:109603. [PMID: 37236529 DOI: 10.1016/j.neuropharm.2023.109603] [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: 03/21/2023] [Revised: 05/18/2023] [Accepted: 05/23/2023] [Indexed: 05/28/2023]
Abstract
Liraglutide has been recently discovered to penetrate the blood-brain barrier to exert neuroprotective effects. However, relevant mechanisms of the protective effects of liraglutide on ischaemic stroke remain to be elucidated. This study examined the mechanism of GLP-1R in regulating the protective effect of liraglutide against ischaemic stroke. Middle cerebral artery occlusion (MCAO) male Sprague-Dawley rat model with/without GLP-1R or Nrf2 knockdown was established and subjected to liraglutide treatment. Then neurological deficit and brain oedema of rats was evaluated and brain tissues were subjected to TTC, Nissl, TUNEL and immunofluorescence staining. Rat primary microglial cells firstly underwent lipopolysaccharide (LPS) treatment, then GLP-1R or Nrf2 knockdown treatment, and finally Liraglutide treatment to research the NLRP3 activation. As a result, Liraglutide protected rats' brain tissues after MCAO, which attenuated brain oedema, infarct volume, neurological deficit score, neuronal apoptosis and Iba1 expression but enhanced live neurons. However, GLP-1R knockdown abrogated these protective effects of liraglutide on MCAO rats. According to in vitro experiments, Liraglutide promoted M2 polarisation, activated Nrf2 and inhibited NLRP3 activation in LPS-induced microglial cells, but GLP-1R or Nrf2 knockdown reversed these effects of Liraglutide on LPS-induced microglial cells. Further, Nrf2 knockdown counteracted the protection of liraglutide on MCAO rats, whereas sulforaphane (agonist of Nrf2) counteracted the effect of Nrf2 knockdown on liraglutide-treated MCAO rats. Collectively, GLP-1R knockdown abrogated the protection of liraglutide on MCAO rats by activating NLRP3 via inactivating Nrf2.
Collapse
Affiliation(s)
- Xian-Kun Tu
- Department of Neurosurgery, Fujian Medical University Union Hospital, Neurosurgical Institute of Fujian Province, 29# Xinquan Road, Fuzhou, Fujian, 350001, China.
| | - Ping-Ping Chen
- Department of Neurosurgery, Fujian Medical University Union Hospital, Neurosurgical Institute of Fujian Province, 29# Xinquan Road, Fuzhou, Fujian, 350001, China
| | - Jing-Yi Chen
- Department of Neurosurgery, Fujian Medical University Union Hospital, Neurosurgical Institute of Fujian Province, 29# Xinquan Road, Fuzhou, Fujian, 350001, China
| | - Yi-Hang Ding
- Department of Neurosurgery, Fujian Medical University Union Hospital, Neurosurgical Institute of Fujian Province, 29# Xinquan Road, Fuzhou, Fujian, 350001, China
| | - Quan Chen
- Department of Neurosurgery, Fujian Medical University Union Hospital, Neurosurgical Institute of Fujian Province, 29# Xinquan Road, Fuzhou, Fujian, 350001, China
| | - Song-Sheng Shi
- Department of Neurosurgery, Fujian Medical University Union Hospital, Neurosurgical Institute of Fujian Province, 29# Xinquan Road, Fuzhou, Fujian, 350001, China
| |
Collapse
|
39
|
Bauer S, Hezinger L, Rexhepi F, Ramanathan S, Kufer TA. NOD-like Receptors-Emerging Links to Obesity and Associated Morbidities. Int J Mol Sci 2023; 24:ijms24108595. [PMID: 37239938 DOI: 10.3390/ijms24108595] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Revised: 05/06/2023] [Accepted: 05/08/2023] [Indexed: 05/28/2023] Open
Abstract
Obesity and its associated metabolic morbidities have been and still are on the rise, posing a major challenge to health care systems worldwide. It has become evident over the last decades that a low-grade inflammatory response, primarily proceeding from the adipose tissue (AT), essentially contributes to adiposity-associated comorbidities, most prominently insulin resistance (IR), atherosclerosis and liver diseases. In mouse models, the release of pro-inflammatory cytokines such as TNF-alpha (TNF-α) and interleukin (IL)-1β and the imprinting of immune cells to a pro-inflammatory phenotype in AT play an important role. However, the underlying genetic and molecular determinants are not yet understood in detail. Recent evidence demonstrates that nucleotide-binding and oligomerization domain (NOD)-like receptor (NLR) family proteins, a group of cytosolic pattern recognition receptors (PRR), contribute to the development and control of obesity and obesity-associated inflammatory responses. In this article, we review the current state of research on the role of NLR proteins in obesity and discuss the possible mechanisms leading to and the outcomes of NLR activation in the obesity-associated morbidities IR, type 2 diabetes mellitus (T2DM), atherosclerosis and non-alcoholic fatty liver disease (NAFLD) and discuss emerging ideas about possibilities for NLR-based therapeutic interventions of metabolic diseases.
