51
|
Nagy AM, Abdelhameed MF, Rihan S, Diab KA, El-Saied M, Mohamed SS, El-Nattat WS, Hammam AMM. Rosemary officinalis extract mitigates potassium dichromate-induced testicular degeneration in male rats: Insights from the Nrf2 and its target genes signaling pathway. Toxicol Rep 2024; 13:101700. [PMID: 39165924 PMCID: PMC11334654 DOI: 10.1016/j.toxrep.2024.101700] [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: 06/08/2024] [Revised: 07/14/2024] [Accepted: 07/23/2024] [Indexed: 08/22/2024] Open
Abstract
This study aimed to investigate the protective effects of Rosemary ethanol extract (ROEE) on testicular damage induced by potassium Dichromate (PDC) in male rats regarding the signaling pathway of Nrf2 and its target genes and proteins. A total of 28 male rats were divided into four groups: control, PDC only (15 mg/kg b.w. orally), PDC + low dose ROEE (220 mg/kg b.w.), and PDC + high dose ROEE (440 mg/kg b.w.). After 28 days of consecutive treatment, the rats were sacrificed for histological, immunohistochemistry, and biochemical analyses. The results revealed that the ROEE treatment up-regulated the Nrf2 and its target genes (NQO1, HO-1) mRNA expressions compared to the PDC group. correspondingly, the protein levels of GCLM, GSH, SOD, and catalase were significantly increased in the ROEE-treated animals compared to the PDC-treated animals. Furthermore, ROEE administration led to increased serum levels of testosterone (T4) and decreased levels of estrogen (E2) compared to the PDC group. Semen analysis and histopathology demonstrated that ROEE administration significantly improved spermatological impairment caused by PDC. The immunoexpression of cytoplasmic HSP-90 was reduced in the ROEE-treated groups, while the expression of androgen receptor (AR) was markedly improved. ROEE exhibited protective effects against PDC-induced testicular damage, likely due to its antioxidant properties. However, further investigation is required to elucidate the underlying mechanisms of action.
Collapse
Affiliation(s)
- Ahmed M. Nagy
- Department of Animal Reproduction &AI, Veterinary Research Institute, National Research Centre, Cairo, Egypt
| | - Mohamed F. Abdelhameed
- Pharmacology Department, Medical research and clinical studies institute, National Research Centre, Cairo, Egypt
| | - Shaimaa Rihan
- Department of Biochemistry, Faculty of Science, Ain Shams University, Cairo, Egypt
| | - Kawthar A. Diab
- Department of Genetics and Cytology, National Research Centre, Cairo, Egypt
| | - Mohamed El-Saied
- Department of Pathology, Faculty of Veterinary Medicine, Cairo University, Giza, Egypt
| | - Shereif S. Mohamed
- Nutrition and Food Science Department, National Research Centre, Cairo, Egypt
| | - Walid S. El-Nattat
- Department of Animal Reproduction &AI, Veterinary Research Institute, National Research Centre, Cairo, Egypt
| | - Abdel Mohsen M. Hammam
- Department of Animal Reproduction &AI, Veterinary Research Institute, National Research Centre, Cairo, Egypt
| |
Collapse
|
52
|
Kong R, Peng L, Bao H, Sun L, Feng Y, Li H, Wang D. The role of Gαq in regulating NLRP3 inflammasome activation. Inflamm Res 2024; 73:2249-2261. [PMID: 39455437 DOI: 10.1007/s00011-024-01961-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Revised: 09/20/2024] [Accepted: 10/11/2024] [Indexed: 10/28/2024] Open
Abstract
BACKGROUND G proteins are a class of important signal transducers in mammalians. G proteins can corpoarated with G proteincoupled receptors (GPCRs) and transmit signals from extracellular stimuli into intracellular response, which will regulate a series of biological functions. G-proteins are heterotrimeric proteins composed of Gα, Gβ, and Gγ subunits. Based on structural and functional similarity of their α-subunits, G proteins are typically grouped into four classes (Gi, Gs, Gq/11, and G12/13). The Gq/11 subfamily consists of Gq, G11, G14, and G15/16 proteins. Gαq is the α-subunit of Gq protein and encoded by GNAQ. Our previous studies revealed that Gαq play an important role in regulating T cell survival and T cell differentiation. Inflammasomes are multiprotein complexes that play a critical role in modulating innate inflammatory response. NLRP3 inflammasome is currently the most extensively studied inflammasome. METHODS We found that Gαq suppressed NLRP3 inflammasome activation in macrophage, Gαq also suppressed NLRP3 inflammasome activation in a LPS-induced sepsis mouse model. Gαq can locate to mitochondria and Gαq was required for the maintenance of mitochondrial homeostasis. Gαq regulated NLRP3 inflammasome activation by modulating mitochondrial reactive oxygen species (mtROS). RESULTS We found that Gαq suppressed NLRP3 inflammasome activation in macrophage, Gαq also suppressed NLRP3 inflammasome activation in a LPS-induced sepsis mouse model. Gαq can locate to mitochondria and Gαq was required for the maintenance of mitochondrial homeostasis. Gαq regulated NLRP3 inflammasome activation by modulating mitochondrial reactive oxygen species (mtROS). CONCLUSION Our results indicate that Gαq regulates NLRP3 inflammasome activation by modulating mitochondrial ROS production. Our research provides new mechanistic insight into the activation of NLRP3 inflammasome. As it has been proved that NLRP3 inflammasome plays an important role in the pathogenesis many diseases such as Alzheimer's disease, cancer, and inflammatory bowel disease, Gαq might become a novel drug target for these diseases in future.
Collapse
Affiliation(s)
- Ruixue Kong
- College of Life Sciences, Shandong Normal University, Jinan, 250014, Shandong, China
| | - Lijun Peng
- Department of Gastroenterology, Linyi People's Hospital, Linyi, 276000, Shandong, China
| | - Honggang Bao
- Department of Laboratory Medicine, Linyi Cancer Hospital, Linyi, 276000, Shandong, China
| | - Lulu Sun
- College of Life Sciences, Shandong Normal University, Jinan, 250014, Shandong, China
| | - Yan Feng
- College of Life Sciences, Shandong Normal University, Jinan, 250014, Shandong, China
| | - Hua Li
- College of Life Sciences, Shandong Normal University, Jinan, 250014, Shandong, China.
| | - Dashan Wang
- Research Center, Shandong Medical College, Linyi, 276000, Shandong, China.
| |
Collapse
|
53
|
Luo L, Zhuang X, Fu L, Dong Z, Yi S, Wang K, Jiang Y, Zhao J, Yang X, Hei F. The role of the interplay between macrophage glycolytic reprogramming and NLRP3 inflammasome activation in acute lung injury/acute respiratory distress syndrome. Clin Transl Med 2024; 14:e70098. [PMID: 39623879 PMCID: PMC11612265 DOI: 10.1002/ctm2.70098] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2024] [Revised: 10/26/2024] [Accepted: 11/04/2024] [Indexed: 12/06/2024] Open
Abstract
Acute lung injury (ALI)/acute respiratory distress syndrome (ARDS) is a severe respiratory condition associated with elevated morbidity and mortality. Understanding their complex pathophysiological mechanisms is crucial for developing new preventive and therapeutic strategies. Recent studies highlight the significant role of inflammation involved in ALI/ARDS, particularly the hyperactivation of the NOD-like receptor thermal protein domain-associated protein 3 (NLRP3) inflammasome in macrophages. This activation drives pulmonary inflammation by releasing inflammatory signalling molecules and is linked to metabolic reprogramming, marked by increased glycolysis and reduced oxidative phosphorylation. However, the relationship between NLRP3 inflammasome activation and macrophage glycolytic reprogramming in ALI/ARDS, as well as the molecular mechanisms regulating these processes, remain elusive. This review provides a detailed description of the interactions and potential mechanisms linking NLRP3 inflammasome activation with macrophage glycolytic reprogramming, proposing that glycolytic reprogramming may represent a promising therapeutic target for mitigating inflammatory responses in ALI/ARDS. KEY POINTS: NLRP3 inflammasome activation is pivotal in mediating the excessive inflammatory response in ALI/ARDS. Glycolytic reprogramming regulates NLRP3 inflammasome activation. Therapeutic potential of targeting glycolytic reprogramming to inhibit NLRP3 inflammasome activation in ALI/ARDS.
Collapse
Affiliation(s)
- Lan Luo
- Department of Extracorporeal Circulation and Mechanical Circulation AssistantsCenter for Cardiac Intensive CareBeijing Anzhen HospitalCapital Medical UniversityBeijingChina
| | - Xiaoli Zhuang
- Department of Extracorporeal Circulation and Mechanical Circulation AssistantsCenter for Cardiac Intensive CareBeijing Anzhen HospitalCapital Medical UniversityBeijingChina
| | - Lin Fu
- Department of Extracorporeal Circulation and Mechanical Circulation AssistantsCenter for Cardiac Intensive CareBeijing Anzhen HospitalCapital Medical UniversityBeijingChina
| | - Ziyuan Dong
- Department of Extracorporeal Circulation and Mechanical Circulation AssistantsCenter for Cardiac Intensive CareBeijing Anzhen HospitalCapital Medical UniversityBeijingChina
| | - Shuyuan Yi
- Department of Extracorporeal Circulation and Mechanical Circulation AssistantsCenter for Cardiac Intensive CareBeijing Anzhen HospitalCapital Medical UniversityBeijingChina
| | - Kan Wang
- Department of Extracorporeal Circulation and Mechanical Circulation AssistantsCenter for Cardiac Intensive CareBeijing Anzhen HospitalCapital Medical UniversityBeijingChina
| | - Yu Jiang
- Department of Extracorporeal Circulation and Mechanical Circulation AssistantsCenter for Cardiac Intensive CareBeijing Anzhen HospitalCapital Medical UniversityBeijingChina
| | - Ju Zhao
- Department of Extracorporeal Circulation and Mechanical Circulation AssistantsCenter for Cardiac Intensive CareBeijing Anzhen HospitalCapital Medical UniversityBeijingChina
| | - Xiaofang Yang
- Department of Extracorporeal Circulation and Mechanical Circulation AssistantsCenter for Cardiac Intensive CareBeijing Anzhen HospitalCapital Medical UniversityBeijingChina
| | - Feilong Hei
- Department of Extracorporeal Circulation and Mechanical Circulation AssistantsCenter for Cardiac Intensive CareBeijing Anzhen HospitalCapital Medical UniversityBeijingChina
| |
Collapse
|
54
|
Chakraborty S, Banerjee M, Jayaraman G, Rajeswari V D. Evaluation of the health impacts and deregulation of signaling pathways in humans induced by microplastics. CHEMOSPHERE 2024; 369:143881. [PMID: 39631686 DOI: 10.1016/j.chemosphere.2024.143881] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Revised: 11/29/2024] [Accepted: 12/01/2024] [Indexed: 12/07/2024]
Abstract
This review assesses the diverse health risk factors associated with microplastic (MP) exposure and their impact on cellular signaling pathways. MPs induce chronic inflammation, oxidative stress, endocrine disruption, apoptosis, and immune dysregulation. They activate signaling pathways such as NF-κB, MAPK, and Nrf2, exacerbating inflammatory responses, oxidative damage, and hormonal imbalances. Understanding the interplay between MPs and signaling pathways is crucial for elucidating the mechanisms underlying MP-induced health effects. Effective risk assessment and management strategies are essential to mitigate the adverse health impacts of MPs on human populations. This research underscores the urgent need for interdisciplinary collaboration to safeguard human health and environmental sustainability in the face of rising MP pollution. In this paper, we also assess the risk factors caused by the microplastics in the pregnant women and the development of the fetus. This review explores the potential risks and challenges associated with MP exposure in newborn babies. It is quite concerning that microplastic particles were recently found in the placental tissue of newborn children for the first time. Although it is unclear how these tiny particles affect different organs, researchers believe that these tiny particles could potentially carry harmful chemicals or disrupt the developing immune system of the fetus. This review overall focuses on the impact of microplastic disrupting different signaling including reproductive health in humans.
Collapse
Affiliation(s)
- Shreya Chakraborty
- Department of Bio-Medical Sciences, School of Biosciences and Technology, Vellore Institute of Technology, Vellore, Tamil Nadu, 632014, India
| | - Manosi Banerjee
- Department of Bio-Medical Sciences, School of Biosciences and Technology, Vellore Institute of Technology, Vellore, Tamil Nadu, 632014, India
| | - Gurunathan Jayaraman
- Department of Bio-Sciences, School of Biosciences and Technology, Vellore Institute of Technology, Vellore, Tamil Nadu, 632014, India
| | - Devi Rajeswari V
- Department of Bio-Medical Sciences, School of Biosciences and Technology, Vellore Institute of Technology, Vellore, Tamil Nadu, 632014, India.
| |
Collapse
|
55
|
Ruan YB, Liu XH, Jiang JZ, Nie T, Ma J. Leydig cells pyroptosis in testis mediates deoxynivalenol-induced male reproductive toxicity in mice. THE SCIENCE OF THE TOTAL ENVIRONMENT 2024; 954:176432. [PMID: 39312968 DOI: 10.1016/j.scitotenv.2024.176432] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Revised: 09/18/2024] [Accepted: 09/19/2024] [Indexed: 09/25/2024]
Abstract
Deoxynivalenol (DON) is a toxic secondary metabolite produced by Fusarium spp. It is widely distributed among various cereals and has attracted much attention as a potential health threat to humans and domestic animals. However, the effects of DON on the reproductive systems of mammals are still ambiguous. In this study, the toxic effects of DON in the male reproduction of mice were investigated. The results showed that DON caused the shedding of sperm cells at all testis levels and the presence of inflammatory cells in the testicular interstitium. The rate of living sperm was significantly reduced, and the rate of sperm deformity was increased after DON exposure. The DON exposure resulted in decreased levels of testosterone (T) and increased levels of follicle-stimulating hormone (FSH) and luteinizing hormone (LH) in the serum. Measurements of oxidative stress markers showed that DON induced oxidative stress in mice testis. Meanwhile, DON triggered the assembly of NLRP3-ASC-Caspase-1 inflammatory complex and pyroptosis in both mice testis and TM3 cells, further causing the activation of GSDMD, promoting the leakage of inflammatory cytokines, including IL-1β and IL-18. Notably, the inhibition of oxidative stress was found to protect pyroptosis in TM3 cells exposed to DON. We identified a novel mechanism of reproductive damage induced by DON, demonstrating the activation of the canonical Caspase-1-dependent pyroptosis pathway and clarifying the protection of antioxidation against pyroptosis damage. Our discovery provided support for the risk assessment of DON and target exploration for clinical treatment related to pyroptosis.
Collapse
Affiliation(s)
- Yong-Bao Ruan
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, PR China
| | - Xiao-Hui Liu
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, PR China
| | - Jun-Ze Jiang
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, PR China
| | - Tong Nie
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, PR China
| | - Jun Ma
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, PR China; Heilongjiang Provincial Key Laboratory of Pathogenic Mechanism for Animal Disease and Comparative Medicine, Harbin 150030, PR China.
| |
Collapse
|
56
|
Han S, Bian R, Chen Y, Liang J, Zhao P, Gu Y, Zhang D. Dysregulation of the Gut Microbiota Contributes to Sevoflurane-Induced Cognitive Dysfunction in Aged Mice by Activating the NLRP3 Inflammasome. Mol Neurobiol 2024; 61:10500-10516. [PMID: 38740706 DOI: 10.1007/s12035-024-04229-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2024] [Accepted: 05/09/2024] [Indexed: 05/16/2024]
Abstract
Postoperative cognitive dysfunction (POCD), a common complication in elderly patients after surgery, seriously affects patients' quality of life. Long-term or repeated inhalation of sevoflurane can cause neuroinflammation, which is a risk factor for POCD. However, the underlying mechanism needs to be further explored. Recent research had revealed a correlation between neurological disorders and changes in the gut microbiota. Dysfunction of the gut microbiota is involved in the occurrence and development of central nervous system diseases. Here, we found that cognitive dysfunction in aged mice induced by sevoflurane exposure (3%, 2 hours daily, for 3 days) was related to gut microbiota dysbiosis, while probiotics improved cognitive function by alleviating dysbiosis. Sevoflurane caused a significant decrease in the abundance of Akkermansia (P<0.05), while probiotics restored the abundance of Akkermansia. Compared to those in the control group, sevoflurane significantly increased the expression of NLRP3 inflammasome-associated proteins in the gut and brain in the sevoflurane-exposed group, thus causing neuroinflammation and synaptic damage, which probiotics can mitigate (con vs. sev, P < 0.01; p+sev vs. sev, P < 0.05). In conclusion, for the first time, our study revealed that dysbiosis of the gut microbiota caused by sevoflurane anesthesia contributes to the NLRP3 inflammasome-mediated neuroinflammation and cognitive dysfunction from the perspective of the gut-brain axis. Perhaps postoperative cognitive impairment in elderly patients can be alleviated or even prevented by regulating the gut microbiota. This study provides new insights and methods for the prevention and treatment of cognitive impairment induced by sevoflurane.
Collapse
Affiliation(s)
- Shanshan Han
- Department of Anesthesiology, Wuxi Maternal and Child Health Care Hospital Affiliated to Jiangnan University, Wuxi, 214002, China
- Department of Basic Medicine, Wuxi School of Medicine, Jiangnan University, Wuxi, 214122, China
| | - Ruxi Bian
- Department of Basic Medicine, Wuxi School of Medicine, Jiangnan University, Wuxi, 214122, China
| | - Yuxuan Chen
- Department of Basic Medicine, Wuxi School of Medicine, Jiangnan University, Wuxi, 214122, China
| | - Junjie Liang
- Department of Basic Medicine, Wuxi School of Medicine, Jiangnan University, Wuxi, 214122, China
| | - Peng Zhao
- Department of Basic Medicine, Wuxi School of Medicine, Jiangnan University, Wuxi, 214122, China
| | - Yanfang Gu
- Department of Gynecology, Jiangnan University Affiliated Hospital, Wuxi, 214002, China.
| | - Dengxin Zhang
- Department of Anesthesiology, Wuxi Maternal and Child Health Care Hospital Affiliated to Jiangnan University, Wuxi, 214002, China.
| |
Collapse
|
57
|
Jing D, Liu J, Qin D, Lin J, Li T, Li Y, Duan M. Obeticholic acid ameliorates sepsis-induced renal mitochondrial damage by inhibiting the NF-κb signaling pathway. Ren Fail 2024; 46:2368090. [PMID: 39108162 PMCID: PMC11308967 DOI: 10.1080/0886022x.2024.2368090] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Revised: 05/19/2024] [Accepted: 06/10/2024] [Indexed: 08/10/2024] Open
Abstract
Acute kidney injury (AKI), a common complication of sepsis, might be caused by overactivated inflammation, mitochondrial damage, and oxidative stress. However, the mechanisms underlying sepsis-induced AKI (SAKI) have not been fully elucidated, and there is a lack of effective therapies for AKI. To this end, this study aimed to investigate whether obeticholic acid (OCA) has a renoprotective effect on SAKI and to explore its mechanism of action. Through bioinformatics analysis, our study confirmed that the mitochondria might be a critical target for the treatment of SAKI. Thus, a septic rat model was established by cecal ligation puncture (CLP) surgery. Our results showed an evoked inflammatory response via the NF-κB signaling pathway and NLRP3 inflammasome activation in septic rats, which led to mitochondrial damage and oxidative stress. OCA, an Farnesoid X Receptor (FXR) agonist, has shown anti-inflammatory effects in numerous studies. However, the effects of OCA on SAKI remain unclear. In this study, we revealed that pretreatment with OCA can inhibit the inflammatory response by reducing the synthesis of proinflammatory factors (such as IL-1β and NLRP3) via blocking NF-κB and alleviating mitochondrial damage and oxidative stress in the septic rat model. Overall, this study provides insight into the excessive inflammation-induced SAKI caused by mitochondrial damage and evidence for the potential use of OCA in SAKI treatment.