Collapse
Affiliation(s)
- Sarah Bauer
- Institute of Nutritional Medicine, Department of Immunology, University of Hohenheim, 70593 Stuttgart, Germany
| | - Lucy Hezinger
- Institute of Nutritional Medicine, Department of Immunology, University of Hohenheim, 70593 Stuttgart, Germany
| | - Fjolla Rexhepi
- Department of Immunology and Cell Biology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, QC J1K 2R1, Canada
| | - Sheela Ramanathan
- Department of Immunology and Cell Biology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, QC J1K 2R1, Canada
| | - Thomas A Kufer
- Institute of Nutritional Medicine, Department of Immunology, University of Hohenheim, 70593 Stuttgart, Germany
| |
Collapse
|
40
|
Liu L, Feng L, Gao J, Hu J, Li A, Zhu Y, Zhang C, Qiu B, Shen Z. Parthenolide targets NLRP3 to treat inflammasome-related diseases. Int Immunopharmacol 2023; 119:110229. [PMID: 37167640 DOI: 10.1016/j.intimp.2023.110229] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Revised: 04/03/2023] [Accepted: 04/20/2023] [Indexed: 05/13/2023]
Abstract
Natural products have attracted extensive attention from researchers in medical fields due to their abundant biological activities. Parthenolide (PTL) is a sesquiterpene lactone originally purified from herb Feverfew (Tanacetum parthenium), recent studies have showed its potential activities of anti-cancer and anti-inflammatory. Acting as the most studied inflammasome, NLRP3 inflammasome played an important role in human diseases including type-2 diabetes (T2D), Alzheimer's disease (AD) and cryopyrin-associated periodic syndromes (CAPS). In this article, we show that PTL specially inhibits the activation of NLRP3 inflammation by block the upstream signal and prevent the assembly of NLRP3 inflammasome complex. Furthermore, we showed the treatment of PTL significantly attenuates the symptoms of lipopolysaccharide (LPS)-induced systemic inflammation and dextran sulfate sodium (DSS)-induced colitis in mice models. Thus, our results demonstrate that PTL alleviates inflammation by targeting NLRP3 inflammasome, which indicate that PTL acting as a promising natural product for the treatment of NLRP3 inflammasome-related diseases.
Collapse
Affiliation(s)
- Liu Liu
- Department of Laboratory Medicine, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230001, China
| | - Linxiang Feng
- Department of Laboratory Medicine, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230001, China
| | - Jiahui Gao
- Department of Laboratory Medicine, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230001, China
| | - Jie Hu
- Department of Laboratory Medicine, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230001, China
| | - Ang Li
- Department of Laboratory Medicine, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230001, China
| | - Yangyang Zhu
- School of Medicine & Institute for Life Sciences, South China University of Technology, Guangzhou 510006, China
| | - Changlong Zhang
- Department of Laboratory Medicine, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230001, China
| | - Bensheng Qiu
- Center for Biomedical Imaging, University of Science and Technology of China, Hefei 230026, China
| | - Zuojun Shen
- Department of Laboratory Medicine, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230001, China.
| |
Collapse
|
41
|
Wan J, Zhu Z, He Z, Wu H, Chen A, Zhu W, Cheng P. Stevioside protects primary articular chondrocytes against IL-1β-induced inflammation and catabolism by targeting integrin. Int Immunopharmacol 2023; 119:110261. [PMID: 37167638 DOI: 10.1016/j.intimp.2023.110261] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Revised: 04/26/2023] [Accepted: 04/28/2023] [Indexed: 05/13/2023]
Abstract
Osteoarthritis (OA) is a common, progressive, and chronic disorder of the joints that is characterized by the inflammation and degradation of articular cartilage and is known to significantly impair quality of daily life. Stevioside (SVS) is a natural diterpenoid glycoside that has anti-inflammatory benefits. Hence, in the current research, it was hypothesized that SVS might exert anti-inflammatory effects on articular chondrocytes and alleviate cartilage degradation in mice with OA. The expression of inflammatory cytokines, like inducible nitric oxide synthase (iNOS), NOD-, LRR- and pyrin domain-containing protein 3 (NLRP3), and cyclooxygenase-2 (COX-2) in chondrocytes after interleukin-1β (IL-1β) exposure, was inhibited by the pretreatment of SVS. As well, SVS inhibited the reduction of collagen II and sry-box transcription factor 9 (SOX9) in chondrocytes stimulated by IL-1β and suppressed the expression of MMP3 and MMP13. Further, after treatment with SVS, cell cytometry, autophagy flux, and related protein expression showed diminished cell apoptosis and reduced autophagy impairment. Moreover, SVS blocked the activation of phosphoinositide-3-kinase/Akt/nuclear factor-kappa beta (PI3K/Akt/NF-κB) and mitogen-activated protein kinase (MAPK) signaling pathways stimulated by IL-1β. This resulted in decreased cellular inflammation. In vivo experiments with intra-articular injections of SVS in mice with the DMM mouse model demonstrated a decrease in cartilage degradation and an improvement in subchondral bone remodeling. After the integrin αVβ3-related knockdown using siRNA, a reversed effect was observed on the anti-inflammatory, anabolic promoting, catabolic blocking, and NF-κB and MAPK signaling pathway inhibition of SVS on chondrocytes treated with IL-1β. The above findings highlighted that SVS blocked IL-1β, triggered an inflammatory response in mice chondrocytes, and prevented cartilage degradation in vivo through integrin αVβ3. This suggested that SVS might serve as a novel therapeutic option for OA.