Collapse
Affiliation(s)
- Danyang Jing
- Department of Critical Care Medicine, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Jingfeng Liu
- Department of Critical Care Medicine, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Da Qin
- Department of Gastroenterology, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Jin Lin
- Department of Critical Care Medicine, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Tian Li
- Department of Critical Care Medicine, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Yu Li
- Department of Critical Care Medicine, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Meili Duan
- Department of Critical Care Medicine, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
58
|
Zhou YT, Li S, Du SL, Zhao JH, Cai YQ, Zhang ZQ. The multifaceted role of macrophage mitophagy in SiO 2-induced pulmonary fibrosis: A brief review. J Appl Toxicol 2024; 44:1854-1867. [PMID: 38644760 DOI: 10.1002/jat.4612] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2024] [Revised: 03/19/2024] [Accepted: 03/28/2024] [Indexed: 04/23/2024]
Abstract
Prolonged exposure to environments with high concentrations of crystalline silica (CS) can lead to silicosis. Macrophages play a crucial role in the pathogenesis of silicosis. In the process of silicosis, silica (SiO2) invades alveolar macrophages (AMs) and induces mitophagy which usually exists in three states: normal, excessive, and/or deficiency. Different mitophagy states lead to corresponding toxic responses, including successful macrophage repair, injury, necrosis, apoptosis, and even pulmonary fibrosis. This is a complex process accompanied by various cytokines. Unfortunately, the details have not been fully systematically summarized. Therefore, it is necessary to elucidate the role of macrophage mitophagy in SiO2-induced pulmonary fibrosis by systematic analysis on the literature reports. In this review, we first summarized the current data on the macrophage mitophagy in the development of SiO2-induced pulmonary fibrosis. Then, we introduce the molecular mechanism on how SiO2-induced mitophagy causes pulmonary fibrosis. Finally, we focus on introducing new therapies based on newly developed mitophagy-inducing strategies. We conclude that macrophage mitophagy plays a multifaceted role in the progression of SiO2-induced pulmonary fibrosis, and reprogramming the macrophage mitophagy state accordingly may be a potential means of preventing and treating pulmonary fibrosis.
Collapse
Affiliation(s)
- Yu-Ting Zhou
- Department of Public Health, Shandong First Medical University, Jinan, China
- Department of Public Health, Jining Medical University, Jining, China
| | - Shuang Li
- Department of Public Health, Jining Medical University, Jining, China
| | - Shu-Ling Du
- Department of Public Health, Jining Medical University, Jining, China
| | - Jia-Hui Zhao
- Department of Public Health, Jining Medical University, Jining, China
| | | | - Zhao-Qiang Zhang
- Department of Public Health, Jining Medical University, Jining, China
| |
Collapse
|
59
|
Xia Y, Zou C, Kang W, Xu T, Shao R, Zeng P, Sun B, Chen J, Qi Y, Wang Z, Lin T, Zhu H, Shen Y, Wang X, Guo S, Cui D. Invasive metastatic tumor-camouflaged ROS responsive nanosystem for targeting therapeutic brain injury after cardiac arrest. Biomaterials 2024; 311:122678. [PMID: 38917705 DOI: 10.1016/j.biomaterials.2024.122678] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2023] [Revised: 05/28/2024] [Accepted: 06/22/2024] [Indexed: 06/27/2024]
Abstract
Drug transmission through the blood-brain barrier (BBB) is considered an arduous challenge for brain injury treatment following the return of spontaneous circulation after cardiac arrest (CA-ROSC). Inspired by the propensity of melanoma metastasis to the brain, B16F10 cell membranes are camouflaged on 2-methoxyestradiol (2ME2)-loaded reactive oxygen species (ROS)-triggered "Padlock" nanoparticles that are constructed by phenylboronic acid pinacol esters conjugated D-a-tocopheryl polyethylene glycol succinate (TPGS-PBAP). The biomimetic nanoparticles (BM@TP/2ME2) can be internalized, mainly mediated by the mutual recognition and interaction between CD44v6 expressed on B16F10 cell membranes and hyaluronic acid on cerebral vascular endothelial cells, and they responsively release 2ME2 by the oxidative stress microenvironment. Notably, BM@TP/2ME2 can scavenge excessive ROS to reestablish redox balance, reverse neuroinflammation, and restore autophagic flux in damaged neurons, eventually exerting a remarkable neuroprotective effect after CA-ROSC in vitro and in vivo. This biomimetic drug delivery system is a novel and promising strategy for the treatment of cerebral ischemia-reperfusion injury after CA-ROSC.
Collapse
Affiliation(s)
- Yiyang Xia
- Department of Anesthesiology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, PR China
| | - Chenming Zou
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai, 200240, PR China
| | - Weichao Kang
- Department of Anesthesiology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, PR China
| | - Tianhua Xu
- Department of Anesthesiology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, PR China
| | - Rongjiao Shao
- Department of Critical Care Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200030, PR China
| | - Ping Zeng
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai, 200240, PR China
| | - Bixi Sun
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai, 200240, PR China
| | - Jie Chen
- Department of Anesthesiology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, PR China
| | - Yiming Qi
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai, 200240, PR China
| | - Zhaozhong Wang
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai, 200240, PR China
| | - Tiancheng Lin
- Department of Anesthesiology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, PR China
| | - Haichao Zhu
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai, 200240, PR China
| | - Yuanyuan Shen
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai, 200240, PR China
| | - Xintao Wang
- Department of Anesthesiology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, PR China.
| | - Shengrong Guo
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai, 200240, PR China.
| | - Derong Cui
- Department of Anesthesiology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, PR China.
| |
Collapse
|
60
|
Zhang C, Qiao P, Zhang J, Luo Y, Xiao C, Shen S, Hasegawa A, Qiao H, Wang G, Abe R, Fu M. A carbamazepine metabolite activates NLRP3 and controls skin homing of CD8 + T-cells in SJS/TEN. J Dermatol Sci 2024; 116:80-89. [PMID: 39500682 DOI: 10.1016/j.jdermsci.2024.10.003] [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: 11/28/2022] [Revised: 10/05/2024] [Accepted: 10/18/2024] [Indexed: 12/09/2024]
Abstract
BACKGROUND Stevens-Johnson syndrome (SJS) and toxic epidermal necrolysis (TEN) are severe adverse drug reactions with extensive keratinocyte death. Carbamazepine (CBZ), the most commonly implicated drug in SJS/TEN, is metabolized by the cytochrome P450 enzyme 3A4 (CYP3A4) into carbamazepine-10,11-epoxide (CBZE) in the liver. While CD8+ cytotoxic T cells play an important role in SJS/TEN, the underlying mechanism of exuberant immune response by CD8+ T cells in these conditions remains incompletely understood. OBJECTIVES To examine the expression of NLRP3 inflammasome and their skin migration in CBZE-induced SJS/TEN. METHODS The expression of the NLRP3 inflammasome complex in skin lesions, sera, and blister fluids of SJS/TEN patients were analyzed by immunohistochemistry and enzyme-linked immunosorbent assay. NLRP3 formation and CD8+ T cell activation status and their functions were examined by immunoblotting, immunofluorescence, and chemotaxis assays. RESULTS The expression of the NLRP3 inflammasome complex was greatly increased in skin lesions of SJS/TEN patients. Moreover, IL-1β and IL-18 levels in sera and blister fluids of SJS/TEN patients were approximately 3-fold higher than those in healthy individuals, with a linear correlation between IL-1β levels and disease activity. CBZE induced NLRP3 inflammasome formation, upregulated CXCL9/CXCL10 levels, and activated CD8+ cytotoxic T cell functions via IL-1β/IL-1R or IL-18/IL-18R signaling in SJS/TEN keratinocytes, which promoted CD8+ cytotoxic T cell migration in SJS/TEN patients. CONCLUSION This study showed that CBZE promoted NLRP3 inflammasome formation and strengthened the activation and function of CD8+ cytotoxic T cells in the skin, which contributed to the initiation and progression of SJS/TEN.
Collapse
Affiliation(s)
- Chen Zhang
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Pei Qiao
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - JieYu Zhang
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - YiXin Luo
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - ChunYing Xiao
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - ShengXian Shen
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Akio Hasegawa
- Division of Dermatology, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - HongJiang Qiao
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Gang Wang
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Riichiro Abe
- Division of Dermatology, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan.
| | - Meng Fu
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, Xi'an, China.
| |
Collapse
|
61
|
Jangra J, Bajad NG, Singh R, Kumar A, Singh SK. Identification of novel potential cathepsin-B inhibitors through pharmacophore-based virtual screening, molecular docking, and dynamics simulation studies for the treatment of Alzheimer's disease. Mol Divers 2024; 28:4381-4401. [PMID: 38517648 DOI: 10.1007/s11030-024-10821-z] [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: 11/28/2023] [Accepted: 02/03/2024] [Indexed: 03/24/2024]
Abstract
Cathepsin B is a cysteine protease lysosomal enzyme involved in several physiological functions. Overexpression of the enzyme enhances its proteolytic activity and causes the breakdown of amyloid precursor protein (APP) into neurotoxic amyloid β (Aβ), a characteristic hallmark of Alzheimer's disease (AD). Therefore, inhibition of the enzyme is a crucial therapeutic aspect for treating the disease. Combined structure and ligand-based drug design strategies were employed in the current study to identify the novel potential cathepsin B inhibitors. Five different pharmacophore models were developed and used for the screening of the ZINC-15 database. The obtained hits were analyzed for the presence of duplicates, interfering PAINS moieties, and structural similarities based on Tanimoto's coefficient. The molecular docking study was performed to screen hits with better target binding affinity. The top seven hits were selected and were further evaluated based on their predicted ADME properties. The resulting best hits, ZINC827855702, ZINC123282431, and ZINC95386847, were finally subjected to molecular dynamics simulation studies to determine the stability of the protein-ligand complex during the run. ZINC123282431 was obtained as the virtual lead compound for cathepsin B inhibition and may be a promising novel anti-Alzheimer agent.
Collapse
Affiliation(s)
- Jatin Jangra
- Pharmaceutical Chemistry Research Laboratory-I, Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (Banaras Hindu University), Varanasi, 221005, India
| | - Nilesh Gajanan Bajad
- Pharmaceutical Chemistry Research Laboratory-I, Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (Banaras Hindu University), Varanasi, 221005, India
| | - Ravi Singh
- Pharmaceutical Chemistry Research Laboratory-I, Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (Banaras Hindu University), Varanasi, 221005, India
| | - Ashok Kumar
- Pharmaceutical Chemistry Research Laboratory-I, Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (Banaras Hindu University), Varanasi, 221005, India
| | - Sushil Kumar Singh
- Pharmaceutical Chemistry Research Laboratory-I, Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (Banaras Hindu University), Varanasi, 221005, India.
| |
Collapse
|
62
|
Tang LP, Guo YN, Mou RW, Liang SS, Lu MH, He YM. Effects of Clausena lansium leaves volatile oil emulsion against Staphylococcus aureus in mice via autophagy modulation. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 135:156159. [PMID: 39442278 DOI: 10.1016/j.phymed.2024.156159] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Revised: 10/10/2024] [Accepted: 10/13/2024] [Indexed: 10/25/2024]
Abstract
BACKGROUND Volatile oil from fresh Clausena lansium (Lour.) Skeels (Rutaceae) (common name Wampee) has been previously extracted by our group from fresh C. lansium leaf and its components were qualitative and quantitatively analyzed by GC-MS. It altered the cell membrane permeability of Staphylococcus aureus and reduced the levels of inflammation factors. However, previous in vivo reports on the anti-inflammatory and the antibacterial properties against S. aureus are scarce. HYPOTHESIS/PURPOSE To evaluate the protective in vivo effects of Wampee leaves volatile oil emulsion (WVOE) against S. aureus-induced pneumonia and elucidate the underlying mechanisms of action. METHODS Wild-type and nucleotide oligomerization domain-like receptor protein 3 (NLRP3)-deficient mice were used. Mice were treated with WVOE for 7 days, and subjected to S. aureus infection by nasal administration on day 5 for 48 h. Lung and blood samples were collected for assessing lung damage and protein abundance. Lung bacterial load, wet/dry ratio, C-reactive protein (CRP) levels, inflammatory cytokines secretion, and lung histopathological injury were examined. RESULTS WVOE effectively reduced lung bacterial load, wet/dry ratio, and CRP levels increased following S. aureus infection in mice. WVOE decreased the secretion of inflammatory cytokines (IL-6 and TNF-α) and lung histopathological injury, and suppressed the NF-κB pathway and NLRP3 inflammasome activation. NLRP3-/- mice exhibited lower bacterial load, inflammatory cytokines levels and lung histopathological injury compared with mice in the model group. Autophagy was enhanced in S. aureus-infected mice, with higher levels of p-mTOR, Beclin-1, Atg 16L1, Atg7, p62, p-p62, and LC3II. WVOE administration restored the autophagy related protein levels. Autophagy was inhibited in NLRP3-/- mice of the control and model groups, and WVOE lost its ability to regulate the autophagy-related proteins enhanced upon S. aureus infection. WVOE enhanced autophagy to alleviate lung injury by inhibiting NLRP3-targeted P62. Furthermore, compared with the 3MA + model group, WVOE reduced the bacterial load and CRP levels, pulmonary septa narrowing, and congestion. NLRP3 protein expression increased due to autophagy inhibition. WVOE exerted a pharmacological effect through the PI3K/AKT/mTOR pathway. CONCLUSION WVOE regulated the PI3K/AKT/mTOR pathway and enhanced autophagy, with NLRP3 playing a crucial role. WVOE exhibited protective effects against S. aureus-induced pneumonia by inhibiting NLRP3 inflammasome activation and enhancing autophagy. These findings expand the understanding of antibacterial properties of WVOE, and provide novel insights into the therapeutic potential of WVOE in managing S. aureus infections.
Collapse
Affiliation(s)
- Lu-Ping Tang
- School of Life Science and Engineering, Foshan University, Foshan 528225, China.
| | - Yan-Na Guo
- School of Life Science and Engineering, Foshan University, Foshan 528225, China
| | - Rui-Wei Mou
- School of Life Science and Engineering, Foshan University, Foshan 528225, China
| | - Shao-Shan Liang
- School of Life Science and Engineering, Foshan University, Foshan 528225, China
| | - Meng-Han Lu
- School of Life Science and Engineering, Foshan University, Foshan 528225, China
| | - Yong-Ming He
- School of Life Science and Engineering, Foshan University, Foshan 528225, China.
| |
Collapse
|
63
|
Pak SW, Kim WI, Lee SJ, Park SH, Cho YK, Kim JS, Kim JC, Kim SH, Shin IS. TXNIP regulates pulmonary inflammation induced by Asian sand dust. Redox Biol 2024; 78:103421. [PMID: 39520910 DOI: 10.1016/j.redox.2024.103421] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2024] [Revised: 11/04/2024] [Accepted: 11/05/2024] [Indexed: 11/16/2024] Open
Abstract
Asian sand dust (ASD), a seasonal dust storm originating from the deserts of China and Mongolia, affects Korea and Japan during the spring, carrying soil particles and a variety of biochemical components. Exposure to ASD has been associated with the onset and exacerbation of respiratory disorders, although the underlying mechanisms remain unclear. This study investigates ASD-induced pulmonary toxicity and its mechanistic pathways, focusing on the role of thioredoxin-interacting protein (TXNIP). Using TXNIP knock-out (KO) mice and adeno-associated virus (AAV)-mediated TXNIP overexpression transgenic mice, we explored how TXNIP modulates ASD-induced pulmonary inflammation. Mice were exposed to ASD via intranasal administration on days 1, 3, and 5 to induce inflammation. ASD exposure led to significant pulmonary inflammation, evidenced by increased inflammatory cell counts and elevated cytokine levels in bronchoalveolar lavage fluid, as well as heightened protein expression of the TXNIP/NOD-like receptor pyrin domain-containing 3 (NLRP3) inflammasome. TXNIP KO mice exhibited attenuated airway inflammation and downregulation of the NLRP3 inflammasome compared to wild-type controls, while AAV-mediated TXNIP overexpression mice showed exacerbated inflammatory responses, including elevated NLRP3 inflammasome expression, compared to AAV-GFP controls. These findings suggest that TXNIP is a key regulator of ASD-induced pulmonary inflammation.
Collapse
Affiliation(s)
- So-Won Pak
- College of Veterinary Medicine and BK21 FOUR Program, Chonnam National University, 77 Yong-bong-ro, Buk-gu, Gwangju, 61186, Republic of Korea
| | - Woong-Il Kim
- College of Veterinary Medicine and BK21 FOUR Program, Chonnam National University, 77 Yong-bong-ro, Buk-gu, Gwangju, 61186, Republic of Korea
| | - Se-Jin Lee
- College of Veterinary Medicine and BK21 FOUR Program, Chonnam National University, 77 Yong-bong-ro, Buk-gu, Gwangju, 61186, Republic of Korea
| | - Sin-Hyang Park
- College of Veterinary Medicine and BK21 FOUR Program, Chonnam National University, 77 Yong-bong-ro, Buk-gu, Gwangju, 61186, Republic of Korea
| | - Young-Kwon Cho
- College of Health Sciences, Cheongju University, 298 Daesung-ro, Sangdang-gu, Cheongju-si, Chungbuk, 28503, Republic of Korea
| | - Joong-Sun Kim
- College of Veterinary Medicine and BK21 FOUR Program, Chonnam National University, 77 Yong-bong-ro, Buk-gu, Gwangju, 61186, Republic of Korea
| | - Jong-Choon Kim
- College of Veterinary Medicine and BK21 FOUR Program, Chonnam National University, 77 Yong-bong-ro, Buk-gu, Gwangju, 61186, Republic of Korea
| | - Sung-Hwan Kim
- Jeonbuk Branch, Korea Institute of Toxicology (KIT), Jeongeup-si, Jeonbuk, 53212, Republic of Korea.
| | - In-Sik Shin
- College of Veterinary Medicine and BK21 FOUR Program, Chonnam National University, 77 Yong-bong-ro, Buk-gu, Gwangju, 61186, Republic of Korea.
| |
Collapse
|
64
|
Kou Y, Jing Q, Yan X, Chen J, Shen Y, Ma Y, Xiang Y, Li X, Liu X, Liu Z, Wei Y, Wang Y. 4-Hydroxybenzoic acid restrains Nlrp3 inflammasome priming and activation via disrupting PU.1 DNA binding activity and direct antioxidation. Chem Biol Interact 2024; 404:111262. [PMID: 39389441 DOI: 10.1016/j.cbi.2024.111262] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Revised: 08/30/2024] [Accepted: 10/08/2024] [Indexed: 10/12/2024]
Abstract
Reactive oxygen species (ROS) production is considered central to triggering the nucleotide-binding domain-like receptor family pyrin domain containing 3 (Nlrp3) inflammasome activation and the subsequent inflammatory responses. Coenzyme Q10 (CoQ10) plays a critical role in maintaining intracellular ROS homeostasis and inhibiting excessive Nlrp3 inflammasome activation. However, direct supplementation of CoQ10 showed unsatisfactory clinical improvement due to its limited absorption and bioavailability. Therefore, stimulating endogenous CoQ10 biosynthesis by supplementing CoQ10 precursors may provide a more promising therapeutic approach. In this study, we described the role of 4-hydroxybenzoic acid (4-HBA), a precursor of CoQ10, in attenuating excessive inflammatory responses. We found that while supplementation of 4-HBA inhibited the priming and activation of Nlrp3 inflammasome, this effect was independent of its metabolic transformation into CoQ10. 4-HBA itself exhibits antioxidative activities. Furthermore, 4-HBA can disrupt the binding activity of PU.1 on the promoters of Tlr4 and Md2, thereby directly suppressing Nlrp3 inflammasome priming during LPS-induced inflammatory responses. Therefore, strategically utilizing 4-HBA or increasing 4-HBA intake may represent a potential strategy for reducing excessive inflammation.
Collapse
Affiliation(s)
- Yanbo Kou
- Laboratory of Infection and Immunity, Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogenic Biology and Immunology, School of Basic Medicine, Xuzhou Medical University, Xuzhou, China; Jiangsu International Laboratory of Immunity and Metabolism, China
| | - Qiyue Jing
- Laboratory of Infection and Immunity, Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogenic Biology and Immunology, School of Basic Medicine, Xuzhou Medical University, Xuzhou, China; Jiangsu International Laboratory of Immunity and Metabolism, China
| | - Xiaoqing Yan
- Laboratory of Emergency Medicine, Second Clinical Medical College, Xuzhou Medical University, Xuzhou, China
| | - Junru Chen
- Laboratory of Infection and Immunity, Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogenic Biology and Immunology, School of Basic Medicine, Xuzhou Medical University, Xuzhou, China; Jiangsu International Laboratory of Immunity and Metabolism, China
| | - Yusi Shen
- Laboratory of Infection and Immunity, Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogenic Biology and Immunology, School of Basic Medicine, Xuzhou Medical University, Xuzhou, China; Jiangsu International Laboratory of Immunity and Metabolism, China
| | - Yulu Ma
- Laboratory of Infection and Immunity, Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogenic Biology and Immunology, School of Basic Medicine, Xuzhou Medical University, Xuzhou, China; Jiangsu International Laboratory of Immunity and Metabolism, China
| | - Yaoyao Xiang
- Laboratory of Infection and Immunity, Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogenic Biology and Immunology, School of Basic Medicine, Xuzhou Medical University, Xuzhou, China; Jiangsu International Laboratory of Immunity and Metabolism, China
| | - Xiangyang Li
- Laboratory of Infection and Immunity, Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogenic Biology and Immunology, School of Basic Medicine, Xuzhou Medical University, Xuzhou, China; Jiangsu International Laboratory of Immunity and Metabolism, China
| | - Xiangye Liu
- Laboratory of Infection and Immunity, Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogenic Biology and Immunology, School of Basic Medicine, Xuzhou Medical University, Xuzhou, China; Jiangsu International Laboratory of Immunity and Metabolism, China
| | - Zhuanzhuan Liu
- Laboratory of Infection and Immunity, Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogenic Biology and Immunology, School of Basic Medicine, Xuzhou Medical University, Xuzhou, China; Jiangsu International Laboratory of Immunity and Metabolism, China
| | - Yanxia Wei
- Laboratory of Infection and Immunity, Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogenic Biology and Immunology, School of Basic Medicine, Xuzhou Medical University, Xuzhou, China; Jiangsu International Laboratory of Immunity and Metabolism, China
| | - Yugang Wang
- Laboratory of Infection and Immunity, Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogenic Biology and Immunology, School of Basic Medicine, Xuzhou Medical University, Xuzhou, China; Jiangsu International Laboratory of Immunity and Metabolism, China.
| |
Collapse
|
65
|
Hua Z, Deng J, Wang G. Moringa isothiocyanate-1 mitigates the damage of oxidative stress and apoptosis in diabetic nephropathy mice. Histol Histopathol 2024; 39:1621-1629. [PMID: 38623789 DOI: 10.14670/hh-18-741] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/17/2024]
Abstract
OBJECTIVE Diabetic nephropathy (DN) is a prevalent cause of end-stage kidney disease worldwide. Moringa isothiocyanate-1 (MIC-1) has shown potential for DN management, however, the exact mechanisms remain unclear. This research intended to evaluate the impact and mechanism of MIC-1 on DN. METHODS Six C57BLKS/J-db/m mice served as controls. Eighteen C57BLKS/J-db/db mice were randomly separated into three groups: db/db, db/db + irbesartan (IBS), and db/db + MIC-1. Three weeks post-drug administration, the body weight and kidney weight of mice in each group were measured. Concurrently, serum creatinine (Scr), urine albumin, insulin, glycosylated hemoglobin (GHb), oxidative stress-, and inflammatory-related factors were determined. Additionally, the pathological injury, apoptosis, apoptosis-related markers, NLRP3, and ASC levels in the kidney tissues were examined utilizing H&E, Masson, PAS, TUNEL staining, and Western blot. RESULTS MIC-1 decreased the body weight, kidney weight, the levels of Glu, Scr, and urine albumin in db/db mice. Moreover, MIC-1 significantly suppressed the levels of MDA, insulin, GHb, TNF-α, IL-1β, and IL-6, while increased the activities of SOD, CAT, and GPX in the serum of db/db mice. MIC-1 also mitigated the kidney tissue injury in db/db mice. Western blot assay showed that MIC-1 enhanced the Bcl-2 level and suppressed the Bax, cleaved caspase-3, cleaved caspase-9, NLRP3, ASC, and caspase-1 levels of the kidney tissues in db/db mice. CONCLUSIONS MIC-1 ameliorated the kidney injury in DN mice, and its mechanism may be associated with the suppression of renal cell apoptosis, oxidative stress, and inflammatory responses.
Collapse
Affiliation(s)
- Zhou Hua
- Department of Nephrology, The People's Hospital of Suichang County, Lishui City, Zhejiang Province, China
| | - Jiuhong Deng
- Department of Endocrinology, Second People's Hospital of Pingyang County, Wenzhou City, Zhejiang Province, China
| | - Guiying Wang
- Department of Nephrology, Shangyu People's Hospital of Shaoxing, Shaoxing City, Zhejiang Province, China.
| |
Collapse
|
66
|
Xu S, Wang D, Tan L, Lu J. The role of NLRP3 inflammasome in type 2 inflammation related diseases. Autoimmunity 2024; 57:2310269. [PMID: 38332696 DOI: 10.1080/08916934.2024.2310269] [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: 08/30/2023] [Accepted: 01/21/2024] [Indexed: 02/10/2024]
Abstract
Type 2 inflammation related diseases, such as atopic dermatitis, asthma, and allergic rhinitis, are diverse and affect multiple systems in the human body. It is common for individuals to have multiple co-existing type 2 inflammation related diseases, which can impose a significant financial and living burden on patients. However, the exact pathogenesis of these diseases is still unclear. The NLRP3 inflammasome is a protein complex composed of the NLRP3 protein, ASC, and Caspase-1, and is activated through various mechanisms, including the NF-κB pathway, ion channels, and lysosomal damage. The NLRP3 inflammasome plays a role in the immune response to pathogens and cellular damage. Recent studies have indicated a strong correlation between the abnormal activation of NLRP3 inflammasome and the onset of type 2 inflammation. Additionally, it has been demonstrated that suppressing NLRP3 expression effectively diminishes the inflammatory response, highlighting its promising therapeutic applications. Therefore, this article reviews the role of NLRP3 inflammasome in the development and therapy of multiple type 2 inflammation related diseases.
Collapse
Affiliation(s)
- Shenming Xu
- Department of Dermatology, Third Xiangya Hospital, Central South University, Changsha, Hunan, People's Republic of China
- Medical Ozone Research Center of Central South University, Changsha, Hunan, People's Republic of China
| | - Dan Wang
- Department of Dermatology, Third Xiangya Hospital, Central South University, Changsha, Hunan, People's Republic of China
- Medical Ozone Research Center of Central South University, Changsha, Hunan, People's Republic of China
| | - Lina Tan
- Department of Dermatology, Third Xiangya Hospital, Central South University, Changsha, Hunan, People's Republic of China
- Medical Ozone Research Center of Central South University, Changsha, Hunan, People's Republic of China
| | - Jianyun Lu
- Department of Dermatology, Third Xiangya Hospital, Central South University, Changsha, Hunan, People's Republic of China
- Medical Ozone Research Center of Central South University, Changsha, Hunan, People's Republic of China
| |
Collapse
|
67
|
HANG W, WANG L, BO Y, ZUO S, WANG S, LI H, BU C, ZHAO J, ZHOU X. Bufei Huoxue capsule alleviates silicosis by inhibiting the activation of nucleotide-like receptor containing pyrin domain 3 inflammasome and macrophages polarization based on network pharmacology. J TRADIT CHIN MED 2024; 44:1236-1246. [PMID: 39617709 PMCID: PMC11589560 DOI: 10.19852/j.cnki.jtcm.20240626.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2023] [Accepted: 03/08/2024] [Indexed: 12/17/2024]
Abstract
OBJECTIVE To predict the targets of Bufei Huoxue capsule (, BFHX) using network pharmacology analysis and to explore its effects and functional targets in a silicotic rat model. METHODS The drug and disease targets were correlated through network pharmacology analysis to explore the targets and signaling pathways of BFHX affecting silicosis. NR8383 cells were cultured to verify the core genes and pathways. A rat model of silicosis was established to verify whether the mechanism behind SiO2-caused pulmonary fibrosis was alleviated by BFHX (0.82 g/kg) and how it affected key targets and pathways. RESULTS Overlapping BFHX and silicotic gene targets produced 159 interactive targets, and 55 were screened by network topology analysis. The results of gene ontology and Kyoto encyclopedia of genes and genomes enrichment analyses suggested that BFHX could affect silicosis through the nucleotide-like receptor containing pyrin domain 3 (NLRP3) inflammasome. In NR8383 cells, the expression of core genes related to the NLRP3 inflammasome could be inhibited by BFHX treatment. BFHX reduced the degree of alveolitis and collagen deposition, attenuating pulmonary fibrosis in SiO2-induced rat model. Pulmonary macrophage pyroptosis after SiO2 exposure was observed under transmission electron microscopy. BFHX alleviated the morphological characteristics of pyroptosis. BFHX also reduced the expression of NLRP3, caspase-1, interleukin-1 beta (IL-1β), IL-18, IL-6, and tumor necrosis factor-alpha in lung tissues of silicotic rat model. BFHX affected the K ion content in bronchoalveolar lavage fluid when assessed by energy dispersive spectrometer testing. The expression of CD68+ and CD206+ were also reduced after BFHX intervention. CONCLUSION NOD-like receptor signaling is vital for BFHX's effects on silicosis. It exerts anti-pulmonary fibrosis effects by inhibiting pulmonary macrophage pyroptosis and polarization through NLRP3 inflammasome activation.
Collapse
Affiliation(s)
- Wenlu HANG
- 1 Department of Respiratory and Critical Care Medicine, Second Affiliated Hospital of Xuzhou Medical University, Xuzhou 221000, China
| | - Lin WANG
- 1 Department of Respiratory and Critical Care Medicine, Second Affiliated Hospital of Xuzhou Medical University, Xuzhou 221000, China
| | - Yun BO
- 2 Medical College of Anhui University of Science and Technology, Anhui 232001, China
| | - Shurun ZUO
- 1 Department of Respiratory and Critical Care Medicine, Second Affiliated Hospital of Xuzhou Medical University, Xuzhou 221000, China
| | - Songquan WANG
- 3 School of Mechatronic Engineering, Jiangsu Normal University, Xuzhou 221000, China
| | - Haiquan LI
- 1 Department of Respiratory and Critical Care Medicine, Second Affiliated Hospital of Xuzhou Medical University, Xuzhou 221000, China
| | - Chunlu BU
- 1 Department of Respiratory and Critical Care Medicine, Second Affiliated Hospital of Xuzhou Medical University, Xuzhou 221000, China
| | - Jie ZHAO
- 1 Department of Respiratory and Critical Care Medicine, Second Affiliated Hospital of Xuzhou Medical University, Xuzhou 221000, China
| | - Xianmei ZHOU
- 4 Department of Respiratory Medicine, Jiangsu Province Hospital of Chinese Medicine, Nanjing 210023, China
| |
Collapse
|
68
|
Ziehr BK, MacDonald JA. Regulation of NLRPs by reactive oxygen species: A story of crosstalk. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2024; 1871:119823. [PMID: 39173681 DOI: 10.1016/j.bbamcr.2024.119823] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/03/2024] [Revised: 06/28/2024] [Accepted: 08/14/2024] [Indexed: 08/24/2024]
Abstract
The nucleotide oligomerization domain (NOD)-like receptors containing pyrin (NLRP) family of cytosolic pattern-recognition receptors play an integral role in host defense following exposure to a diverse set of pathogenic and sterile threats. The canonical event following ligand recognition is the formation of a heterooligomeric signaling complex termed the inflammasome that produces pro-inflammatory cytokines. Dysregulation of this process is associated with many autoimmune, cardiovascular, metabolic, and neurodegenerative diseases. Despite the range of activating stimuli which affect varied cell types, recent literature makes evident that reactive oxygen species (ROS) are integral to the initiation and propagation of inflammasome signaling. Notably, ROS production and inflammasome activation act in a positive feedback loop to promote this potent immune response. While NLRP3 is by far the most extensively studied NLRP, there is also sufficient literature to make these conclusions for other NLRPs family members. In all cases, a knowledge gap exists regarding the molecular targets and effects of ROS. Future research to define these targets and to parse the order and timing of ROS-mediated NLRP activation will provide meaningful insights into inflammasome biology. This will create novel therapeutic opportunities for the numerous illnesses that are impacted by inflammasome activity.
Collapse
Affiliation(s)
- Bjoern K Ziehr
- Department of Biochemistry & Molecular Biology, Cumming School of Medicine, University of Calgary, Calgary, Alberta T2N 4Z6, Canada
| | - Justin A MacDonald
- Department of Biochemistry & Molecular Biology, Cumming School of Medicine, University of Calgary, Calgary, Alberta T2N 4Z6, Canada.
| |
Collapse
|
69
|
Gao X, Feng X, Hou T, Huang W, Ma Z, Zhang D. The roles of flavonoids in the treatment of inflammatory bowel disease and extraintestinal manifestations: A review. FOOD BIOSCI 2024; 62:105431. [DOI: 10.1016/j.fbio.2024.105431] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
70
|
Wei J, Tian Y, Wei J, Guan M, Yu X, Xie J, Fan G. Bilirubin regulates cell death type by alleviating macrophage mitochondrial dysfunction caused by cigarette smoke extract. Redox Rep 2024; 29:2382946. [PMID: 39074442 PMCID: PMC11288206 DOI: 10.1080/13510002.2024.2382946] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/31/2024] Open
Abstract
OBJECTIVES To explore the effects and mechanisms of bilirubin on mitochondrial function and type of macrophage cell death after exposure to cigarette smoke extract (CSE). METHODS RAW264.7 macrophages were treated with different concentrations of CSE and bilirubin solutions and divided into four groups: control, CSE, bilirubin, and bilirubin + CSE groups. The necrotic and apoptotic states of the macrophages were determined using an Annexin V-fluorescein 5-isothiocyanate/propidium iodide (FITC/PI) staining kit. Cytoplasmic NOD-like receptor family, pyrin domain containing 3 (NLRP3) expression in macrophages was detected by immunofluorescence and the levels of IL-1β and IL-18 in the supernatants of culture medium were detected by enzyme linked immunosorbent assay (ELISA) test. A JC-1 mitochondrial membrane potential detection kit was used to assess mitochondrial membrane damage and the adenosine triphosphate (ATP) assay kit was used to determine intracellular ATP levels. After the macrophages were stained with reactive oxygen species (ROS) specific dye, 2',7'-Dichlorodihydrofluorescein diacetate (DCFH-DA), the fluorescence intensity and proportion of ROS-positive macrophages were measured using flow cytometry. RESULTS We observed that compared with those of 0 μM (control group), concentrations of 5, 10, or 20 μΜ bilirubin significantly decreased cell viability, which was increased by bilirubin exposure below 1 μM. The effect of CSE on macrophage viability was concentration- and time-dependent. Bilirubin of 0.2 μM could alleviate the inhibition of macrophage viability caused by 5% CSE. In addition, bilirubin intervention could reduce the occurrence of necrosis and pyroptosis to a certain extent. CONCLUSIONS CSE could cause mitochondrial dysfunction in macrophages, as demonstrated by a decrease in mitochondrial membrane potential and intracellular ATP levels and an increase in ROS production, while bilirubin could relieve mitochondrial dysfunction caused by CSE.
Collapse
Affiliation(s)
- Jingjing Wei
- Department of Pediatrics, Shanxi Medical University, Taiyuan, Shanxi, People’s Republic of China
| | - Yuan Tian
- Department of Pediatrics, Shanxi Medical University, Taiyuan, Shanxi, People’s Republic of China
| | - Jinshu Wei
- Department of Pediatrics, Shanxi Medical University, Taiyuan, Shanxi, People’s Republic of China
| | - Meiqi Guan
- Department of Pediatrics, Shanxi Medical University, Taiyuan, Shanxi, People’s Republic of China
| | - Xiaoya Yu
- Department of Pediatrics, Shanxi Medical University, Taiyuan, Shanxi, People’s Republic of China
| | - Jianing Xie
- Department of Pediatrics, Shanxi Medical University, Taiyuan, Shanxi, People’s Republic of China
| | - Guoquan Fan
- School of Basic Medical Science, Shanxi Medical University, Taiyuan, Shanxi, People’s Republic of China
| |
Collapse
|
71
|
Saker D, Sencar L, Coskun G, Sapmaz Ercakalli T, Yilmaz DM, Polat S. Galantamine and wedelolactone combined treatment suppresses LPS-induced NLRP3 inflammasome activation in microglial cells. Immunopharmacol Immunotoxicol 2024; 46:805-814. [PMID: 39279139 DOI: 10.1080/08923973.2024.2405579] [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: 11/07/2023] [Accepted: 08/09/2024] [Indexed: 09/18/2024]
Abstract
CONTEXT Inflammasome NLR family pyrin domain-containing 3 (NLRP3) is associated with neurological disorders. Neuroinflammation can be suppressed by inhibiting NLRP3 inflammasome activation, decreasing neurodegenerative disorder progression. We devised a therapeutic technique that can reduce neuroinflammation induced by microglial activation, avoiding neurodegeneration. We aimed to investigate the mechanisms underlying the pharmacological effects of galantamine and wedelolactone by evaluating the response of the nuclear factor kappa B (NF-κB) signaling pathway and NLRP3 inflammasome in lipopolysaccharide (LPS)-activated N9 microglia. METHODS LPS and adenosine triphosphate were used to activate the NLRP3 inflammasome in N9 microglial cells, which were pretreated with galantamine and wedelolactone. Caspase-1, NLRP3, NF-κB, and interleukin (IL)-1β levels were measured using RT-qPCR and immunostaining. RESULTS Combined administration of galantamine and wedelolactone rescued microglial cells from LPS-induced cell death. Furthermore, treatment with galantamine and wedelolactone led to the suppression of NF-κB expression. NLRP3, caspase-1, and IL-1β levels were decreased by the combined treatment. DISCUSSION AND CONCLUSION The concurrent administration of galantamine and wedelolactone effectively suppresses the production of inflammatory cytokines and NLRP3 inflammasome activation in microglia. This inhibitory effect is likely linked to the NF-κB signaling pathway modulation. Therefore, this combined treatment is a potential therapeutic approach for neuroinflammatory diseases.
Collapse
Affiliation(s)
- Dilek Saker
- Department of Histology and Embryology, Faculty of Medicine, Çukurova University, Adana, Turkey
| | - Leman Sencar
- Department of Histology and Embryology, Faculty of Medicine, Çukurova University, Adana, Turkey
| | - Gulfidan Coskun
- Department of Histology and Embryology, Faculty of Medicine, Çukurova University, Adana, Turkey
| | - Tugce Sapmaz Ercakalli
- Department of Histology and Embryology, Faculty of Medicine, Çukurova University, Adana, Turkey
| | | | - Sait Polat
- Department of Histology and Embryology, Faculty of Medicine, Çukurova University, Adana, Turkey
| |
Collapse
|
72
|
Liu J, Liu H, Tang H, Ran L, Wang D, Yang F, Zhang H, Teng X, Chen D. Golgi apparatus regulated pyroptosis through the miR-32-5p/Golga7/NLRP3 axis in chicken splenic lymphocytes exposure to ammonia. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2024; 362:124923. [PMID: 39260552 DOI: 10.1016/j.envpol.2024.124923] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/17/2024] [Revised: 09/03/2024] [Accepted: 09/07/2024] [Indexed: 09/13/2024]
Abstract
Ammonia, a common toxic gas, posed a hazard to both human and chickens. The Golgi apparatus, an essential organelle, helped maintain the internal environment of the organism and supported the protein foundation for the endoplasmic reticulum to be involved in pyroptosis. Thus, the Golgi apparatus has garnered significant attention. The purpose of our research was to explore the mechanisms of Golgin A7 (Golga7) involved in pyroptosis after chicken exposure to ammonia. To reach our goal, we first created an in vitro ammonia model to study the effect of ammonia on chicken splenic lymphocyte pyroptosis. Then, leveraging this model, we established Golga7 and miR-32-5p knockdown and overexpression models to investigate their roles in ammonia-induced pyroptosis. We found the ultrastructural changes in the nucleus, Golgi apparatus, and mitochondria of chicken splenic lymphocytes exposure to ammonia. The damage of mitochondria increased the level of Reactive Oxygen Species (ROS), which caused the down-regulation of miR-32-5p. The miR-32-5p inhibitor increased the expression of Golga7 and pyroptosis-related genes (NOD-like receptor protein 3 (NLRP3), Cysteine aspartase-1 (Caspase-1), Golgin A3 (Golga3), Nuclear Factor-kappa B (NF-κB), and Tumor Necrosis Factor-alpha (TNF-α)), which induced the pyroptosis, but when miR-32-5p mimic/si-Golga7 (Golga7 inhibitor) was utilized, these effects were reduced. Our research demonstrated that miR-32-5p/Golga7 regulated NLRP3 involving in the pyroptosis of chicken splenic cells exposed to ammonia. Our study provided a valuable foundation for the prevention and treatment chickens ammonia poisoning in the livestock production.