Collapse
Affiliation(s)
- Junlai Wan
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei, China
| | - Ziqing Zhu
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei, China
| | - Zhiyi He
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei, China
| | - Hua Wu
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei, China
| | - Anmin Chen
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei, China
| | - Wentao Zhu
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei, China.
| | - Peng Cheng
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei, China.
| |
Collapse
|
42
|
Li L, Tian Y. The role of metabolic reprogramming of tumor-associated macrophages in shaping the immunosuppressive tumor microenvironment. Biomed Pharmacother 2023; 161:114504. [PMID: 37002579 DOI: 10.1016/j.biopha.2023.114504] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Revised: 03/04/2023] [Accepted: 03/07/2023] [Indexed: 03/15/2023] Open
Abstract
Macrophages are potent immune effector cells in innate immunity and exert dual-effects in the tumor microenvironment (TME). Tumor-associated macrophages (TAMs) make up a significant portion of TME immune cells. Similar to M1/M2 macrophages, TAMs are also highly plastic, and their functions are regulated by cytokines, chemokines and other factors in the TME. The metabolic changes in TAMs are significantly associated with polarization towards a protumour or antitumour phenotype. The metabolites generated via TAM metabolic reprogramming in turn promote tumor progression and immune tolerance. In this review, we explore the metabolic reprogramming of TAMs in terms of energy, amino acid and fatty acid metabolism and the potential roles of these changes in immune suppression.
Collapse
Affiliation(s)
- Lunxu Li
- Department of General Surgery, Shengjing Hospital of China Medical University, Shenyang, China
| | - Yu Tian
- Department of General Surgery, Shengjing Hospital of China Medical University, Shenyang, China.
| |
Collapse
|
43
|
Zhu H, Zhang L, Xiao F, Wu L, Guo Y, Zhang Z, Xiao Y, Sun G, Yang Q, Guo H. Melatonin-Driven NLRP3 Inflammation Inhibition Via Regulation of NF-κB Nucleocytoplasmic Transport: Implications for Postoperative Cognitive Dysfunction. Inflammation 2023:10.1007/s10753-023-01822-5. [PMID: 37185803 DOI: 10.1007/s10753-023-01822-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Revised: 04/02/2023] [Accepted: 04/13/2023] [Indexed: 05/17/2023]
Abstract
The aseptic inflammatory response of the central nervous system is one of the important causes of neurodegenerative diseases in individuals and is also recognized in postoperative cognitive dysfunction (POCD). Inflammasome is thought to be closely related to brain homeostasis. However, there are few drugs targeting the inflammasome to suppress inflammation in clinical practice. Here, we showed that the neuroinflammatory response mediated by the NLRP3 (NLR family, pyrin domain containing 3) inflammasome was involved in the pathological process of POCD. Melatonin protected mice from nerve damage by inhibiting activation of the NLRP3-caspase-1-interleukin 1 beta (IL-β) pathway and thus reduced the secretion of IL-1β inflammatory factors in microglia. Further research found that melatonin has a potential binding effect with NLRP3 protein, and at the same time could reduce the phosphorylation of nuclear factor kappa-B (NF-κB) and inhibit its nuclear translocation. The underlying mechanism was that melatonin inhibited the expression of acetylation of histone H3 and melatonin attenuated the binding of NF-κb to the NLRP3 promoter region 1-200 bp, where there are two potential binding target sites of NF-κb and NLRP3, namely the sequences 5'-GGGAACCCCC-3' and 5'-GGAAATCCA -3'. Therefore, we confirmed a novel mechanism of action of melatonin in the prevention and treatment of POCD.