Collapse
Affiliation(s)
- Jiahao Liu
- Key Laboratory of Animal Medicine in Sichuan Province, Southwest Minzu University, Chengdu, 610041, China
| | - Haifeng Liu
- Department of Veterinary Surgery, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, 611130, China
| | - Haojinming Tang
- Key Laboratory of Animal Medicine in Sichuan Province, Southwest Minzu University, Chengdu, 610041, China
| | - Longjun Ran
- Key Laboratory of Animal Medicine in Sichuan Province, Southwest Minzu University, Chengdu, 610041, China
| | - Danni Wang
- Key Laboratory of Animal Medicine in Sichuan Province, Southwest Minzu University, Chengdu, 610041, China
| | - Falong Yang
- Key Laboratory of Animal Medicine in Sichuan Province, Southwest Minzu University, Chengdu, 610041, China
| | - Huanrong Zhang
- Key Laboratory of Animal Medicine in Sichuan Province, Southwest Minzu University, Chengdu, 610041, China
| | - Xiaohua Teng
- College of Animal Science and Technology, Northeast Agricultural University, Harbin, 150030, China
| | - Dechun Chen
- Key Laboratory of Animal Medicine in Sichuan Province, Southwest Minzu University, Chengdu, 610041, China.
| |
Collapse
|
73
|
Owen AR, Farias A, Levins AM, Wang Z, Higham SL, Mack M, Tregoning JS, Johansson C. Exposure to bacterial PAMPs before RSV infection exacerbates innate inflammation and disease via IL-1α and TNF-α. Mucosal Immunol 2024; 17:1184-1198. [PMID: 39127259 PMCID: PMC11631774 DOI: 10.1016/j.mucimm.2024.08.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Revised: 07/17/2024] [Accepted: 08/01/2024] [Indexed: 08/12/2024]
Abstract
Respiratory syncytial virus (RSV) can cause severe lower respiratory tract infections. Understanding why some individuals get more serious disease may help with diagnosis and treatment. One possible risk factor underlying severe disease is bacterial exposure before RSV infection. Bacterial exposure has been associated with increased respiratory viral-induced disease severity but the mechanism remains unknown. Respiratory bacterial infections or exposure to their pathogen associated molecular patterns (PAMPs) trigger innate immune inflammation, characterised by neutrophil and inflammatory monocyte recruitment and the production of inflammatory cytokines. We hypothesise that these changes to the lung environment alter the immune response and disease severity during subsequent RSV infection. To test this, we intranasally exposed mice to LPS, LTA or Acinetobacter baumannii (an airway bacterial pathogen) before RSV infection and observed an early induction of disease, measured by weight loss, at days 1-3 after infection. Neutrophils or inflammatory monocytes were not responsible for driving this exacerbated weight loss. Instead, exacerbated disease was associated with increased IL-1α and TNF-α, which orchestrated the recruitment of innate immune cells into the lung. This study shows that exposure to bacterial PAMPs prior to RSV infection increases the expression of IL-1α and TNF-α, which dysregulate the immune response resulting in exacerbated disease.
Collapse
Affiliation(s)
- Amber R Owen
- Respiratory Infections, National Heart and Lung Institute, Imperial College London, United Kingdom
| | - Ana Farias
- Respiratory Infections, National Heart and Lung Institute, Imperial College London, United Kingdom
| | - Anne-Marie Levins
- Respiratory Infections, National Heart and Lung Institute, Imperial College London, United Kingdom
| | - Ziyin Wang
- Department of Infectious Disease, Imperial College London, United Kingdom
| | - Sophie L Higham
- Department of Infectious Disease, Imperial College London, United Kingdom
| | - Matthias Mack
- Department of Nephrology, University Hospital Regensburg, Regensburg, Germany
| | - John S Tregoning
- Department of Infectious Disease, Imperial College London, United Kingdom
| | - Cecilia Johansson
- Respiratory Infections, National Heart and Lung Institute, Imperial College London, United Kingdom.
| |
Collapse
|
74
|
Szabo A, Akkouh I, Osete JR, de Assis DR, Kondratskaya E, Hughes T, Ueland T, Andreassen OA, Djurovic S. NLRP3 inflammasome mediates astroglial dysregulation of innate and adaptive immune responses in schizophrenia. Brain Behav Immun 2024; 124:144-156. [PMID: 39617069 DOI: 10.1016/j.bbi.2024.11.030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/22/2024] [Revised: 11/19/2024] [Accepted: 11/27/2024] [Indexed: 12/07/2024] Open
Abstract
Mounting evidence indicates the involvement of neuroinflammation in the development of schizophrenia (SCZ), but the potential role of astroglia in this phenomenon remains poorly understood. We assessed the molecular and functional consequences of inflammasome activation using induced pluripotent stem cell (iPSC)-derived astrocytes generated from SCZ patients and healthy controls (CTRL). Screening protein levels in astrocytes at baseline identified lower expression of the NLRP3-ASC complex in SCZ, but increased Caspase-1 activity upon specific NLRP3 stimulation compared to CTRL. Using transcriptional profiling, we found corresponding downregulations of NLRP3 and ASC/PYCARD in both iPSC-derived astrocytes, and in a large (n = 429) brain postmortem case-control sample. Functional analyses following NLRP3 activation revealed an inflammatory phenotype characterized by elevated production of IL-1β/IL-18 and skewed priming of helper T lymphocytes (Th1/Th17) by SCZ astrocytes. This phenotype was rescued by specific inhibition of NLRP3 activation, demonstrating its dependence on the NLRP3 inflammasome. Taken together, SCZ iPSC-astrocytes display unique, NLRP3-dependent inflammatory characteristics that are manifested via various cellular functions, as well as via dysregulated innate and adaptive immune responses.
Collapse
Affiliation(s)
- Attila Szabo
- Centre for Precision Psychiatry, Institute of Clinical Medicine, University of Oslo, Oslo, Norway; Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway; Department of Medical Genetics, Oslo University Hospital, Oslo, Norway; K.G. Jebsen Centre for Neurodevelopmental Disorders, University of Oslo, Oslo, Norway.
| | - Ibrahim Akkouh
- Centre for Precision Psychiatry, Institute of Clinical Medicine, University of Oslo, Oslo, Norway; Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway; Department of Medical Genetics, Oslo University Hospital, Oslo, Norway
| | - Jordi Requena Osete
- Centre for Precision Psychiatry, Institute of Clinical Medicine, University of Oslo, Oslo, Norway; Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway; Department of Medical Genetics, Oslo University Hospital, Oslo, Norway
| | - Denis Reis de Assis
- Centre for Precision Psychiatry, Institute of Clinical Medicine, University of Oslo, Oslo, Norway; Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway; Department of Medical Genetics, Oslo University Hospital, Oslo, Norway
| | - Elena Kondratskaya
- Centre for Precision Psychiatry, Institute of Clinical Medicine, University of Oslo, Oslo, Norway; Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway; Department of Medical Genetics, Oslo University Hospital, Oslo, Norway
| | - Timothy Hughes
- Centre for Precision Psychiatry, Institute of Clinical Medicine, University of Oslo, Oslo, Norway; Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway; Department of Medical Genetics, Oslo University Hospital, Oslo, Norway
| | - Thor Ueland
- Research Institute of Internal Medicine, Oslo University Hospital, Rikshospitalet, Oslo, Norway; Faculty of Medicine, University of Oslo, Norway; K.G. Jebsen Thrombosis Research and Expertise Centre, University of Tromsø, Tromsø, Norway
| | - Ole A Andreassen
- Centre for Precision Psychiatry, Institute of Clinical Medicine, University of Oslo, Oslo, Norway; Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway
| | - Srdjan Djurovic
- Department of Medical Genetics, Oslo University Hospital, Oslo, Norway; K.G. Jebsen Centre for Neurodevelopmental Disorders, University of Oslo, Oslo, Norway; Department of Clinical Science, University of Bergen, Bergen, Norway
| |
Collapse
|
75
|
Hao T, Zhang X, Liu Q, Zhan R, Tang Y, Bu X, Li W, Du J, Li Y, Mai K, Ai Q. Phosphatidylethanolamine exerts anti-inflammatory action by regulating mitochondrial function in macrophages of large yellow croaker (Larimichthys crocea). FASEB J 2024; 38:e70180. [PMID: 39570029 DOI: 10.1096/fj.202401279rr] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Revised: 10/13/2024] [Accepted: 11/04/2024] [Indexed: 11/22/2024]
Abstract
Phosphatidylethanolamine (PE) is a ubiquitous bioactive lipid in cells, which participates in regulating many metabolic processes. Exogenous PE has been reported to play a positive regulatory role in macrophage inflammatory responses. However, the molecular mechanisms of PE in regulating macrophage inflammation are not completely understood. In the present study, transcriptomic analysis of PE-stimulated macrophages of large yellow croaker revealed that differentially expressed genes were mainly active in cellular components of the mitochondrial respiratory chain, which corresponded to the significant enrichment of the oxidative phosphorylation pathway. Consistent with this result, PE significantly increased ATP content and protein expression of NDUFB3 (mitochondrial respiratory chain complex I subunit) in macrophages. Meanwhile, transcriptomic data showed that PE treatment downregulated the transcript levels of nlrp3 and upregulated the transcript levels of suppressor of cytokine signaling 3 (socs3), suggesting that PE may alleviate macrophage inflammation by interfering with the activation of NLRP3 inflammasome. Further analysis showed that PE significantly attenuated dietary PA-mediated macrophage inflammation via NLRP3-Caspase-1 in vitro and in vivo. Given that PE abundance is strongly correlated with mitochondrial function, the present study hypothesized that PE-mediated inflammatory modulation may be attributed to the positive effects on mitochondrial function. As expected, PE significantly ameliorated PA-induced mitochondrial dysfunction and reduced intracellular reactive oxygen species production and malondialdehyde content in macrophages, indicating that the improvement of mitochondrial function is an important mechanism involved in the positive effect of PE on PA-induced inflammation. In conclusion, this study elucidates the critical role of mitochondrial function in PE-mediated regulation of inflammation in macrophages, which expands the understanding of the regulatory mechanisms of phospholipid metabolism on dietary fatty acid-induced inflammation. This study may provide new intervention targets and nutritional regulation strategies for improving chronic inflammatory diseases.
Collapse
Affiliation(s)
- Tingting Hao
- Key Laboratory of Aquaculture Nutrition and Feed (Ministry of Agriculture and Rural Affairs) and Key Laboratory of Mariculture (Ministry of Education), Ocean University of China, Qingdao, Shandong, People's Republic of China
| | - Xinwen Zhang
- Key Laboratory of Aquaculture Nutrition and Feed (Ministry of Agriculture and Rural Affairs) and Key Laboratory of Mariculture (Ministry of Education), Ocean University of China, Qingdao, Shandong, People's Republic of China
| | - Qiangde Liu
- Key Laboratory of Aquaculture Nutrition and Feed (Ministry of Agriculture and Rural Affairs) and Key Laboratory of Mariculture (Ministry of Education), Ocean University of China, Qingdao, Shandong, People's Republic of China
| | - Rui Zhan
- Key Laboratory of Aquaculture Nutrition and Feed (Ministry of Agriculture and Rural Affairs) and Key Laboratory of Mariculture (Ministry of Education), Ocean University of China, Qingdao, Shandong, People's Republic of China
| | - Yuhang Tang
- Key Laboratory of Aquaculture Nutrition and Feed (Ministry of Agriculture and Rural Affairs) and Key Laboratory of Mariculture (Ministry of Education), Ocean University of China, Qingdao, Shandong, People's Republic of China
| | - Xianyong Bu
- Key Laboratory of Aquaculture Nutrition and Feed (Ministry of Agriculture and Rural Affairs) and Key Laboratory of Mariculture (Ministry of Education), Ocean University of China, Qingdao, Shandong, People's Republic of China
| | - Weijia Li
- Key Laboratory of Aquaculture Nutrition and Feed (Ministry of Agriculture and Rural Affairs) and Key Laboratory of Mariculture (Ministry of Education), Ocean University of China, Qingdao, Shandong, People's Republic of China
| | - Jianlong Du
- Key Laboratory of Aquaculture Nutrition and Feed (Ministry of Agriculture and Rural Affairs) and Key Laboratory of Mariculture (Ministry of Education), Ocean University of China, Qingdao, Shandong, People's Republic of China
| | - Yueru Li
- Key Laboratory of Aquaculture Nutrition and Feed (Ministry of Agriculture and Rural Affairs) and Key Laboratory of Mariculture (Ministry of Education), Ocean University of China, Qingdao, Shandong, People's Republic of China
| | - Kangsen Mai
- Key Laboratory of Aquaculture Nutrition and Feed (Ministry of Agriculture and Rural Affairs) and Key Laboratory of Mariculture (Ministry of Education), Ocean University of China, Qingdao, Shandong, People's Republic of China
- Laboratory for Marine Fisheries Science and Food Production Processes, Qingdao National Laboratory for Marine Science and Technology, Qingdao, Shandong, People's Republic of China
| | - Qinghui Ai
- Key Laboratory of Aquaculture Nutrition and Feed (Ministry of Agriculture and Rural Affairs) and Key Laboratory of Mariculture (Ministry of Education), Ocean University of China, Qingdao, Shandong, People's Republic of China
- Laboratory for Marine Fisheries Science and Food Production Processes, Qingdao National Laboratory for Marine Science and Technology, Qingdao, Shandong, People's Republic of China
| |
Collapse
|
76
|
Pei X, Ma S, Hong L, Zuo Z, Xu G, Chen C, Shen Y, Liu D, Li C, Li D. Molecular insights of T-2 toxin exposure-induced neurotoxicity and the neuroprotective effect of dimethyl fumarate. Food Chem Toxicol 2024; 196:115166. [PMID: 39617286 DOI: 10.1016/j.fct.2024.115166] [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: 10/27/2024] [Revised: 11/24/2024] [Accepted: 11/28/2024] [Indexed: 12/06/2024]
Abstract
T-2 toxin, a potent environmental pollutant, has been proved to stimulate neuroinflammation, while the connection between T-2 toxin and pyroptosis remain elusive. Dimethyl fumarate (DMF), recently identified as a neuroprotectant and pyroptosis inhibitor, has potential therapeutic applications that are underexplored. Based on present study in vitro and vivo, we demonstrated that T-2 toxin induced the activation of NLRP3-Caspase-1 inflammasome in hippocampal neurons. In addition to proinflammatory mediator overexpression, gasdermin D (GSDMD)-dependently pyroptosis in the mouse hippocampal neuron cell line (HT22) treated by T-2 toxin was determined in our study. Moreover, the palliative effect of knockdown sequence of high mobility group B1 protein (HMGB1) provided more details for T-2 toxin-initiated pyroptosis. Importantly, we confirmed that DMF, as a novel inhibitor of GSDMD, could alleviate pyroptosis induced by T-2 toxin in an GSDMD targeting manner. In summary, our studies exposed the evidence that T-2 toxin could induce NLRP3 inflammasome activation and hippocampal neuronal pyroptosis. More notably, DMF was turn out to be a critical executioner for attenuating GSDMD-mediated pyroptosis. Our data found a new function of DMF and suggested a novel therapy strategy against mycotoxin-triggered neuronal inflammation, which leads to varieties of neurological diseases.
Collapse
Affiliation(s)
- Xingyao Pei
- Open Fund of Key Laboratory of Smart Breeding (Co-construction By Ministry and Province), Ministry of Agriculture and Rural Affairs, Tianjin Agricultural University, Jinjing Road No.22, Xiqing District, Tianjin 300392, China; Tianjin Key Laboratory of Agricultural Animal Breeding and Healthy Husbandry, College of Animal Science and Veterinary Medicine, Tianjin Agricultural University, Jinjing Road No.22, Xiqing District, Tianjin 300392, China
| | - Shuhui Ma
- Tianjin Key Laboratory of Agricultural Animal Breeding and Healthy Husbandry, College of Animal Science and Veterinary Medicine, Tianjin Agricultural University, Jinjing Road No.22, Xiqing District, Tianjin 300392, China
| | - Liang Hong
- Tianjin Key Laboratory of Agricultural Animal Breeding and Healthy Husbandry, College of Animal Science and Veterinary Medicine, Tianjin Agricultural University, Jinjing Road No.22, Xiqing District, Tianjin 300392, China
| | - Zonghui Zuo
- Tianjin Key Laboratory of Agricultural Animal Breeding and Healthy Husbandry, College of Animal Science and Veterinary Medicine, Tianjin Agricultural University, Jinjing Road No.22, Xiqing District, Tianjin 300392, China
| | - Gang Xu
- Tianjin Key Laboratory of Agricultural Animal Breeding and Healthy Husbandry, College of Animal Science and Veterinary Medicine, Tianjin Agricultural University, Jinjing Road No.22, Xiqing District, Tianjin 300392, China
| | - Chun Chen
- Tianjin Key Laboratory of Agricultural Animal Breeding and Healthy Husbandry, College of Animal Science and Veterinary Medicine, Tianjin Agricultural University, Jinjing Road No.22, Xiqing District, Tianjin 300392, China
| | - Yao Shen
- Tianjin Key Laboratory of Agricultural Animal Breeding and Healthy Husbandry, College of Animal Science and Veterinary Medicine, Tianjin Agricultural University, Jinjing Road No.22, Xiqing District, Tianjin 300392, China
| | - Dingkuo Liu
- Tianjin Key Laboratory of Agricultural Animal Breeding and Healthy Husbandry, College of Animal Science and Veterinary Medicine, Tianjin Agricultural University, Jinjing Road No.22, Xiqing District, Tianjin 300392, China; Tianjin Key Laboratory of Biological Feed Additive Enterprise, S&E Burgeoning Biotechnology (Tianjin) Co., Ltd, Tianjin 300383, China
| | - Cun Li
- Tianjin Key Laboratory of Agricultural Animal Breeding and Healthy Husbandry, College of Animal Science and Veterinary Medicine, Tianjin Agricultural University, Jinjing Road No.22, Xiqing District, Tianjin 300392, China
| | - Daowen Li
- Open Fund of Key Laboratory of Smart Breeding (Co-construction By Ministry and Province), Ministry of Agriculture and Rural Affairs, Tianjin Agricultural University, Jinjing Road No.22, Xiqing District, Tianjin 300392, China; Tianjin Key Laboratory of Agricultural Animal Breeding and Healthy Husbandry, College of Animal Science and Veterinary Medicine, Tianjin Agricultural University, Jinjing Road No.22, Xiqing District, Tianjin 300392, China; Tianjin Key Laboratory of Biological Feed Additive Enterprise, S&E Burgeoning Biotechnology (Tianjin) Co., Ltd, Tianjin 300383, China.
| |
Collapse
|
77
|
He R, Liu Y, Fu W, He X, Liu S, Xiao D, Tao Y. Mechanisms and cross-talk of regulated cell death and their epigenetic modifications in tumor progression. Mol Cancer 2024; 23:267. [PMID: 39614268 PMCID: PMC11606237 DOI: 10.1186/s12943-024-02172-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Accepted: 11/07/2024] [Indexed: 12/01/2024] Open
Abstract
Cell death is a fundamental part of life for metazoans. To maintain the balance between cell proliferation and metabolism of human bodies, a certain number of cells need to be removed regularly. Hence, the mechanisms of cell death have been preserved during the evolution of multicellular organisms. Tumorigenesis is closely related with exceptional inhibition of cell death. Mutations or defects in cell death-related genes block the elimination of abnormal cells and enhance the resistance of malignant cells to chemotherapy. Therefore, the investigation of cell death mechanisms enables the development of drugs that directly induce tumor cell death. In the guidelines updated by the Cell Death Nomenclature Committee (NCCD) in 2018, cell death was classified into 12 types according to morphological, biochemical and functional classification, including intrinsic apoptosis, extrinsic apoptosis, mitochondrial permeability transition (MPT)-driven necrosis, necroptosis, ferroptosis, pyroptosis, PARP-1 parthanatos, entotic cell death, NETotic cell death, lysosome-dependent cell death, autophagy-dependent cell death, immunogenic cell death, cellular senescence and mitotic catastrophe. The mechanistic relationships between epigenetic controls and cell death in cancer progression were previously unclear. In this review, we will summarize the mechanisms of cell death pathways and corresponding epigenetic regulations. Also, we will explore the extensive interactions between these pathways and discuss the mechanisms of cell death in epigenetics which bring benefits to tumor therapy.
Collapse
Affiliation(s)
- Ruimin He
- Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, 410078, China
- Cancer Research Institute and School of Basic Medicine, Central South University, Changsha, Hunan, 410078, China
- Department of Pathology, Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Xiangya Hospital, Central South University, Hunan, 410078, China
| | - Yifan Liu
- Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, 410078, China
- Cancer Research Institute and School of Basic Medicine, Central South University, Changsha, Hunan, 410078, China
- Department of Pathology, Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Xiangya Hospital, Central South University, Hunan, 410078, China
| | - Weijie Fu
- Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, 410078, China
- Cancer Research Institute and School of Basic Medicine, Central South University, Changsha, Hunan, 410078, China
- Department of Pathology, Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Xiangya Hospital, Central South University, Hunan, 410078, China
| | - Xuan He
- Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, 410078, China
- Cancer Research Institute and School of Basic Medicine, Central South University, Changsha, Hunan, 410078, China
- Department of Pathology, Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Xiangya Hospital, Central South University, Hunan, 410078, China
| | - Shuang Liu
- Department of Oncology, Institute of Medical Sciences, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China.
| | - Desheng Xiao
- Department of Pathology, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China.
| | - Yongguang Tao
- Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, 410078, China.
- Cancer Research Institute and School of Basic Medicine, Central South University, Changsha, Hunan, 410078, China.
- Department of Pathology, Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Xiangya Hospital, Central South University, Hunan, 410078, China.
- Department of Pathology, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China.
- Department of Thoracic Surgery, Hunan Key Laboratory of Early Diagnosis and Precision Therapy in Lung Cancer, Second Xiangya Hospital, Central South University, Changsha, 410011, China.
- Furong Laboratory, Xiangya School of Medicine, Central South University, Hunan, 410078, China.
| |
Collapse
|
78
|
Filannino FM, Ruggiero M, Panaro MA, Lofrumento DD, Trotta T, Benameur T, Cianciulli A, Calvello R, Zoila F, Porro C. Irisin Attenuates Neuroinflammation Targeting the NLRP3 Inflammasome. Molecules 2024; 29:5623. [PMID: 39683782 DOI: 10.3390/molecules29235623] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Revised: 11/25/2024] [Accepted: 11/26/2024] [Indexed: 12/18/2024] Open
Abstract
Neuroinflammation is defined as an immune response involving various cell types, particularly microglia, which monitor the neuroimmune axis. Microglia activate in two distinct ways: M1, which is pro-inflammatory and capable of inducing phagocytosis and releasing pro-inflammatory factors, and M2, which has anti-inflammatory properties. Inflammasomes are large protein complexes that form in response to internal danger signals, activating caspase-1 and leading to the release of pro-inflammatory cytokines such as interleukin 1β. Irisin, a peptide primarily released by muscles during exercise, was examined for its effects on BV2 microglial cells in vitro. Even at low concentrations, irisin was observed to influence the NLRP3 inflammasome, showing potential as a neuroprotective and anti-inflammatory agent after stimulation with lipopolysaccharides (LPSs). Irisin helped maintain microglia in their typical physiological state and reduced their migratory capacity. Irisin also increased Arg-1 protein expression, a marker of M2 polarization, while downregulating NLRP3, Pycard, caspase-1, IL-1β, and CD14. The results of this study indicate that irisin may serve as a crucial mediator of neuroprotection, thus representing an innovative tool for the prevention of neurodegenerative diseases.
Collapse
Affiliation(s)
| | - Melania Ruggiero
- Department of Biosciences, Biotechnologies and Environment, University of Bari, I-70125 Bari, Italy
| | - Maria Antonietta Panaro
- Department of Biosciences, Biotechnologies and Environment, University of Bari, I-70125 Bari, Italy
| | - Dario Domenico Lofrumento
- Department of Biological and Environmental Sciences and Technologies, Section of Human Anatomy, University of Salento, I-73100 Lecce, Italy
| | - Teresa Trotta
- Department of Clinical and Experimental Medicine, University of Foggia, I-71100 Foggia, Italy
| | - Tarek Benameur
- Department of Biomedical Sciences, College of Medicine, King Faisal University, Al-Ahsa 31982, Saudi Arabia
| | - Antonia Cianciulli
- Department of Biosciences, Biotechnologies and Environment, University of Bari, I-70125 Bari, Italy
| | - Rosa Calvello
- Department of Biosciences, Biotechnologies and Environment, University of Bari, I-70125 Bari, Italy
| | - Federico Zoila
- Department of Clinical and Experimental Medicine, University of Foggia, I-71100 Foggia, Italy
| | - Chiara Porro
- Department of Clinical and Experimental Medicine, University of Foggia, I-71100 Foggia, Italy
| |
Collapse
|
79
|
Zhou X, Tao Y, Shi Y. Unraveling the NLRP family: Structure, function, activation, critical influence on tumor progression, and potential as targets for cancer therapy. Cancer Lett 2024; 605:217283. [PMID: 39366544 DOI: 10.1016/j.canlet.2024.217283] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Revised: 09/27/2024] [Accepted: 09/30/2024] [Indexed: 10/06/2024]
Abstract
The innate immune system serves as the body's initial defense, swiftly detecting danger via pattern recognition receptors (PRRs). Among these, nucleotide-binding oligomerization domain-like receptor family pyrin domain-containing proteins (NLRPs) are pivotal in recognizing pathogen-associated and damage-associated molecular patterns, thereby triggering immune responses. NLRPs, the most extensively studied subset within the NLR family, form inflammasomes that regulate inflammation, essential for innate immunity activation. Recent research highlights NLRPs' significant impact on various human diseases, including cancer. With differential expression across organs, NLRPs influence cancer progression by modulating immune reactions, cell fate, and proliferation. Their clinical significance in cancer makes them promising therapeutic targets. This review provides a comprehensive overview of the structure, function, activation mechanism of the NLRPs family and its potential role in cancer progression. In addition, we particularly focused on the concept of NLRP as a therapeutic target and its potential value in combination with immune checkpoint inhibitors.
Collapse
Affiliation(s)
- Xueqing Zhou
- Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, 410013, China; Cancer Research Institute and School of Basic Medicine, Central South University, Changsha, Hunan, 410013, China
| | - Yongguang Tao
- Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, 410013, China; Cancer Research Institute and School of Basic Medicine, Central South University, Changsha, Hunan, 410013, China.
| | - Ying Shi
- Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, 410013, China; Cancer Research Institute and School of Basic Medicine, Central South University, Changsha, Hunan, 410013, China.
| |
Collapse
|
80
|
Pan BY, Chen CS, Chen FY, Shen MY. Multifaceted Role of Apolipoprotein C3 in Cardiovascular Disease Risk and Metabolic Disorder in Diabetes. Int J Mol Sci 2024; 25:12759. [PMID: 39684468 DOI: 10.3390/ijms252312759] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2024] [Revised: 11/26/2024] [Accepted: 11/26/2024] [Indexed: 12/18/2024] Open
Abstract
Apolipoprotein C3 (APOC3) plays a critical role in regulating triglyceride levels and serves as a key predictor of cardiovascular disease (CVD) risk, particularly in patients with diabetes. While APOC3 is known to inhibit lipoprotein lipase, recent findings reveal its broader influence across lipoprotein metabolism, where it modulates the structure and function of various lipoproteins. Therefore, this review examines the complex metabolic cycle of APOC3, emphasizing the impact of APOC3-containing lipoproteins on human metabolism, particularly in patients with diabetes. Notably, APOC3 affects triglyceride-rich lipoproteins and causes structural changes in high-, very low-, intermediate-, and low-density lipoproteins, thereby increasing CVD risk. Evidence suggests that elevated APOC3 levels-above the proposed safe range of 10-15 mg/dL-correlate with clinically significant CVD outcomes. Recognizing APOC3 as a promising biomarker for CVD, this review underscores the urgent need for high-throughput, clinically feasible methods to further investigate its role in lipoprotein physiology in both animal models and human studies. Additionally, we analyze the relationship between APOC3-related genes and lipoproteins, reinforcing the value of large-population studies to understand the impact of APOC3 on metabolic diseases. Ultimately, this review supports the development of therapeutic strategies targeting APOC3 reduction as a preventive approach for diabetes-related CVD.
Collapse
Affiliation(s)
- Bo-Yi Pan
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung 40402, Taiwan
| | - Chen-Sheng Chen
- The Ph.D. Program for Cancer Biology and Drug Discovery, China Medical University and Academia Sinica, Taichung 40402, Taiwan
| | - Fang-Yu Chen
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung 40402, Taiwan
| | - Ming-Yi Shen
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung 40402, Taiwan
- Department of Medical Research, China Medical University Hospital, Taichung 40402, Taiwan
- Department of Nursing, Asia University, Taichung 413305, Taiwan
| |
Collapse
|
81
|
Theodorakis N, Kreouzi M, Hitas C, Anagnostou D, Nikolaou M. Adipokines and Cardiometabolic Heart Failure with Preserved Ejection Fraction: A State-of-the-Art Review. Diagnostics (Basel) 2024; 14:2677. [PMID: 39682585 DOI: 10.3390/diagnostics14232677] [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: 11/06/2024] [Revised: 11/25/2024] [Accepted: 11/26/2024] [Indexed: 12/18/2024] Open
Abstract
BACKGROUND Cardiometabolic heart failure with preserved ejection fraction (HFpEF) is largely driven by obesity-related factors, including adipokines and bioactive peptides primarily secreted by the adipose tissue, such as leptin, adiponectin, and resistin. These molecules link metabolic dysregulation to cardiovascular dysfunction, influencing HFpEF progression and patient outcomes Methods: A comprehensive literature search was conducted in PubMed up to 20 November 2024, using keywords and MeSH terms, such as "HFpEF", "adipokines", "leptin", "adiponectin", and "resistin", yielding 723 results. Boolean operators refined the search, and reference lists of key studies were reviewed. After screening for duplicates and irrelevant studies, 103 articles were included, providing data on adipokines' roles in HFpEF pathophysiology, biomarkers, and therapeutic implications. RESULTS Both preclinical and clinical studies have demonstrated that adipokines play a role in modulating cardiovascular function, thereby contributing to the development of cardiometabolic HFpEF. Leptin promotes myocardial hypertrophy, fibrosis, endothelial dysfunction, and inflammation, though contradictory evidence suggests potential cardioprotective roles in subgroups like obese African American women. Adiponectin generally offers protective effects but presents a paradox, where elevated levels may correlate with worse outcomes, which may reflect either a compensatory response to cardiac dysfunction or a maladaptive state characterized by adiponectin resistance. Resistin is associated with increased cardiovascular risk through pro-inflammatory and pro-fibrotic effects, though its role in HFpEF requires further clarification. Other adipokines, like retinol-binding protein 4 and omentin-1, have emerged as potential contributors. Despite growing insights, clinical translation remains limited, underscoring a significant gap between experimental evidence and therapeutic application. CONCLUSIONS Future research should focus on targeted interventions that modulate adipokine pathways to potentially improve HFpEF outcomes. Innovative treatment strategies addressing underlying metabolic disturbances and adipokine dysregulation are essential for advancing the management of this challenging condition.
Collapse
Affiliation(s)
- Nikolaos Theodorakis
- School of Medicine, National and Kapodistrian University of Athens, 75 Mikras Asias, 11527 Athens, Greece
- Department of Cardiology & Heart Failure Outpatient Clinic, Sismanogleio-Amalia Fleming General Hospital, 14 25is Martiou Str., 15127 Melissia, Greece
| | - Magdalini Kreouzi
- Department of Internal Medicine, Sismanogleio-Amalia Fleming General Hospital, 14 25is Martiou Str., 15127 Melissia, Greece
| | - Christos Hitas
- Department of Cardiology & Heart Failure Outpatient Clinic, Sismanogleio-Amalia Fleming General Hospital, 14 25is Martiou Str., 15127 Melissia, Greece
| | - Dimitrios Anagnostou
- Department of Cardiology & Heart Failure Outpatient Clinic, Sismanogleio-Amalia Fleming General Hospital, 14 25is Martiou Str., 15127 Melissia, Greece
| | - Maria Nikolaou
- Department of Cardiology & Heart Failure Outpatient Clinic, Sismanogleio-Amalia Fleming General Hospital, 14 25is Martiou Str., 15127 Melissia, Greece
| |
Collapse
|
82
|
Drago E, Fioredda F, Penco F, Prigione I, Bertoni A, Del Zotto G, Bocca P, Massaccesi E, Lanciotti M, Moratto D, Thurner L, Caorsi R, Gattorno M, Volpi S. Inborn Error of WAS Presenting with SARS-CoV-2-Related Multisystem Inflammatory Syndrome in Children. J Clin Immunol 2024; 45:49. [PMID: 39581942 DOI: 10.1007/s10875-024-01840-4] [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: 12/03/2023] [Accepted: 11/08/2024] [Indexed: 11/26/2024]
Abstract
Multisystem inflammatory syndrome in children (MIS-C) has been reported in patients with inborn errors of immunity (IEI), providing insights into disease pathogenesis. Here, we present the first case of MIS-C in a child affected by Wiskott-Aldrich syndrome (WAS) gene mutation, elucidating underlying predisposing factors and the involved inflammatory pathways. Genetic analysis revealed a frameshift truncating variant in the WAS gene, resulting in WAS protein expression between mild and severe forms, despite a clinical phenotype resembling X-linked thrombocytopenia (XLT). IL-1β secretion by LPS-stimulated peripheral blood mononuclear cells from patient during MIS-C was lower compared to healthy subjects but increased during follow-up. Conversely, the percentage of ASC (apoptosis-associated speck-like protein containing a CARD) specks in the patient's circulating monocytes during the acute phase was higher than in healthy subjects. The type I interferon (IFN) signature during MIS-C was normal, in contrast to the raised IFN signature measured far from the acute event. This case supports the association of IEI with MIS-C, potentially linked to delayed immune responses to SARS-CoV-2. The XLT phenotype underlies a subclinical immunodysregulation involving the NLRP3 inflammasome and the type-I IFN response.
Collapse
Affiliation(s)
- Enrico Drago
- Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health (DINOGMI), Università degli Studi di Genova, Genova, Italy.
- UOC Reumatologia e Malattie Autoinfiammatorie, IRCCS Istituto Giannina Gaslini, Genova, Italy.
| | | | - Federica Penco
- UOC Reumatologia e Malattie Autoinfiammatorie, IRCCS Istituto Giannina Gaslini, Genova, Italy
| | - Ignazia Prigione
- UOC Reumatologia e Malattie Autoinfiammatorie, IRCCS Istituto Giannina Gaslini, Genova, Italy
| | - Arinna Bertoni
- UOC Reumatologia e Malattie Autoinfiammatorie, IRCCS Istituto Giannina Gaslini, Genova, Italy
| | - Genny Del Zotto
- Core Facilities Laboratory, Integrated Department of Services and Laboratories, IRCCS Istituto Giannina Gaslini, Genova, Italy
| | - Paola Bocca
- UOC Reumatologia e Malattie Autoinfiammatorie, IRCCS Istituto Giannina Gaslini, Genova, Italy
| | | | | | - Daniele Moratto
- Flow Cytometry Unit, Clinical Chemistry Laboratory, ASST Spedali Civili di Brescia, Brescia, Italy
| | - Lorenz Thurner
- José Carreras Center for Immuno and Gene Therapy, Department of Internal Medicine I, Saarland University, Homburg, Germany
| | - Roberta Caorsi
- UOC Reumatologia e Malattie Autoinfiammatorie, IRCCS Istituto Giannina Gaslini, Genova, Italy
| | - Marco Gattorno
- UOC Reumatologia e Malattie Autoinfiammatorie, IRCCS Istituto Giannina Gaslini, Genova, Italy
| | - Stefano Volpi
- Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health (DINOGMI), Università degli Studi di Genova, Genova, Italy
- UOC Reumatologia e Malattie Autoinfiammatorie, IRCCS Istituto Giannina Gaslini, Genova, Italy
| |
Collapse
|
83
|
Chen Y, He X, Chen Y, Zhang R, Zhang T, Zhang T, Wu L. IL-33 deficiency inhibits Toxoplasma gondii infection by promoting NLRP3 inflammasome. Parasitol Res 2024; 123:391. [PMID: 39570453 DOI: 10.1007/s00436-024-08414-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2024] [Accepted: 11/12/2024] [Indexed: 11/22/2024]
Abstract
NLRP3 inflammasome-mediated inflammatory responses play pivotal functions in innate immunity. However, its homeostatic regulation still needs to be better understood. Here we explore the effect and potential mechanism of IL-33 on NLRP3 inflammasome upon Toxoplasma gondii infection through a series of molecular biology and immunological experiments, including western blot, qRT-PCR, and ELISA. We demonstrated that T. gondii infection induces the expression of IL-33, and IL-33-deficient (IL-33-/-) mice exhibit longer survival time than wild-type (WT) mice upon T. gondii infection. IL-33 deficiency promotes the expression of NLRP3 and ASC and the secretion of IL-1β, while exogenous IL-33 inhibits NLRP3 inflammasome. Furthermore, T. gondii infection results in the M2 polarization of macrophages, exacerbated by exogenous IL-33, which also promotes the proliferation of T. gondii. These findings showed that IL-33 deficiency attenuates T. gondii infection by promoting NLRP3 inflammasome, advancing the understanding of the role of IL-33 in inflammation.