Collapse
Affiliation(s)
- Hong Zhu
- Department of Neurosurgery, The Second Affiliated Hospital of Nanchang University, 1# Minde Road, Nanchang, China
- Jiangxi Key Laboratory of Neurological Tumors and Cerebrovascular Diseases, Nanchang, China
- Jiangxi Health Commission Key Laboratory of Neurological Medicine, Nanchang, China
- Institute of Neuroscience, Nanchang University, Nanchang, China
| | - Lieliang Zhang
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Nanchang, China
- Key Laboratory of Anesthesiology of Jiangxi Province, Nanchang, China
| | - Feng Xiao
- Department of Neurosurgery, The Second Affiliated Hospital of Nanchang University, 1# Minde Road, Nanchang, China
- Jiangxi Key Laboratory of Neurological Tumors and Cerebrovascular Diseases, Nanchang, China
- Jiangxi Health Commission Key Laboratory of Neurological Medicine, Nanchang, China
- Institute of Neuroscience, Nanchang University, Nanchang, China
| | - Lei Wu
- Department of Neurosurgery, The Second Affiliated Hospital of Nanchang University, 1# Minde Road, Nanchang, China
- Jiangxi Key Laboratory of Neurological Tumors and Cerebrovascular Diseases, Nanchang, China
- Jiangxi Health Commission Key Laboratory of Neurological Medicine, Nanchang, China
- Institute of Neuroscience, Nanchang University, Nanchang, China
| | - Yun Guo
- Department of Neurosurgery, The Second Affiliated Hospital of Nanchang University, 1# Minde Road, Nanchang, China
- Jiangxi Key Laboratory of Neurological Tumors and Cerebrovascular Diseases, Nanchang, China
- Jiangxi Health Commission Key Laboratory of Neurological Medicine, Nanchang, China
- Institute of Neuroscience, Nanchang University, Nanchang, China
| | - Zhe Zhang
- Department of Neurosurgery, The Second Affiliated Hospital of Nanchang University, 1# Minde Road, Nanchang, China
- Jiangxi Key Laboratory of Neurological Tumors and Cerebrovascular Diseases, Nanchang, China
- Jiangxi Health Commission Key Laboratory of Neurological Medicine, Nanchang, China
- Institute of Neuroscience, Nanchang University, Nanchang, China
| | - Yao Xiao
- Department of Neurosurgery, The Second Affiliated Hospital of Nanchang University, 1# Minde Road, Nanchang, China
- Jiangxi Key Laboratory of Neurological Tumors and Cerebrovascular Diseases, Nanchang, China
- Jiangxi Health Commission Key Laboratory of Neurological Medicine, Nanchang, China
- Institute of Neuroscience, Nanchang University, Nanchang, China
| | - Gufeng Sun
- Department of Neurosurgery, The Second Affiliated Hospital of Nanchang University, 1# Minde Road, Nanchang, China
- Jiangxi Key Laboratory of Neurological Tumors and Cerebrovascular Diseases, Nanchang, China
- Jiangxi Health Commission Key Laboratory of Neurological Medicine, Nanchang, China
- Institute of Neuroscience, Nanchang University, Nanchang, China
| | - Qing Yang
- Department of Respiratory Medicine, The Second Affiliated Hospital of Nanchang University, 1# Minde Road, Nanchang, China.
| | - Hua Guo
- Department of Neurosurgery, The Second Affiliated Hospital of Nanchang University, 1# Minde Road, Nanchang, China.
- Jiangxi Key Laboratory of Neurological Tumors and Cerebrovascular Diseases, Nanchang, China.
- Jiangxi Health Commission Key Laboratory of Neurological Medicine, Nanchang, China.
- Institute of Neuroscience, Nanchang University, Nanchang, China.
| |
Collapse
|
44
|
Xu H, Xu Z, Long S, Li Z, Jiang J, Zhou Q, Huang X, Wu X, Wei W, Li X. The role of the gut microbiome and its metabolites in cerebrovascular diseases. Front Microbiol 2023; 14:1097148. [PMID: 37125201 PMCID: PMC10140324 DOI: 10.3389/fmicb.2023.1097148] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2022] [Accepted: 03/23/2023] [Indexed: 05/02/2023] Open
Abstract
The gut microbiome is critically involved in maintaining normal physiological function in the host. Recent studies have revealed that alterations in the gut microbiome contribute to the development and progression of cerebrovascular disease via the microbiota-gut-brain axis (MGBA). As a broad communication network in the human body, MGBA has been demonstrated to have significant interactions with various factors, such as brain structure and function, nervous system diseases, etc. It is also believed that the species and composition of gut microbiota and its metabolites are intrinsically linked to vascular inflammation and immune responses. In fact, in fecal microbiota transplantation (FMT) research, specific gut microbiota and downstream-related metabolites have been proven to not only participate in various physiological processes of human body, but also affect the occurrence and development of cerebrovascular diseases directly or indirectly through systemic inflammatory immune response. Due to the high mortality and disability rate of cerebrovascular diseases, new treatments to improve intestinal dysbacteriosis have gradually attracted widespread attention to better ameliorate the poor prognosis of cerebrovascular diseases in a non-invasive way. This review summarizes the latest advances in the gut microbiome and cerebrovascular disease research and reveals the profound impact of gut microbiota dysbiosis and its metabolites on cerebrovascular diseases. At the same time, we elucidated molecular mechanisms whereby gut microbial metabolites regulate the expression of specific interleukins in inflammatory immune responses. Moreover, we further discuss the feasibility of novel therapeutic strategies targeting the gut microbiota to improve the outcome of patients with cerebrovascular diseases. Finally, we provide new insights for standardized diagnosis and treatment of cerebrovascular diseases.
Collapse
Affiliation(s)
- Hongyu Xu
- Department of Neurosurgery, Zhongnan Hospital, Wuhan University, Wuhan, Hubei, China
- Brain Research Center, Zhongnan Hospital, Wuhan University, Wuhan, Hubei, China
| | - Ziyue Xu
- Department of Neurosurgery, Zhongnan Hospital, Wuhan University, Wuhan, Hubei, China
- Brain Research Center, Zhongnan Hospital, Wuhan University, Wuhan, Hubei, China
| | - Shengrong Long
- Department of Neurosurgery, Zhongnan Hospital, Wuhan University, Wuhan, Hubei, China
- Brain Research Center, Zhongnan Hospital, Wuhan University, Wuhan, Hubei, China
| | - Zhengwei Li
- Department of Neurosurgery, Zhongnan Hospital, Wuhan University, Wuhan, Hubei, China
| | - Jiazhi Jiang
- Department of Neurosurgery, Zhongnan Hospital, Wuhan University, Wuhan, Hubei, China
- Brain Research Center, Zhongnan Hospital, Wuhan University, Wuhan, Hubei, China
| | - Qiangqiang Zhou
- Department of Neurosurgery, Zhongnan Hospital, Wuhan University, Wuhan, Hubei, China
- Brain Research Center, Zhongnan Hospital, Wuhan University, Wuhan, Hubei, China
| | - Xiaopeng Huang
- Department of Neurosurgery, Zhongnan Hospital, Wuhan University, Wuhan, Hubei, China
- Brain Research Center, Zhongnan Hospital, Wuhan University, Wuhan, Hubei, China
| | - Xiaohui Wu
- Department of Neurosurgery, Zhongnan Hospital, Wuhan University, Wuhan, Hubei, China
| | - Wei Wei
- Department of Neurosurgery, Zhongnan Hospital, Wuhan University, Wuhan, Hubei, China
- Brain Research Center, Zhongnan Hospital, Wuhan University, Wuhan, Hubei, China
| | - Xiang Li
- Department of Neurosurgery, Zhongnan Hospital, Wuhan University, Wuhan, Hubei, China
- Brain Research Center, Zhongnan Hospital, Wuhan University, Wuhan, Hubei, China
| |
Collapse
|
45
|
Wang T, Xu H, Dong R, Wu S, Guo Y, Wang D. Effectiveness of targeting the NLRP3 inflammasome by using natural polyphenols: A systematic review of implications on health effects. Food Res Int 2023; 165:112567. [PMID: 36869555 DOI: 10.1016/j.foodres.2023.112567] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2022] [Revised: 10/13/2022] [Accepted: 01/30/2023] [Indexed: 02/05/2023]
Abstract
Globally, inflammation and metabolic disorders pose serious public health problems and are major health concerns. It has been shown that natural polyphenols are effective in the treatment of metabolic diseases, including anti-inflammation, anti-diabetes, anti-obesity, neuron-protection, and cardio-protection. NLRP3 inflammasome, which are multiprotein complexes located within the cytosol, play an important role in the innate immune system. However, aberrant activation of the NLRP3 inflammasome were discovered as essential molecular mechanisms in triggering inflammatory processes as well as implicating it in several major metabolic diseases, such as type 2 diabetes mellitus, obesity, atherosclerosis or cardiovascular disease. Recent studies indicate that natural polyphenols can inhibit NLRP3 inflammasome activation. In this review, the progress of natural polyphenols preventing inflammation and metabolic disorders via targeting NLRP3 inflammasome is systemically summarized. From the viewpoint of interfering NLRP3 inflammasome activation, the health effects of natural polyphenols are explained. Recent advances in other beneficial effects, clinical trials, and nano-delivery systems for targeting NLRP3 inflammasome are also reviewed. NLRP3 inflammasome is targeted by natural polyphenols to exert multiple health effects, which broadens the understanding of polyphenol mechanisms and provides valuable guidance to new researchers in this field.