Collapse
Affiliation(s)
- Yizhong Chen
- The Department of Immunology, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, China
| | - Xiaoli He
- The Department of Immunology, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, China
| | - Yuqin Chen
- The Department of Immunology, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, China
| | - Rongzhao Zhang
- The Department of Immunology, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, China
| | - Tengwen Zhang
- The Department of Immunology, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, China
| | - Tao Zhang
- The Department of Immunology, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, China.
| | - Linqing Wu
- The Department of Immunology, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, China.
| |
Collapse
|
84
|
Vallese A, Cordone V, Ferrara F, Guiotto A, Gemmo L, Cervellati F, Hayek J, Pecorelli A, Valacchi G. NLRP3 inflammasome-mitochondrion loop in autism spectrum disorder. Free Radic Biol Med 2024; 225:581-594. [PMID: 39433111 DOI: 10.1016/j.freeradbiomed.2024.10.297] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/16/2024] [Revised: 10/17/2024] [Accepted: 10/18/2024] [Indexed: 10/23/2024]
Abstract
Autism spectrum disorder (ASD) is a neurodevelopmental disorder characterized by deficits in social communication and the presence of restricted interests and repetitive behavior. To date, no single cause has been demonstrated but both genetic and environmental factors are believed to be involved in abnormal brain development. In recent years, immunological and mitochondrial dysfunctions acquired particular interest in the study of the molecular mechanisms underlying the pathophysiology of ASD. For this reason, our study focused on evaluating the mitochondrial component and activation of the NLRP3 inflammasome, a critical player of the innate immune system. The assembly of NLRP3 with ASC mediates activation of Caspase-1, which in turn, by proteolytic cleavage, activates Gasdermin D and the proinflammatory cytokines IL-1β/IL-18 with their subsequent secretion. Using primary fibroblasts of autistic and control patients we studied basal and stimulated conditions. Specifically, LPS and ATP were used to activate the NLRP3 inflammasome and MCC950 for its inhibition. In addition, FCCP was used as a mitochondrial stressor and MitoTEMPO as a scavenger of mitochondrial ROS. Our results showed a hyperactivation of NLRP3 inflammasome in ASDs, as evidenced by the co-localization of the two main components, NLRP3 and ASC, by the higher levels of ASC specks, oligomers and dimers and by the increased amounts of active Caspase-1 and IL-1β. In addition, increased mitochondrial superoxide anion and reduced mitochondrial membrane potential were detected in ASD cells. These data are in accordance with the abnormal mitochondrial morphology evidenced by transmission electron microscopy analysis. Interestingly, NLRP3 inflammasome inhibition with MCC950 improved mitochondrial parameters, while the use of MitoTEMPO, in addition to decrease mitochondrial ROS production, was able to prevent NLRP3 inflammasome activation suggesting for the first time an abnormal bidirectional crosstalk between mitochondria and NLRP3 inflammasome in ASD.
Collapse
Affiliation(s)
- Andrea Vallese
- Dept. of Environmental and Prevention Sciences, University of Ferrara, Ferrara, Italy; Animal Science Dept., Plants for Human Health Institute, North Carolina State University, Kannapolis, NC, USA
| | - Valeria Cordone
- Dept. of Environmental and Prevention Sciences, University of Ferrara, Ferrara, Italy
| | - Francesca Ferrara
- Dept. of Chemical, Pharmaceutical and Agricultural Sciences, University of Ferrara, Ferrara, Italy
| | - Anna Guiotto
- Dept. of Environmental and Prevention Sciences, University of Ferrara, Ferrara, Italy; Animal Science Dept., Plants for Human Health Institute, North Carolina State University, Kannapolis, NC, USA
| | - Laura Gemmo
- Dept. of Environmental and Prevention Sciences, University of Ferrara, Ferrara, Italy
| | - Franco Cervellati
- Dept. of Neuroscience and Rehabilitation, University of Ferrara, Ferrara, Italy
| | | | - Alessandra Pecorelli
- Dept. of Environmental and Prevention Sciences, University of Ferrara, Ferrara, Italy; Dept. of Food, Bioprocessing and Nutrition Sciences, Plants for Human Health Institute, North Carolina State University, Kannapolis, NC, USA.
| | - Giuseppe Valacchi
- Dept. of Environmental and Prevention Sciences, University of Ferrara, Ferrara, Italy; Animal Science Dept., Plants for Human Health Institute, North Carolina State University, Kannapolis, NC, USA; Dept. of Food and Nutrition, Kyung Hee University, Seoul, South Korea.
| |
Collapse
|
85
|
Abdel Mageed SS, Elimam H, Elesawy AE, Abulsoud AI, Raouf AA, Tabaa MME, Mohammed OA, Zaki MB, Abd-Elmawla MA, El-Dakroury WA, Mangoura SA, Elrebehy MA, Elballal MS, Mohamed AA, Ashraf A, Abdel-Reheim MA, Eleragi AMS, Abdellatif H, Doghish AS. Unraveling the impact of miRNAs on gouty arthritis: diagnostic significance and therapeutic opportunities. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024:10.1007/s00210-024-03603-9. [PMID: 39560752 DOI: 10.1007/s00210-024-03603-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Accepted: 11/03/2024] [Indexed: 11/20/2024]
Abstract
Gouty arthritis is a prevalent inflammatory illness. Gout attacks begin when there is an imbalance in the body's uric acid metabolism, which leads to urate buildup and the development of the ailment. A family of conserved, short non-coding RNAs known as microRNAs (miRNAs) can regulate post-transcriptional protein synthesis by attaching to the 3' untranslated region (UTR) of messenger RNA (mRNA). An increasing amount of research is pointing to miRNAs as potential players in several inflammatory diseases, including gouty arthritis. miRNAs may influence the progression of the disease by regulating immune function and inflammatory responses. This review mainly focused on miRNAs and how they contribute to gouty arthritis. It also looked at how miRNAs could be used as diagnostic, prognostic, and potential therapeutic targets.
Collapse
Affiliation(s)
- Sherif S Abdel Mageed
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Badr University in Cairo (BUC), Badr City, 11829, Cairo, Egypt
| | - Hanan Elimam
- Biochemistry, Department of Biochemistry, Faculty of Pharmacy, University of Sadat City, Sadat City, 32897, Menoufia, Egypt
| | - Ahmed E Elesawy
- Department of Biochemistry, Faculty of Pharmacy, Badr University in Cairo (BUC), Badr City, 11829, Cairo, Egypt
| | - Ahmed I Abulsoud
- Biochemistry Department, Faculty of Pharmacy, Heliopolis University, Cairo, 11785, Egypt
- Biochemistry and Molecular Biology Department, Faculty of Pharmacy (Boys), Al-Azhar University, Nasr City, 11231, Cairo, Egypt
| | - Ahmed Amr Raouf
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Badr University in Cairo (BUC), Badr City, 11829, Cairo, Egypt
| | - Manar Mohammed El Tabaa
- Pharmacology & Environmental Toxicology, Environmental Studies & Research Institute (ESRI), University of Sadat City, Sadat City, 32897, Menoufia, Egypt
| | - Osama A Mohammed
- Department of Pharmacology, College of Medicine, University of Bisha, 61922, Bisha, Saudi Arabia
| | - Mohamed Bakr Zaki
- Biochemistry, Department of Biochemistry, Faculty of Pharmacy, University of Sadat City, Sadat City, 32897, Menoufia, Egypt
| | - Mai A Abd-Elmawla
- Department of Biochemistry, Faculty of Pharmacy, Cairo University, Cairo, Egypt
| | - Walaa A El-Dakroury
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Badr University in Cairo (BUC), Badr City, 11829, Cairo, Egypt
| | - Safwat Abdelhady Mangoura
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Badr University in Cairo (BUC), Badr City, 11829, Cairo, Egypt
| | - Mahmoud A Elrebehy
- Department of Biochemistry, Faculty of Pharmacy, Galala University, New Galala City, 43713, Suez,, Egypt
| | - Mohammed S Elballal
- Department of Biochemistry, Faculty of Pharmacy, Badr University in Cairo (BUC), Badr City, 11829, Cairo, Egypt
- BK21 FOUR Team and Integrated Research Institute for Drug Development, College of Pharmacy, Dongguk University, Goyang, Republic of Korea
| | - Aya A Mohamed
- Department of Pharmacognosy, Faculty of Pharmacy, Badr University in Cairo (BUC), Badr City, 11829, Cairo, Egypt
| | - Alaa Ashraf
- Department of Clinical Pharmacy and Pharmacy Practice, Faculty of Pharmacy, Badr University in Cairo (BUC), Badr City, 11829, Cairo, Egypt
| | | | - Ali M S Eleragi
- Department of Microorganisms and Clinical Parasitology, College of Medicine, University of Bisha, 61922, Bisha, Saudi Arabia
| | - Hussein Abdellatif
- Department of Human and Clinical Anatomy, College of Medicine and Health Sciences, Sultan Qaboos University, Muscat, Oman
- Department of Anatomy and Embryology, Faculty of Medicine, University of Mansoura, Mansoura, 35516, Egypt
| | - Ahmed S Doghish
- Department of Biochemistry, Faculty of Pharmacy, Badr University in Cairo (BUC), Badr City, 11829, Cairo, Egypt.
- Biochemistry and Molecular Biology Department, Faculty of Pharmacy (Boys), Al-Azhar University, Nasr City, 11231, Cairo, Egypt.
| |
Collapse
|
86
|
Ge X, Zhang L, Ye M, Fei A, Wang L, Yuan A. Electroacupuncture improves diabetic cognitive impairment rats by suppressing NLRP3 inflammasome activation via induction of mitophagy. Metab Brain Dis 2024; 40:14. [PMID: 39560708 DOI: 10.1007/s11011-024-01464-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Accepted: 09/15/2024] [Indexed: 11/20/2024]
Abstract
Diabetic cognitive impairment pose a significant threat to public health in our aging society. However, the underlying molecular mechanisms have not been fully elucidated, which warrants further investigation. This study aimed to investigate the effects of electroacupuncture on cognitive impairment and its associated mechanisms. The diabetes model was induced via intraperitoneal injection of streptozotocin (STZ) into Sprague-Dawley rats combined with a high-fat and high-sugar diet. The learning and memory abilities of the rats were assessed using behavioral tests. Electron microscopy and hematoxylin-eosin (H&E) staining were used to identify the histological changes of neurons in the CA1 area of the rat hippocampal CA1. An examination of related indicators was performed by Western blotting including NLRP3 inflammasome-associated proteins Caspase-1, IL-18, IL1β, NLRP3, and P62, and mitophagy-related proteins Pink1, LC3, and Parkin. After modeling, rats displayed impaired learning and memory functions. The administration of electroacupuncture treatment alleviated diabetic cognitive impairment, described as shorter escape latency and an increased frequency of platform crossings. The damaged morphological and ultrastructural changes of neurons in rat hippocampal CA1 area can be alleviated through electroacupuncture treatment. Furthermore, in-depth studies suggest that electroacupuncture treatment can suppress NLRP3 inflammasome activation through induction of Pink1, LC3 and Parkin expression. Electroacupuncture treatment can attenuate NLRP3 inflammasome activation by promoting mitophagy, eventually improving cognitive impairment in (STZ)-treated rats with a high-fat die.
Collapse
Affiliation(s)
- Xia Ge
- Department of Endocrinology, The Second Affiliated Hospital of Anhui, University of Chinese Medicine, Hefei, 230001, Anhui, China
| | - Lele Zhang
- Anhui University of Chinese Medicine, Hefei, 230012, Anhui, China
| | - Min Ye
- Anhui University of Chinese Medicine, Hefei, 230012, Anhui, China
| | - Aihua Fei
- Department of Endocrinology, The Second Affiliated Hospital of Anhui, University of Chinese Medicine, Hefei, 230001, Anhui, China
| | - Ling Wang
- Wangjing Hospital of China Academy of Chinese Medical Sciences, Beijing, 100102, Anhui, China
| | - Aihong Yuan
- Department of acupuncture and rehabilitation, The First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei, 230031, Anhui, China.
| |
Collapse
|
87
|
Jantaruk P, Roytrakul S, Sistayanarain A, Kunthalert D. Potential role of the antimicrobial peptide Tachyplesin III in regulating nontypeable Haemophilus influenzae-induced inflammation in airway epithelial cells. Arch Microbiol 2024; 206:471. [PMID: 39560721 DOI: 10.1007/s00203-024-04196-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Revised: 11/04/2024] [Accepted: 11/06/2024] [Indexed: 11/20/2024]
Abstract
The respiratory bacterium nontypeable (non-encapsulated) Haemophilus influenzae (NTHi) is a key pathogen driving exacerbations in chronic obstructive pulmonary disease (COPD), and is associated with an excessive airway inflammation. Increasing issues with tolerance and unwanted side effects of existing pharmaceuticals present an urgent need for new, effective and minimally toxic therapeutic options. This study aimed to investigate the potential role of Tachyplesin III, an antimicrobial peptide derived from the hemolysates of Southeast Asian horseshoe crabs, in regulating NTHi-induced airway inflammation. The results revealed that Tachyplesin III effectively inhibited the production of IL-1β in NTHi-stimulated human lung epithelial cells (A549), without causing cytotoxic effects. Additionally, Tachyplesin III significantly reduced TNF-α, PGE2 and NO production in NTHi-stimulated A549 cells. Moreover, this peptide inhibited the nuclear translocation of the NF-κB p65 subunit in NTHi-stimulated lung epithelial cells. It also reduced transcriptional activation of NF-κB target genes, as shown by lower mRNA levels of IL-1β, TNF-α, COX-2 and iNOS, which correlated with corresponding decreases in their protein expression. Tachyplesin III peptide also inhibited pro-IL-1β and NLRP3 protein expression and prevented NTHi-induced caspase-1 cleavage and IL-1β maturation. Together, our findings demonstrate that Tachyplesin III effectively reduced NTHi-mediated inflammation via the NF-κB/NLRP3 inflammasome signaling pathway, highlighting its important anti-inflammatory activity. Complementing these findings, in silico analysis revealed key pharmacokinetic and toxicological attributes, establishing a foundational understanding of Tachyplesin III as a promising therapeutic agent for managing NTHi-associated inflammation.
Collapse
Affiliation(s)
- Pornpimon Jantaruk
- Department of Microbiology and Parasitology, Faculty of Medical Science, Naresuan University, Phitsanulok, 65000, Thailand
| | - Sittiruk Roytrakul
- Functional Proteomics Technology Laboratory, National Center for Genetic Engineering and Biotechnology, National Science and Technology Development Agency, Pathum Thani, 12120, Thailand
| | - Anchalee Sistayanarain
- Department of Microbiology and Parasitology, Faculty of Medical Science, Naresuan University, Phitsanulok, 65000, Thailand
| | - Duangkamol Kunthalert
- Department of Microbiology and Parasitology, Faculty of Medical Science, Naresuan University, Phitsanulok, 65000, Thailand.
- Centre of Excellence in Medical Biotechnology (CEMB), Faculty of Medical Science, Naresuan University, Phitsanulok, 65000, Thailand.
| |
Collapse
|
88
|
Plaisance EP, Bergeron JM, Bolyard ML, Hathorne HY, Graziano CM, Hartzes A, Genschmer KR, Alvarez JA, Goss AM, Gaggar A, Fontaine KR. Low-Dose Ketone Monoester Administration in Adults with Cystic Fibrosis: A Pilot and Feasibility Study. Nutrients 2024; 16:3957. [PMID: 39599743 PMCID: PMC11597165 DOI: 10.3390/nu16223957] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Revised: 11/12/2024] [Accepted: 11/15/2024] [Indexed: 11/29/2024] Open
Abstract
INTRODUCTION Cystic fibrosis transmembrane conductance regulator (CFTR) modulators have greatly improved outcomes in persons with CF (pwCF); however, there is still significant heterogeneity in clinical responses, particularly with regard to respiratory infection and inflammation. Exogenous administration of ketones has profound systemic anti-inflammatory effects and produces several nutrient-signaling and metabolic effects that may benefit multiple organ systems affected in pwCF. This pilot study was designed to determine the feasibility of administration of a ketone monoester (KME) to increase circulating D-beta hydroxybutyrate concentrations (D-βHB) and to improve subjective measures of CF-specific quality of life and markers of inflammation in serum and sputum in adults with CF. METHODS Fourteen participants receiving modulator therapy were randomized to receive either KME (n = 9) or placebo control (PC, n = 5) for 5-7 days during hospitalization for treatment of acute pulmonary exacerbation or as outpatients under standard care. RESULTS The KME was well tolerated, with only mild reports of gastrointestinal distress. D-βHB concentrations increased from 0.2 ± 0.1 mM to 1.6 ± 0.6 mM in the KME group compared to 0.2 ± 0.0 to 0.3 ± 0.1 in the PC group (p = 0.011) within 15 min following consumption and remained elevated, relative to baseline, for over 2 h. Pulmonary function was not altered after single- or short-term KME administration, but participants in the KME group self-reported higher subjective respiratory scores compared to PC in both cases (p = 0.031). Plasma inflammatory markers were not statistically different between groups following the short-term (5-7 d) intervention (p > 0.05). However, an exploratory analysis of plasma pre- and post-IL-6 concentrations was significant (p = 0.028) in the KME group but not PC. Sputum IFNγ (p = 0.057), IL-12p70 (p = 0.057), IL-1β (p = 0.100), IL-15 (p = 0.057), IL-1α (p = 0.114), and MPO (p = 0.133) were lower in the KME group compared to PC but did not achieve statistical significance. CONCLUSIONS With the emerging role of exogenous ketones as nutrient signaling molecules and mediators of metabolism, we showed that KME is well tolerated, increases circulating D-βHB concentrations, and produces outcomes that justify the need for large-scale clinical trials to investigate the role of KME on whole-body and tissue lipid accumulation and inflammation in pwCF.
Collapse
Affiliation(s)
- Eric P. Plaisance
- Department of Nutrition Sciences, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Jonathan M. Bergeron
- Division of Pulmonary, Allergy, & Critical Care Medicine, University of Alabama at Birmingham, Birmingham, AL 35294, USA (K.R.G.)
| | - Mickey L. Bolyard
- Department of Health & Kinesiology, University of Utah, Salt Lake City, UT 84112, USA
| | - Heather Y. Hathorne
- Division of Pulmonary, Allergy, & Critical Care Medicine, University of Alabama at Birmingham, Birmingham, AL 35294, USA (K.R.G.)
| | - Christina M. Graziano
- Department of Human Studies, University of Alabama at Birmingham, Birmingham, AL 35294, USA;
| | - Anastasia Hartzes
- Department of Biostatistics, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Kristopher R. Genschmer
- Division of Pulmonary, Allergy, & Critical Care Medicine, University of Alabama at Birmingham, Birmingham, AL 35294, USA (K.R.G.)
| | - Jessica A. Alvarez
- Division of Endocrinology, Metabolism and Lipids, Department of Medicine, Emory University, Atlanta, GA 30322, USA;
| | - Amy M. Goss
- Department of Nutrition Sciences, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Amit Gaggar
- Division of Pulmonary, Allergy, & Critical Care Medicine, University of Alabama at Birmingham, Birmingham, AL 35294, USA (K.R.G.)
| | - Kevin R. Fontaine
- Department of Health Behavior, University of Alabama at Birmingham, Birmingham, AL 35294, USA;
| |
Collapse
|
89
|
Qi WH, Tang N, Zhao ZJ, Li XQ. Transient receptor potential channels in viral infectious diseases: Biological characteristics and regulatory mechanisms. J Adv Res 2024:S2090-1232(24)00541-1. [PMID: 39551130 DOI: 10.1016/j.jare.2024.11.022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2024] [Revised: 11/11/2024] [Accepted: 11/12/2024] [Indexed: 11/19/2024] Open
Abstract
BACKGROUND Viral infectious diseases have long posed a challenge to humanity. In recent decades, transient receptor potential (TRP) channels have emerged as newly investigated cation channels. Increasing evidence suggests that TRP channel-mediated Ca2+ homeostasis disruptions, along with associated pathological changes, are critical factors in the onset and progression of viral infectious diseases. However, the precise roles and mechanisms of TRP channels in these diseases remain to be systematically elucidated. AIM OF REVIEW The aim of this review is to systematically summarize recent advances in understanding TRP channels in viral infections, and based on current progress and challenges, propose future directions for research. KEY SCIENTIFIC CONCEPTS OF REVIEW This review summarizes the classification and biological functions of the TRP family, explores the mechanisms by which TRP channels contribute to viral infections, and highlights specific mechanisms at three levels: virus, host, and outcome. These include the direct role in viral biology and replication, the indirect role in host immunity and inflammation, and the resulting pathological changes. Additionally, we discuss the potential applications of the TRP family in the treatment of viral infectious diseases and propose future research directions.
Collapse
Affiliation(s)
- Wen-Hui Qi
- Department of Chinese Materia Medica and Natural Medicines, School of Pharmacy, Air Force Medical University, Xi'an, Shaanxi 710032, China; Key Laboratory of Gastrointestinal Pharmacology of Chinese Materia Medica of the State Administration of Traditional Chinese Medicine, Department of Pharmacology, Air Force Medical University, Xi'an, Shaanxi 710032, China; Research Institution, Xijing Hospital, Air Force Medical University, Xi'an, Shaanxi 710032, China.
| | - Na Tang
- Department of Chinese Materia Medica and Natural Medicines, School of Pharmacy, Air Force Medical University, Xi'an, Shaanxi 710032, China; Key Laboratory of Gastrointestinal Pharmacology of Chinese Materia Medica of the State Administration of Traditional Chinese Medicine, Department of Pharmacology, Air Force Medical University, Xi'an, Shaanxi 710032, China; Research Institution, Xijing Hospital, Air Force Medical University, Xi'an, Shaanxi 710032, China.
| | - Zhi-Jing Zhao
- Research Institution, Xijing Hospital, Air Force Medical University, Xi'an, Shaanxi 710032, China; Department of Cardiology, Xijing Hospital, Air Force Medical University, Xi'an, Shaanxi 710032, China.
| | - Xiao-Qiang Li
- Department of Chinese Materia Medica and Natural Medicines, School of Pharmacy, Air Force Medical University, Xi'an, Shaanxi 710032, China; Key Laboratory of Gastrointestinal Pharmacology of Chinese Materia Medica of the State Administration of Traditional Chinese Medicine, Department of Pharmacology, Air Force Medical University, Xi'an, Shaanxi 710032, China; Research Institution, Xijing Hospital, Air Force Medical University, Xi'an, Shaanxi 710032, China.
| |
Collapse
|
90
|
Arcos M, Goodla L, Kim H, Desai SP, Liu R, Yin K, Liu Z, Martin DR, Xue X. PINK1-deficiency facilitates mitochondrial iron accumulation and colon tumorigenesis. Autophagy 2024:1-17. [PMID: 39512202 DOI: 10.1080/15548627.2024.2425594] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 10/25/2024] [Accepted: 11/01/2024] [Indexed: 11/15/2024] Open
Abstract
Mitophagy, the process by which cells eliminate damaged mitochondria, is mediated by PINK1 (PTEN induced kinase 1). Our recent research indicates that PINK1 functions as a tumor suppressor in colorectal cancer by regulating cellular metabolism. Interestingly, PINK1 ablation activated the NLRP3 (NLR family pyrin domain containing 3) inflammasome, releasing IL1B (interleukin 1 beta). However, inhibiting the NLRP3-IL1B signaling pathway with an IL1R (interleukin 1 receptor) antagonist or NLRP3 inhibitor did not hinder colon tumor growth after PINK1 loss. To identify druggable targets in PINK1-deficient tumors, ribonucleic acid sequencing analysis was performed on colon tumors from pink1 knockout and wild-type mice. Gene Set Enrichment Analysis highlighted the enrichment of iron ion transmembrane transporter activity. Subsequent qualitative polymerase chain reaction and western blot analysis revealed an increase in mitochondrial iron transporters, including mitochondrial calcium uniporter, in PINK1-deficient colon tumor cells and tissues. Live-cell iron staining demonstrated elevated cellular and mitochondrial iron levels in PINK1-deficient cells. Clinically used drugs deferiprone and minocycline reduced mitochondrial iron and superoxide levels, resulting in decreased colon tumor cell growth in vitro and in vivo. Manipulating the mitochondrial iron uptake protein MCU (mitochondrial calcium uniporter) also affected cell and xenograft tumor growth. This study suggests that therapies aimed at reducing mitochondrial iron levels may effectively inhibit colon tumor growth, particularly in patients with low PINK1 expression.Abbreviation: ANOVA: analysis of variance; APC: adenomatous polyposis coli; cAMP: cyclic adenosine monophosphate; CDX2: caudal type homeobox 2; CGAS: cyclic GMP-AMP synthase; CRC: colorectal cancer; DNA: deoxyribonucleic acid; DFP: deferiprone; DMEM: Dulbecco's modified Eagle medium; DSS: dextran sodium sulfate; ERT2-Cre: Cre recombinase fused to a triple mutant form of the human estrogen receptor; EV: empty vector; GLB: glybenclamide/glyburide; H&E: hematoxylin and eosin; ICP-MS: inductively coupled plasma mass spectrometer; IL1B: interleukin 1 beta; kDa: kilodalton; MCU: mitochondrial calcium uniporter; MKI67: marker of proliferation Ki-67; mRNA: messenger ribonucleic acid; MTT: 3-(4,5-dimethylthiazol-2-Yl)-2,5-diphenyltetrazolium bromide; NLRP3: NLR family pyrin domain containing 3; OE: overexpression; PBS: phosphate-buffered saline; p-CREB: phosphorylated cAMP responsive element binding protein; PINK1: PTEN induced kinase 1; p-PRKAA/AMPK: phosphorylated protein kinase AMP-activated catalytic subunit alpha; qPCR: qualitative polymerase chain reaction; RNA-seq: ribonucleic acid sequencing; ROS: reactive oxygen species; sg: single guide; sh: short hairpin; SLC25A28: solute carrier family 25 member 28; SLC25A37/MFRN: solute carrier family 25 member 37; STING1: stimulator of interferon response cGAMP interactor 1; TP53/p53: tumor protein p53; TUBA: tubulin alpha; µL: microliter; µm: micrometer; µM: micromolar; mm: millimeter.
Collapse
Affiliation(s)
- Mariella Arcos
- Department of Biochemistry and Molecular Biology, University of New Mexico, Albuquerque, NM, USA
| | - Lavanya Goodla
- Department of Biochemistry and Molecular Biology, University of New Mexico, Albuquerque, NM, USA
| | - Hyeoncheol Kim
- Department of Biochemistry and Molecular Biology, University of New Mexico, Albuquerque, NM, USA
| | - Sharina P Desai
- Department of Molecular Genetics Microbiology, University of New Mexico, Albuquerque, NM, USA
| | - Rui Liu
- Department of Pharmaceutical Sciences, University of New Mexico, Albuquerque, NM, USA
| | - Kunlun Yin
- Department of Biochemistry and Molecular Biology, University of New Mexico, Albuquerque, NM, USA
| | - Zhaoli Liu
- Department of Biochemistry and Molecular Biology, University of New Mexico, Albuquerque, NM, USA
| | - David R Martin
- Department of Pathology, University of New Mexico, Albuquerque, NM, USA
| | - Xiang Xue
- Department of Biochemistry and Molecular Biology, University of New Mexico, Albuquerque, NM, USA
| |
Collapse
|
91
|
Pu Y, Han S, Chen J, Liu Z. MANF inhibits NLRP3 inflammasome activation by competitively binding to DDX3X in paraquat-stimulated alveolar macrophages. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2024; 287:117331. [PMID: 39547060 DOI: 10.1016/j.ecoenv.2024.117331] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/07/2024] [Revised: 11/10/2024] [Accepted: 11/10/2024] [Indexed: 11/17/2024]
Abstract
NLRP3 inflammasome activation in macrophages is involved in paraquat-induced acute lung injury (ALI). MANF exerts an inhibitory effect against inflammation and cell death. The aim of this study was to investigate the role of MANF in paraquat-stimulated alveolar macrophages and the potential mechanism. Paraquat-induced ALI mouse model was established by intraperitoneally injection of 30 mg/kg of paraquat. The lung pathological changes were observed by hematoxylin and eosin staining. The expression of MANF/DDX3X/NLRP3/Caspase-1 in mice lung macrophages was evaluated by double immunofluorescence staining and western blot. NLRP3 inflammasome activation and pro-inflammatory cytokines (IL-1β and IL-18) in paraquat-stimulated macrophage transfected with MANF overexpression plasmid (pcDNA3.1-MANF) or siRNA-MANF were measured by Western blot. The protein-protein interaction of MANF/DDX3X/NLRP3 was verified by Co-immunoprecipitation. As a result, MANF/DDX3X/NLRP3/Caspase-1 were upregulated in alveolar macrophages of paraquat-induced ALI in mice. In paraquat-stimulated alveolar macrophages, upregulation of MANF and DDX3X were also observed, accompanied by NLRP3 inflammasome activation. In addition, overexpression of MANF inhibited NLRP3 inflammasome activation in paraquat-stimulated alveolar macrophages. In contrast, knockdown of MANF aggravated NLRP3 inflammasome activation. Co-immunoprecipitation results revealed that DDX3X could bind to MANF and NLRP3, but MANF could not bind to NLRP3 in paraquat-stimulated alveolar macrophages. Furthermore, Co-immunoprecipitation of truncated three fragments of DDX3X confirmed MANF can interact with the helicase core of DDX3X which is the binding site for NLRP3. Taken together, MANF exerted a protective effect against paraquat-induced cytotoxicity by inhibiting the NLRP3 inflammasome activation in macrophages via competitive binding to the helicase core of DDX3X.
Collapse
Affiliation(s)
- Yi Pu
- Department of Critical Care Medicine, Shengjing Hospital of China Medical University, No.36, Sanhao Street, Heping District, Shenyang, Liaoning 110004, China
| | - Siying Han
- Department of Critical Care Medicine, Shengjing Hospital of China Medical University, No.36, Sanhao Street, Heping District, Shenyang, Liaoning 110004, China
| | - Jie Chen
- Department of Cardiology, Shengjing Hospital of China Medical University, No.36, Sanhao Street, Heping District, Shenyang, Liaoning 110004, China
| | - Zhenning Liu
- Department of Critical Care Medicine, Shengjing Hospital of China Medical University, No.36, Sanhao Street, Heping District, Shenyang, Liaoning 110004, China.
| |
Collapse
|
92
|
Liu Y, Fu X, Sun J, Cui R, Yang W. AdipoRon exerts an antidepressant effect by inhibiting NLRP3 inflammasome activation in microglia via promoting mitophagy. Int Immunopharmacol 2024; 141:113011. [PMID: 39213872 DOI: 10.1016/j.intimp.2024.113011] [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: 07/07/2024] [Revised: 08/14/2024] [Accepted: 08/21/2024] [Indexed: 09/04/2024]
Abstract
Depression is a serious mental disorder that threatens patients' physical and mental health worldwide. The activation of the NLR family pyrin domain-containing 3 (NLRP3) inflammasome is essential for microglia-mediated neuroinflammation and neuronal damage in depression. Numerous pathophysiological factors, such as mitochondrial dysfunction and impaired mitophagy, have an essential role in activating the NLRP3 inflammasome. AdipoRon is a potent adiponectin receptor agonist; however, its antidepressant effects have not been thoroughly investigated. In this study, we found that AdipoRon ameliorated depression-like behavior and neuronal damage induced by chronic unpredictable mild stress (CUMS). Further research demonstrated that AdipoRon inhibited the activation of the NLRP3 inflammasome and protected hippocampal neurons from microglial cytotoxicity by promoting mitophagy, increasing the clearance of damaged mitochondria, and reducing mtROS accumulation. Importantly, inhibition of mitophagy attenuated the antidepressant and neuroprotective effects of AdipoRon. Overall, these findings indicate that AdipoRon alleviates depression by inhibiting NLRP3 inflammasome activation in microglia via improving mitophagy.
Collapse
Affiliation(s)
- Yaqi Liu
- Department of Neurology, The Second Hospital of Jilin University, Changchun, Jilin Province, China; Jilin Provincial Key Laboratory on Molecular and Chemical Genetics, The Second Hospital of Jilin University, Changchun, Jilin Province, China
| | - Xiying Fu
- Jilin Provincial Key Laboratory on Molecular and Chemical Genetics, The Second Hospital of Jilin University, Changchun, Jilin Province, China; Department of Endocrinology, The Second Hospital of Jilin University, Changchun, Jilin Province, China
| | - Jiangjin Sun
- Department of Neurology, The Second Hospital of Jilin University, Changchun, Jilin Province, China; Jilin Provincial Key Laboratory on Molecular and Chemical Genetics, The Second Hospital of Jilin University, Changchun, Jilin Province, China
| | - Ranji Cui
- Jilin Provincial Key Laboratory on Molecular and Chemical Genetics, The Second Hospital of Jilin University, Changchun, Jilin Province, China.
| | - Wei Yang
- Department of Neurology, The Second Hospital of Jilin University, Changchun, Jilin Province, China; Jilin Provincial Key Laboratory on Molecular and Chemical Genetics, The Second Hospital of Jilin University, Changchun, Jilin Province, China.
| |
Collapse
|
93
|
Leal VNC, Bork F, Mateo Tortola M, von Guilleaume JC, Greve CL, Bugl S, Danker B, Bittner ZA, Grimbacher B, Pontillo A, Weber ANR. Bruton's tyrosine kinase (BTK) and matrix metalloproteinase-9 (MMP-9) regulate NLRP3 inflammasome-dependent cytokine and neutrophil extracellular trap responses in primary neutrophils. J Allergy Clin Immunol 2024:S0091-6749(24)01186-2. [PMID: 39547282 DOI: 10.1016/j.jaci.2024.10.035] [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: 02/26/2024] [Revised: 10/02/2024] [Accepted: 10/22/2024] [Indexed: 11/17/2024]
Abstract
BACKGROUND Inflammation is a double-edged state of immune activation that is required to resolve threats harmful to the host, but can also cause severe collateral damage. Polymorphonuclear neutrophils (PMNs), the primary leukocyte population in humans, mediate inflammation through the release of cytokines and neutrophil extracellular traps (NETs). Although the pathophysiological importance of NETs is unequivocal, the multiple molecular pathways driving NET release are not fully defined. Recently, NET release was linked to the NLRP3 inflammasome, which is regulated by Bruton's tyrosine kinase (BTK) in macrophages. OBJECTIVE As NLRP3 inflammasome regulation by BTK has not been studied in neutrophils, we explored a potential regulatory role of BTK in primary murine and human neutrophils and matched monocytes or macrophages from Btk-deficient versus wild-type mice, or from healthy donors versus BTK-deficient patients with X-linked agammaglobulinemia. METHODS Cytokine, myeloperoxidase, and matrix metalloproteinase-9 (MMP-9) release were quantified by ELISA, NET release, and inflammasome formation by immunofluorescence microscopy. RESULTS Surprisingly, in both mouse and human primary neutrophils, we observed a significant increase in NLRP3 inflammasome-dependent IL-1β and NETs when BTK was absent or inhibited, whereas IL-1β release was decreased in corresponding primary mouse macrophages or human PBMCs. This suggests a novel negative regulatory role of BTK in terms of neutrophil NLRP3 activation. IL-1β and NET release in both mouse and human primary neutrophils was strictly dependent on NLRP3, caspase-1 and, surprisingly, MMP-9. CONCLUSIONS This study highlights BTK and MMP-9 as novel and versatile inflammasome regulators and may have implications for the clinical use of BTK inhibitors.
Collapse
Affiliation(s)
- Vinicius N C Leal
- Institute of Immunology, Department of Innate Immunity, University of Tübingen, Tübingen, Germany; Laboratory of Immunogenetics, Department of Immunology, Institute of Biomedical Science, University of São Paulo, São Paulo, Brazil
| | - Francesca Bork
- Institute of Immunology, Department of Innate Immunity, University of Tübingen, Tübingen, Germany
| | - Maria Mateo Tortola
- Institute of Immunology, Department of Innate Immunity, University of Tübingen, Tübingen, Germany
| | | | - Carsten L Greve
- Institute of Immunology, Department of Innate Immunity, University of Tübingen, Tübingen, Germany
| | - Stefanie Bugl
- Institute of Immunology, Department of Innate Immunity, University of Tübingen, Tübingen, Germany
| | - Bettina Danker
- Institute of Immunology, Department of Innate Immunity, University of Tübingen, Tübingen, Germany
| | - Zsofia A Bittner
- Institute of Immunology, Department of Innate Immunity, University of Tübingen, Tübingen, Germany
| | - Bodo Grimbacher
- Klinik für Rheumatologie/Klinische Immunologie, Universitätsklinikum Freiburg, Freiburg, Germany; DZIF - German Center for Infection Research, Satellite Center Freiburg, Freiburg, Germany; CIBSS - Centre for Integrative Biological Signalling Studies, Albert-Ludwigs University, Freiburg, Germany; RESIST - Cluster of Excellence 2155 to Hanover Medical School, Satellite Center Freiburg, Freiburg, Germany
| | - Alessandra Pontillo
- Laboratory of Immunogenetics, Department of Immunology, Institute of Biomedical Science, University of São Paulo, São Paulo, Brazil
| | - Alexander N R Weber
- Institute of Immunology, Department of Innate Immunity, University of Tübingen, Tübingen, Germany; iFIT-Cluster of Excellence (EXC 2180) "Image-Guided and Functionally Instructed Tumor Therapies," University of Tübingen, Tübingen, Germany; German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ) Partner Site Tübingen, Tübingen, Germany; CMFI-Cluster of Excellence (EXC 2124) "Controlling Microbes to Fight Infection," University of Tübingen, Tübingen, Germany.
| |
Collapse
|
94
|
Tsuruno Y, Nagano A, Sugita K, Onishi S, Tabata Y, Kedoin C, Murakami M, Yano K, Kawano T, Hasuzawa N, Nomura M, Kaji T, Bitoh Y, Ieiri S. Favorable inhibitory effect of clodronate on hepatic steatosis in short bowel syndrome model rats. Pediatr Surg Int 2024; 40:307. [PMID: 39537943 PMCID: PMC11561034 DOI: 10.1007/s00383-024-05858-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 10/16/2024] [Indexed: 11/16/2024]
Abstract
PURPOSE This study investigated the anti-inflammatory effect of clodronate, a vesicular nucleotide transporter (VNUT) inhibitor, on intestinal-failure-associated liver disease (IFALD) in a rat model of short bowel syndrome (SBS). METHODS The rats underwent jugular vein catheterization for continuous total parenteral nutrition (TPN) and 90% small bowel resection. The animals were divided into the following groups: TPN/SBS (Control group), TPN/SBS/intravenous administration of low-dose clodronate (20 mg/kg twice per week; Low group), or TPN/SBS/intravenous administration of high-dose clodronate (60 mg/kg twice per week; High group). On day 7, the rats were euthanized. Hepatic steatosis and hepatocellular injury were also assessed. RESULTS Hepatic steatosis and lobular inflammation in the liver were observed in all groups. The High group showed histologically reduced hepatic steatosis compared with the Control group. IL-6 and Nlrp3 expression in the High group was significantly suppressed compared to that in the Control group. The expression of other inflammatory cytokines tended to be lower in the High dose group than in the control group. The lipid metabolism gene expression in the liver specimens showed no significant differences among the groups. CONCLUSION The high-dose administration of clodronate may, therefore, inhibit hepatic steatosis and inflammation associated with IFALD in patients with SBS.