Collapse
Affiliation(s)
- Taotao Wang
- Department of Clinical Nutrition, Affiliated Hospital of Jiangsu University, 212000 Zhenjiang, China
| | - Hong Xu
- School of Grain Science and Technology, Jiangsu University of Science and Technology, 212100 Zhenjiang, China
| | - Ruixia Dong
- College of Horticulture, Jinling Institute of Technology, 211169 Nanjing, China
| | - Shanshan Wu
- College of Agriculture & Biotechnology, Zhejiang University, 310058 Hanzhou, China
| | - Yuanxin Guo
- School of Grain Science and Technology, Jiangsu University of Science and Technology, 212100 Zhenjiang, China.
| | - Dongxu Wang
- School of Grain Science and Technology, Jiangsu University of Science and Technology, 212100 Zhenjiang, China.
| |
Collapse
|
46
|
van Gemert Y, Kruisbergen NNL, Blom AB, van den Bosch MHJ, van der Kraan PM, Pieterman EJ, Princen HMG, van Lent PLEM. IL-1β inhibition combined with cholesterol-lowering therapies decreases synovial lining thickness and spontaneous cartilage degeneration in a humanized dyslipidemia mouse model. Osteoarthritis Cartilage 2023; 31:340-350. [PMID: 36442605 DOI: 10.1016/j.joca.2022.09.014] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Revised: 09/02/2022] [Accepted: 09/26/2022] [Indexed: 11/26/2022]
Abstract
INTRODUCTION Both systemic inflammation and dyslipidemia contribute to osteoarthritis (OA) development and have been suggested as a possible link between metabolic disease and OA development. Recently, the CANTOS trial showed a reduction in knee and hip replacements after inhibition of IL-1β in patients with a history of cardiovascular disease and high inflammatory risk. In this light, we investigated whether inhibition of IL-1β combined with cholesterol-lowering therapies can reduce OA development in dyslipidemic APOE∗3Leiden mice under pro-inflammatory dietary conditions. MATERIALS AND METHODS Female ApoE3∗Leiden mice were fed a cholesterol-supplemented Western-Type diet (WTD) for 38 weeks. After 14 weeks, cholesterol-lowering and anti-inflammatory treatments were started. Treatments included atorvastatin alone or with an anti-IL1β antibody, and atorvastatin combined with proprotein convertase subtilisin-kexin type 9 (PCSK9) inhibitor alirocumab without or with the anti-IL1β antibody. Knee joints were analyzed for cartilage degradation, synovial inflammation and ectopic bone formation using histology at end point. RESULTS Cholesterol-lowering treatment successfully decreased systemic inflammation in dyslipidemic mice, which was not further affected by inhibition of IL-1β. Synovial thickening and cartilage degeneration were significantly decreased in mice that received cholesterol-lowering treatment combined with inhibition of IL-1β (P < 0.01, P < 0.05, respectively) compared to mice fed a WTD alone. Ectopic bone formation was comparable between all groups. CONCLUSION These results indicate that inhibition of IL-1β combined with cholesterol-lowering therapy diminishes synovial thickening and cartilage degeneration in mice and may imply that this combination therapy could be beneficial in patients with metabolic inflammation.
Collapse
Affiliation(s)
- Y van Gemert
- Experimental Rheumatology, Radboud University Medical Center, Nijmegen, the Netherlands
| | - N N L Kruisbergen
- Experimental Rheumatology, Radboud University Medical Center, Nijmegen, the Netherlands
| | - A B Blom
- Experimental Rheumatology, Radboud University Medical Center, Nijmegen, the Netherlands
| | - M H J van den Bosch
- Experimental Rheumatology, Radboud University Medical Center, Nijmegen, the Netherlands
| | - P M van der Kraan
- Experimental Rheumatology, Radboud University Medical Center, Nijmegen, the Netherlands
| | - E J Pieterman
- Metabolic Health Research, TNO, Leiden, the Netherlands
| | - H M G Princen
- Metabolic Health Research, TNO, Leiden, the Netherlands
| | - P L E M van Lent
- Experimental Rheumatology, Radboud University Medical Center, Nijmegen, the Netherlands.