Collapse
Affiliation(s)
- Yudai Tsuruno
- Department of Pediatric Surgery, Medical and Dental Area, Research and Education Assembly, Research Field in Medical and Health Sciences, Kagoshima University, 8-35-1, Sakuragaoka, Kagoshima, 890-8520, Japan
- Division of Pediatric Surgery, Department of Surgery, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Ayaka Nagano
- Department of Pediatric Surgery, Medical and Dental Area, Research and Education Assembly, Research Field in Medical and Health Sciences, Kagoshima University, 8-35-1, Sakuragaoka, Kagoshima, 890-8520, Japan
| | - Koshiro Sugita
- Department of Pediatric Surgery, Medical and Dental Area, Research and Education Assembly, Research Field in Medical and Health Sciences, Kagoshima University, 8-35-1, Sakuragaoka, Kagoshima, 890-8520, Japan.
| | - Shun Onishi
- Department of Pediatric Surgery, Medical and Dental Area, Research and Education Assembly, Research Field in Medical and Health Sciences, Kagoshima University, 8-35-1, Sakuragaoka, Kagoshima, 890-8520, Japan
| | - Yumiko Tabata
- Department of Pediatric Surgery, Medical and Dental Area, Research and Education Assembly, Research Field in Medical and Health Sciences, Kagoshima University, 8-35-1, Sakuragaoka, Kagoshima, 890-8520, Japan
| | - Chihiro Kedoin
- Department of Pediatric Surgery, Medical and Dental Area, Research and Education Assembly, Research Field in Medical and Health Sciences, Kagoshima University, 8-35-1, Sakuragaoka, Kagoshima, 890-8520, Japan
| | - Masakazu Murakami
- Department of Pediatric Surgery, Medical and Dental Area, Research and Education Assembly, Research Field in Medical and Health Sciences, Kagoshima University, 8-35-1, Sakuragaoka, Kagoshima, 890-8520, Japan
| | - Keisuke Yano
- Department of Pediatric Surgery, Medical and Dental Area, Research and Education Assembly, Research Field in Medical and Health Sciences, Kagoshima University, 8-35-1, Sakuragaoka, Kagoshima, 890-8520, Japan
| | - Takafumi Kawano
- Department of Pediatric Surgery, Medical and Dental Area, Research and Education Assembly, Research Field in Medical and Health Sciences, Kagoshima University, 8-35-1, Sakuragaoka, Kagoshima, 890-8520, Japan
| | - Nao Hasuzawa
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Kurume University School of Medicine, Kurume, Japan
| | - Masatoshi Nomura
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Kurume University School of Medicine, Kurume, Japan
| | - Tatsuru Kaji
- Department of Pediatric Surgery, Kurume University School of Medicine, Kurume, Japan
| | - Yuko Bitoh
- Division of Pediatric Surgery, Department of Surgery, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Satoshi Ieiri
- Department of Pediatric Surgery, Medical and Dental Area, Research and Education Assembly, Research Field in Medical and Health Sciences, Kagoshima University, 8-35-1, Sakuragaoka, Kagoshima, 890-8520, Japan
| |
Collapse
|
95
|
Kim WI, Pak SW, Lee SJ, Park SH, Lim JO, Kim DI, Shin IS, Kim SH, Kim JC. Copper oxide nanoparticles exacerbate chronic obstructive pulmonary disease by activating the TXNIP-NLRP3 signaling pathway. Part Fibre Toxicol 2024; 21:46. [PMID: 39529109 PMCID: PMC11552314 DOI: 10.1186/s12989-024-00608-3] [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: 07/03/2024] [Accepted: 10/28/2024] [Indexed: 11/16/2024] Open
Abstract
BACKGROUND Although copper oxide nanoparticles (CuONPs) offer certain benefits to humans, they can be toxic to organs and exacerbate underlying diseases upon exposure. Chronic obstructive pulmonary disease (COPD), induced by smoking, can worsen with exposure to various harmful particles. However, the specific impact of CuONPs on COPD and the underlying mechanisms remain unknown. In this study, we investigated the toxic effects of CuONPs on the respiratory tract, the pathophysiology of CuONPs exposure-induced COPD, and the mechanism of CuONPs toxicity, focusing on thioredoxin-interacting protein (TXNIP) signaling using a cigarette smoke condensate (CSC)-induced COPD model. RESULTS In the toxicity study, CuONPs exposure induced an inflammatory response in the respiratory tract, including inflammatory cell infiltration, cytokine production, and mucus secretion, which were accompanied by increased TXNIP, NOD-like receptor protein 3 (NLRP3), caspase-1, and interleukin (IL)-1β. In the COPD model, CuONPs exposure induced the elevation of various indexes related to COPD, as well as increased TXNIP expression. Additionally, TNXIP-knockout (KO) mice showed a significantly decreased expression of NLRP3, caspase-1, and IL-1β and inflammatory responses in CuONPs-exposed COPD mice. These results were consistent with the results of an in vitro experiment using H292 cells. By contrast, TNXIP-overexpressed mice had a markedly increased expression of NLRP3, caspase-1, and IL-1β and inflammatory responses in CuONPs-exposed COPD mice. CONCLUSIONS We elucidated the exacerbating effect of CuONPs exposure on the respiratory tract with underlying COPD, as well as related signaling transduction via TXNIP regulation. CuONPs exposure significantly increased inflammatory responses in the respiratory tract, which was correlated with elevated TXNIP-NLRP3 signaling.
Collapse
Affiliation(s)
- Woong-Il Kim
- College of Veterinary Medicine and BK21 FOUR Program, Chonnam National University, Gwangju, 61186, Republic of Korea
| | - So-Won Pak
- College of Veterinary Medicine and BK21 FOUR Program, Chonnam National University, Gwangju, 61186, Republic of Korea
| | - Se-Jin Lee
- College of Veterinary Medicine and BK21 FOUR Program, Chonnam National University, Gwangju, 61186, Republic of Korea
| | - Sin-Hyang Park
- College of Veterinary Medicine and BK21 FOUR Program, Chonnam National University, Gwangju, 61186, Republic of Korea
| | - Je-Oh Lim
- Herbal Medicine Resources Research Center, Korea Institute of Oriental Medicine, Naju, 58245, Republic of Korea
| | - Dong-Il Kim
- College of Veterinary Medicine and BK21 FOUR Program, Chonnam National University, Gwangju, 61186, Republic of Korea
| | - In-Sik Shin
- College of Veterinary Medicine and BK21 FOUR Program, Chonnam National University, Gwangju, 61186, Republic of Korea
| | - Sung-Hwan Kim
- Jeonbuk Branch Institute, Korea Institute of Toxicology, Jeongeup, 56212, Republic of Korea.
| | - Jong-Choon Kim
- College of Veterinary Medicine and BK21 FOUR Program, Chonnam National University, Gwangju, 61186, Republic of Korea.
| |
Collapse
|
96
|
Li L, Xu T, Qi X. Balanced regulation of ROS production and inflammasome activation in preventing early development of colorectal cancer. Immunol Rev 2024. [PMID: 39523732 DOI: 10.1111/imr.13417] [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] [Indexed: 11/16/2024]
Abstract
Reactive oxygen species (ROS) production and inflammasome activation are the key components of the innate immune response to microbial infection and sterile insults. ROS are at the intersection of inflammation and immunity during cancer development. Balanced regulation of ROS production and inflammasome activation serves as the central hub of innate immunity, determining whether a cell will survive or undergo cell death. However, the mechanisms underlying this balanced regulation remain unclear. Mitochondria and NADPH oxidases are the two major sources of ROS production. Recently, NCF4, a component of the NADPH oxidase complex that primarily contributes to ROS generation in phagocytes, was reported to balance ROS production and inflammasome activation in macrophages. The phosphorylation and puncta distribution of NCF4 shifts from the membrane-bound NADPH complex to the perinuclear region, promoting ASC speck formation and inflammasome activation, which triggers downstream IL-18-IFN-γ signaling to prevent the progression of colorectal cancer (CRC). Here, we review ROS signaling and inflammasome activation studies in colitis-associated CRC and propose that NCF4 acts as a ROS sensor that balances ROS production and inflammasome activation. In addition, NCF4 is a susceptibility gene for Crohn's disease (CD) and CRC. We discuss the evidence demonstrating NCF4's crucial role in facilitating cell-cell contact between immune cells and intestinal cells, and mediating the paracrine effects of inflammatory cytokines and ROS. This coordination of the signaling network helps create a robust immune microenvironment that effectively prevents epithelial cell mutagenesis and tumorigenesis during the early stage of colitis-associated CRC.
Collapse
Affiliation(s)
- Longjun Li
- Key Laboratory for Experimental Teratology of the Ministry of Education, Advanced Medical Research Institute, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Tao Xu
- Key Laboratory for Experimental Teratology of the Ministry of Education, Advanced Medical Research Institute, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Xiaopeng Qi
- Key Laboratory for Experimental Teratology of the Ministry of Education, Advanced Medical Research Institute, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
- State Key Laboratory for Innovation and Transformation of Luobing Theory; The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, Jinan, Shandong, China
| |
Collapse
|
97
|
Ashmore AA, Balasubramanian B, Phillips A, Asher V, Bali A, Ordóñez-Morán P, Khan R. Bioinformatic and experimental data pertaining to the role of the NLRP3 inflammasome in ovarian cancer. J Cancer Res Clin Oncol 2024; 150:488. [PMID: 39516433 PMCID: PMC11549120 DOI: 10.1007/s00432-024-05988-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2024] [Accepted: 10/04/2024] [Indexed: 11/16/2024]
Abstract
The Nod-Like Receptor (NLR) family pyrin domain containing 3 (NLRP3) inflammasome plays a role in regulating inflammatory signaling and is a well-established contributor to pyroptotic cell death. It has been investigated extensively in cancer but there remains limited evidence of its role within ovarian cancer (OC). Bioinformatic investigation of gene expression data has highlighted that higher expression of NLRP3 and genes associated with the NLRP3 complex appear to be positively correlated with OC and may also have prognostic significance. However, heterogeneity exists within the results and experimental data is limited and contradictory. If the NLRP3 inflammasome is to be exploited as a therapeutic target, further laboratory-based investigation is required to determine its role in cancer. Furthermore, its relationship with clinically important characteristics such as histopathological subtype may be of key significance in developing targeted therapies towards specific cohorts of patients.
Collapse
Affiliation(s)
- Ayisha A Ashmore
- Derby Gynaecological Cancer Centre, Royal Derby Hospital, University Hospitals of Derby and Burton, Derby, UK.
- Translational Medical Sciences Unit, School of Medicine, University of Nottingham, Nottingham, UK.
| | - Brinda Balasubramanian
- Translational Medical Sciences Unit, Biodiscovery Institute, Centre for Cancer Sciences, School of Medicine, University of Nottingham, Nottingham, UK
| | - Andrew Phillips
- Translational Medical Sciences Unit, School of Medicine, University of Nottingham, Nottingham, UK
| | - Viren Asher
- Translational Medical Sciences Unit, School of Medicine, University of Nottingham, Nottingham, UK
| | - Anish Bali
- Derby Gynaecological Cancer Centre, Royal Derby Hospital, University Hospitals of Derby and Burton, Derby, UK
- Translational Medical Sciences Unit, School of Medicine, University of Nottingham, Nottingham, UK
| | - Paloma Ordóñez-Morán
- Translational Medical Sciences Unit, Biodiscovery Institute, Centre for Cancer Sciences, School of Medicine, University of Nottingham, Nottingham, UK
| | - Raheela Khan
- Translational Medical Sciences Unit, School of Medicine, University of Nottingham, Nottingham, UK
| |
Collapse
|
98
|
Lamantia V, Bissonnette S, Beaudry M, Cyr Y, Rosiers CD, Baass A, Faraj M. EPA and DHA inhibit LDL-induced upregulation of human adipose tissue NLRP3 inflammasome/IL-1β pathway and its association with diabetes risk factors. Sci Rep 2024; 14:27146. [PMID: 39511203 PMCID: PMC11543682 DOI: 10.1038/s41598-024-73672-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Accepted: 09/19/2024] [Indexed: 11/15/2024] Open
Abstract
Elevated numbers of atherogenic lipoproteins (apoB) predict the incidence of type 2 diabetes (T2D). We reported that this may be mediated via the activation of the NLRP3 inflammasome, as low-density lipoproteins (LDL) induce interleukin-1 beta (IL-1β) secretion from human white adipose tissue (WAT) and macrophages. However, mitigating nutritional approaches remained unknown. We tested whether omega-3 eicosapentaenoic and docosahexaenoic acids (EPA and DHA) treat LDL-induced upregulation of WAT IL-1β-secretion and its relation to T2D risk factors. Twelve-week intervention with EPA and DHA (2.7 g/day, Webber Naturals) abolished baseline group-differences in WAT IL-1β-secretion between subjects with high-apoB (N = 17) and low-apoB (N = 16) separated around median plasma apoB. Post-intervention LDL failed to trigger IL-1β-secretion and inhibited it in lipopolysaccharide-stimulated WAT. Omega-3 supplementation also improved β-cell function and postprandial fat metabolism in association with higher blood EPA and mostly DHA. It also blunted the association of WAT NLRP3 and IL1B expression and IL-1β-secretion with multiple cardiometabolic risk factors including adiposity. Ex vivo, EPA and DHA inhibited WAT IL-1β-secretion in a dose-dependent manner. In conclusion, EPA and DHA treat LDL-induced upregulation of WAT NLRP3 inflammasome/IL-1β pathway and related T2D risk factors. This may aid in the prevention of T2D and related morbidities in subjects with high-apoB.Clinical Trail Registration ClinicalTrials.gov (NCT04496154): Omega-3 to Reduce Diabetes Risk in Subjects with High Number of Particles That Carry "Bad Cholesterol" in the Blood - Full Text View - ClinicalTrials.gov.
Collapse
Affiliation(s)
- Valérie Lamantia
- Faculty of Medicine, Université de Montréal, Montréal, QC, Canada
- Institut de Recherches Cliniques de Montréal (IRCM), 110, Avenue des Pins Ouest, Montréal, QC, H2W 1R7, Canada
- Montréal Diabetes Research Center (MDRC), Montréal, QC, Canada
| | - Simon Bissonnette
- Faculty of Medicine, Université de Montréal, Montréal, QC, Canada
- Institut de Recherches Cliniques de Montréal (IRCM), 110, Avenue des Pins Ouest, Montréal, QC, H2W 1R7, Canada
- Montréal Diabetes Research Center (MDRC), Montréal, QC, Canada
| | - Myriam Beaudry
- Faculty of Medicine, Université de Montréal, Montréal, QC, Canada
- Institut de Recherches Cliniques de Montréal (IRCM), 110, Avenue des Pins Ouest, Montréal, QC, H2W 1R7, Canada
| | - Yannick Cyr
- Faculty of Medicine, Université de Montréal, Montréal, QC, Canada
- Institut de Recherches Cliniques de Montréal (IRCM), 110, Avenue des Pins Ouest, Montréal, QC, H2W 1R7, Canada
- Montréal Diabetes Research Center (MDRC), Montréal, QC, Canada
| | - Christine Des Rosiers
- Faculty of Medicine, Université de Montréal, Montréal, QC, Canada
- Montréal Heart Institute, Montréal, QC, Canada
| | - Alexis Baass
- Faculty of Medicine, Université de Montréal, Montréal, QC, Canada
- Institut de Recherches Cliniques de Montréal (IRCM), 110, Avenue des Pins Ouest, Montréal, QC, H2W 1R7, Canada
- Faculty of Medicine, McGill University, Montreal, QC, Canada
| | - May Faraj
- Faculty of Medicine, Université de Montréal, Montréal, QC, Canada.
- Institut de Recherches Cliniques de Montréal (IRCM), 110, Avenue des Pins Ouest, Montréal, QC, H2W 1R7, Canada.
- Montréal Diabetes Research Center (MDRC), Montréal, QC, Canada.
- Faculty of Medicine, McGill University, Montreal, QC, Canada.
| |
Collapse
|
99
|
Smyth T, Payton A, Hickman E, Rager JE, Jaspers I. Leveraging a comprehensive unbiased RNAseq database to characterize human monocyte-derived macrophage gene expression profiles within commonly employed in vitro polarization methods. Sci Rep 2024; 14:26753. [PMID: 39500943 PMCID: PMC11538326 DOI: 10.1038/s41598-024-78000-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2024] [Accepted: 10/28/2024] [Indexed: 11/08/2024] Open
Abstract
Macrophages are pivotal innate immune cells which exhibit high phenotypic plasticity and can exist in different polarization states dependent on exposure to external stimuli. Numerous methods have been employed to simulate macrophage polarization states to test their function in vitro. However, limited research has explored whether these polarization methods yield comparable populations beyond key gene, cytokine, and cell surface marker expression. Here, we employ an unbiased comprehensive analysis using data organized through the all RNA-seq and ChIP-seq sample and signature search (ARCHS4) database, which compiles all RNAseq data deposited into the National Center for Biotechnology Information (NCBI) Sequence Read Archive (SRA). In silico analyses were carried out demonstrating that commonly employed macrophage polarization methods generate distinct gene expression profiles in macrophage subsets that remained poorly described until now. Our analyses confirm existing knowledge on broad macrophage polarization, while expanding nuanced differences between M2a and M2c subsets, suggesting non-interchangeable stimuli for M2a polarization. Furthermore, we characterize divergent gene expression patterns in M1 macrophages following standard polarization protocols, indicating significant subset distinctions. Consequently, equivalence cannot be assumed among polarization regimens for in vitro macrophage studies, particularly in simulating diverse pathogen responses.
Collapse
Affiliation(s)
- Timothy Smyth
- Curriculum in Toxicology & Environmental Medicine, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Center for Environmental Medicine, Asthma, and Lung Biology, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Alexis Payton
- Center for Environmental Medicine, Asthma, and Lung Biology, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Elise Hickman
- Curriculum in Toxicology & Environmental Medicine, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Department of Environmental Sciences and Engineering, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Julia E Rager
- Curriculum in Toxicology & Environmental Medicine, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Center for Environmental Medicine, Asthma, and Lung Biology, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Department of Environmental Sciences and Engineering, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Ilona Jaspers
- Curriculum in Toxicology & Environmental Medicine, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.
- Center for Environmental Medicine, Asthma, and Lung Biology, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.
- Department of Pediatrics, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.
- , 116 Manning Drive, Campus Box 7310, Chapel Hill, NC, 27599-7310, USA.
| |
Collapse
|
100
|
Duret T, Elmallah M, Rollin J, Gatault P, Jiang LH, Roger S. Role of purinoreceptors in the release of extracellular vesicles and consequences on immune response and cancer progression. Biomed J 2024:100805. [PMID: 39510381 DOI: 10.1016/j.bj.2024.100805] [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: 09/04/2024] [Revised: 10/24/2024] [Accepted: 11/02/2024] [Indexed: 11/15/2024] Open
Abstract
Cell-to-cell communication is a major process for accommodating cell functioning to changes in the environments and to preserve tissue and organism homeostasis. It is achieved by different mechanisms characterized by the origin of the message, the molecular nature of the messenger, its speed of action and its reach. Purinergic signalling is a powerful mechanism initiated by extracellular nucleotides, such as ATP, acting on plasma membrane purinoreceptors. Purinergic signalling is tightly controlled in time and space by the action of ectonucleotidases. Recent studies have highlighted the critical role of purinergic signalling in controlling the generation, release and fate of extracellular vesicles and, in this way, mediating long-distance responses. Most of these discoveries have been made in immune and cancer cells. This review is aimed at establishing the current knowledge on the way which purinoreceptors control extracellular vesicle-mediated communications and consequences for recipient cells.
Collapse
Affiliation(s)
- Thomat Duret
- Université de Tours, Inserm UMR1327 ISCHEMIA « Membrane Signalling and Inflammation in Reperfusion Injuries », Tours, France; Fédération Hospitalo-Universitaire Survival optimization in organ Transplantation (FHU SUPORT), Tours, France
| | - Mohammed Elmallah
- Université de Tours, Inserm UMR1327 ISCHEMIA « Membrane Signalling and Inflammation in Reperfusion Injuries », Tours, France
| | - Jérôme Rollin
- Université de Tours, Inserm UMR1327 ISCHEMIA « Membrane Signalling and Inflammation in Reperfusion Injuries », Tours, France; Service d'Hématologie-Hémostase, CHRU de Tours, Tours, France
| | - Philippe Gatault
- Université de Tours, Inserm UMR1327 ISCHEMIA « Membrane Signalling and Inflammation in Reperfusion Injuries », Tours, France; Service de Néphrologie, Hypertension, Dialyse et Transplantation Rénale, CHRU Tours, Tours, France; Fédération Hospitalo-Universitaire Survival optimization in organ Transplantation (FHU SUPORT), Tours, France
| | - Lin-Hua Jiang
- Université de Tours, Inserm UMR1327 ISCHEMIA « Membrane Signalling and Inflammation in Reperfusion Injuries », Tours, France; School of Basic Medical Sciences, Xinxiang Medical University, China; School of Biomedical Sciences, University of Leeds, Leeds, United Kingdom
| | - Sébastien Roger
- Université de Tours, Inserm UMR1327 ISCHEMIA « Membrane Signalling and Inflammation in Reperfusion Injuries », Tours, France; Fédération Hospitalo-Universitaire Survival optimization in organ Transplantation (FHU SUPORT), Tours, France.
| |
Collapse
|