| |
Collapse
|
47
|
Li L, Li J, Li S, Chen H, Wu Y, Qiu Y. IL-37 alleviates alveolar bone resorption and inflammatory response through the NF-κB/NLRP3 signaling pathway in male mice with periodontitis. Arch Oral Biol 2023; 147:105629. [PMID: 36680836 DOI: 10.1016/j.archoralbio.2023.105629] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Revised: 01/15/2023] [Accepted: 01/17/2023] [Indexed: 01/20/2023]
Abstract
OBJECTIVE Periodontitis is an inflammatory disease, characterized by periodontal pocket formation and alveolar bone resorption, is one of the most common oral diseases. Interleukin-37 (IL-37) is a novel inflammatory suppressor that plays an important role in many inflammatory diseases. This study aimed to investigate the role of IL-37 in periodontitis DESIGN: A mouse model of periodontitis was established by Porphyromonas gingivalis. After four weeks treatment of recombinant human IL-37 (rhIL-37), the effects of IL-37 on the gingival index and tooth loosening degree of periodontitis mice were observed. H&E staining and micro-CT scanning were used to analyze the bone resorption of the maxillary. The number of osteoclasts was counted by TRAP staining and the differentiation of osteoclasts was evaluated by immunohistochemistry. The expression of inflammatory cytokines was detected by ELISA, and the protein expressions of the NF-κB/NLRP3 pathway were analyzed by WB. RESULTS RhIL-37 significantly decreased the gingival index and tooth mobility degree, inhibited maxillary bone resorption, decreased the number of osteoclasts and the expression of calcitonin receptor (CTR), periodontal cathepsin K (CTSK) and receptor activator of NF-κB ligand (RANKL), and increased the expression of osteoprotegerin (OPG) in periodontitis mice. At the same time, rhIL-37 significantly decreased the expression of IL-1β, IL-6 and TNF-α, and increased the expression of IL-10 in the gingival tissue of periodontitis mice. In addition, rhIL-37 significantly inhibited the protein expressions of p-p65, NLRP3, ASC, caspase-1 and IL-1β in periodontitis mice. CONCLUSION IL-37 may alleviate alveolar bone resorption and inflammation response in periodontitis through the NF-κB/NLRP3 pathway.
Collapse
Affiliation(s)
- Lihua Li
- Department of Stomatology, Affiliated Hospital of North Sichuan Medical College, China
| | - Junxiong Li
- Department of Stomatology, Affiliated Hospital of North Sichuan Medical College, China
| | - Siyu Li
- Department of Stomatology, Affiliated Hospital of North Sichuan Medical College, China
| | - Hongjun Chen
- Department of Stomatology, Affiliated Hospital of North Sichuan Medical College, China
| | - Yan Wu
- Department of Stomatology, Affiliated Hospital of North Sichuan Medical College, China
| | - Ya Qiu
- Medical Research Center, Affiliated Hospital of North Sichuan Medical College, China.
| |
Collapse
|
48
|
Zhang N, Cao W, He X, Xing Y, Yang N. Long Non-Coding RNAs in Retinal Ganglion Cell Apoptosis. Cell Mol Neurobiol 2023; 43:561-574. [PMID: 35226226 PMCID: PMC11415166 DOI: 10.1007/s10571-022-01210-x] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2021] [Accepted: 02/17/2022] [Indexed: 12/19/2022]
Abstract
Traumatic optic neuropathy or other neurodegenerative diseases, including optic nerve transection, glaucoma, and diabetic retinopathy, can lead to progressive and irreversible visual damage. Long non-coding RNAs (lncRNAs), which belong to the family of non-protein-coding transcripts, have been linked to the pathogenesis, progression, and prognosis of these lesions. Retinal ganglion cells (RGCs) are critical for the transmission of visual information to the brain, damage to which results in visual loss. Apoptosis has been identified as one of the most essential modes of RGC death. Emerging evidence suggests that lncRNAs can regulate RGC degeneration by directly or indirectly modulating apoptosis-associated signaling pathways. This review presents a comprehensive overview of the role of lncRNAs in RGC apoptosis at transcriptional, post-transcriptional, translational, and post-translational levels, emphasizing on the potential mechanisms of action. The current limitations and future perspectives of exploring the connection between lncRNAs and RGC apoptosis have been summarized. Understanding the intricate molecular interaction network of lncRNAs and RGC apoptosis will open new avenues for the identification of novel diagnostic biomarkers, therapeutic targets, and molecules for prognostic evaluation of diseases related to RGC injury.
Collapse
Affiliation(s)
- Ningzhi Zhang
- Department of Ophthalmology, Renmin Hospital of Wuhan University, #238 Jiefang Road, Wuhan, 430060, China
| | - Wenye Cao
- Department of Ophthalmology, Renmin Hospital of Wuhan University, #238 Jiefang Road, Wuhan, 430060, China
| | - Xuejun He
- Department of Ophthalmology, Renmin Hospital of Wuhan University, #238 Jiefang Road, Wuhan, 430060, China
| | - Yiqiao Xing
- Department of Ophthalmology, Renmin Hospital of Wuhan University, #238 Jiefang Road, Wuhan, 430060, China
| | - Ning Yang
- Department of Ophthalmology, Renmin Hospital of Wuhan University, #238 Jiefang Road, Wuhan, 430060, China.
| |
Collapse
|
49
|
Yousaf H, Khan MIU, Ali I, Munir MU, Lee KY. Emerging role of macrophages in non-infectious diseases: An update. Biomed Pharmacother 2023; 161:114426. [PMID: 36822022 DOI: 10.1016/j.biopha.2023.114426] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Revised: 02/16/2023] [Accepted: 02/17/2023] [Indexed: 02/23/2023] Open
Abstract
In the past three decades, a huge body of evidence through various research studies conducted on animal models, has demonstrated that the macrophages are centralized of all the leukocytes involved in diseases and, particularly, their role in non-infectious diseases has been studied extensively for which they have also been referred to as the "double-edged swords". The most versatile of all immunocytes, macrophages play a key role in health and diseases. Various experimental models have demonstrated the conventional paradigms such as the M1/M2 dichotomy, which is not as obvious and presents a complex characterization of the macrophages in the disease immunology. In human diseases, this M1-M2 continuum shows a complex web of mechanisms, which are majorly divided into the pro-inflammatory roles (derived mainly by the cytokines: IL-1, IL-6, IL-12, IL-23, and tumor necrosis factor) and anti-inflammatory roles (CCl-17, CCl-22, CCL-2, transforming growth factor (TGF), and interleukin-10), which are involved in the wound healing and pathogen-suppression. The conventional division of these macrophages as M1 and M2 is derived from the opposing functions of these macrophages; where M1 is involved in the tissue damage and pro-inflammatory roles and M2 promotes cell proliferation and the resolution of inflammation. Both these pathways down-regulate each other in diseases through a plethora of enzymatic and cytokine mediators.
Collapse
Affiliation(s)
- Hassan Yousaf
- Institute of Molecular Biology and Biotechnology (IMBB), University of Lahore, Lahore, Pakistan
| | - Malik Ihsan Ullah Khan
- Institute of Molecular Biology and Biotechnology (IMBB), University of Lahore, Lahore, Pakistan.
| | - Iftikhar Ali
- Department of Life Sciences, University of Management and Technology, Lahore, Pakistan
| | - Muhammad Usman Munir
- Department of Pharmaceutical Chemistry, College of Pharmacy, Jouf University Sakaka, Aljouf 72388, Saudi Arabia
| | - Ka Yiu Lee
- Swedish Winter Sports Research Centre, Department of Health Sciences, Mid Sweden University, Ostersund, Sweden.
| |
Collapse
|
50
|
Jing W, Liu C, Su C, Liu L, Chen P, Li X, Zhang X, Yuan B, Wang H, Du X. Role of reactive oxygen species and mitochondrial damage in rheumatoid arthritis and targeted drugs. Front Immunol 2023; 14:1107670. [PMID: 36845127 PMCID: PMC9948260 DOI: 10.3389/fimmu.2023.1107670] [Citation(s) in RCA: 51] [Impact Index Per Article: 25.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2022] [Accepted: 01/30/2023] [Indexed: 02/11/2023] Open
Abstract
Rheumatoid arthritis (RA) is an autoimmune disease characterized by synovial inflammation, pannus formation, and bone and cartilage damage. It has a high disability rate. The hypoxic microenvironment of RA joints can cause reactive oxygen species (ROS) accumulation and mitochondrial damage, which not only affect the metabolic processes of immune cells and pathological changes in fibroblastic synovial cells but also upregulate the expression of several inflammatory pathways, ultimately promoting inflammation. Additionally, ROS and mitochondrial damage are involved in angiogenesis and bone destruction, thereby accelerating RA progression. In this review, we highlighted the effects of ROS accumulation and mitochondrial damage on inflammatory response, angiogenesis, bone and cartilage damage in RA. Additionally, we summarized therapies that target ROS or mitochondria to relieve RA symptoms and discuss the gaps in research and existing controversies, hoping to provide new ideas for research in this area and insights for targeted drug development in RA.
Collapse
Affiliation(s)
- Weiyao Jing
- Department of Acupuncture-Moxibustion and Tuina, Gansu University of Chinese Medicine, Lanzhou, China
| | - Cui Liu
- Department of Acupuncture-Moxibustion and Tuina, Gansu University of Chinese Medicine, Lanzhou, China
| | - Chenghong Su
- Department of Acupuncture-Moxibustion and Tuina, Gansu University of Chinese Medicine, Lanzhou, China
| | - Limei Liu
- Department of Acupuncture-Moxibustion and Tuina, Gansu University of Chinese Medicine, Lanzhou, China
| | - Ping Chen
- Department of Rheumatic and Bone Disease, Gansu Provincial Hospital of Traditional Chinese Medicine, Lanzhou, China
| | - Xiangjun Li
- Department of Rheumatic and Bone Disease, Gansu Provincial Hospital of Traditional Chinese Medicine, Lanzhou, China
| | - Xinghua Zhang
- Department of Acupuncture, Gansu Provincial Hospital of Traditional Chinese Medicine, Lanzhou, China
| | - Bo Yuan
- Department of Acupuncture and Pain, Affiliated Hospital of Gansu University of Traditional Chinese Medicine, Lanzhou, China
| | - Haidong Wang
- Department of Rheumatic and Bone Disease, Gansu Provincial Hospital of Traditional Chinese Medicine, Lanzhou, China
| | - Xiaozheng Du
- Department of Acupuncture-Moxibustion and Tuina, Gansu University of Chinese Medicine, Lanzhou, China
| |
Collapse
|