1
|
Tan M, Chu JSC, Swiger DR. Exploring the Medicinal Potential of Taraxacum Kok-Saghyz ( TKS) Using Widely Targeted Metabolomics. Metabolites 2025; 15:306. [PMID: 40422883 DOI: 10.3390/metabo15050306] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2025] [Revised: 04/13/2025] [Accepted: 04/28/2025] [Indexed: 05/28/2025] Open
Abstract
Background/Objectives: Plant-derived secondary metabolites have long contributed to the discovery of novel therapeutic agents, especially in the treatment of parasitic and infectious diseases in developing countries. Metabolomics provides a systems-level approach to understanding plant biochemistry, enabling the discovery of secondary metabolites with pharmacological relevance. Taraxacum kok-saghyz (TKS), widely known for its rubber-producing capabilities, remains underexplored as a medicinal plant. Given the well-established therapeutic properties of Taraxacum officinale and the emerging pharmacological profiles of related species, this study investigates the metabolic composition of TKS roots and leaves to uncover bioactive compounds with antioxidant, anti-inflammatory, or hepatoprotective potential. Methods: Widely targeted metabolomics was conducted on 10-month-old field-grown Kultevar™ TKS plants using ultra-performance liquid chromatography-tandem mass spectrometry (UPLC-MS/MS). Samples were hand-harvested and preserved on dry ice to maintain biochemical integrity. Metabolite identification and classification were performed using the MWDB and KEGG databases. Principal component analysis (PCA) and orthogonal partial least squares discriminant analysis (OPLS-DA) were used to evaluate metabolic variation between tissues. Results: A total of 1813 metabolites were identified, including flavonoids, alkaloids, lipids, amino acids, and phenolic compounds. Differential analysis revealed 964 significantly altered metabolites-609 downregulated and 355 upregulated in roots relative to leaves. Multivariate analysis confirmed clear tissue-specific metabolic profiles. KEGG pathway enrichment highlighted the involvement of flavonoid biosynthesis, amino acid metabolism, and lipid metabolism pathways, suggesting bioactive potential. This study presents the first comprehensive metabolic profile of TKS, highlighting its potential value beyond rubber production. The detection of numerous therapeutic secondary metabolites supports its promise as a pharmaceutical and nutraceutical resource. Further functional validation of identified compounds is warranted.
Collapse
Affiliation(s)
- Michele Tan
- Kultevat Inc., 1100 Corporate Square Drive Suite 261, Creve Coeur, MO 63132, USA
| | | | - Daniel Robin Swiger
- Kultevat Inc., 1100 Corporate Square Drive Suite 261, Creve Coeur, MO 63132, USA
| |
Collapse
|
2
|
Tai GJ, Ma YJ, Feng JL, Li JP, Qiu S, Yu QQ, Liu RH, Wankumbu SC, Wang X, Li XX, Xu M. NLRP3 inflammasome-mediated premature immunosenescence drives diabetic vascular aging dependent on the induction of perivascular adipose tissue dysfunction. Cardiovasc Res 2025; 121:77-96. [PMID: 38643484 DOI: 10.1093/cvr/cvae079] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Revised: 11/29/2023] [Accepted: 02/06/2024] [Indexed: 04/23/2024] Open
Abstract
AIMS The vascular aging process accelerated by type 2 diabetes mellitus (T2DM) is responsible for the elevated risk of associated cardiovascular diseases. Metabolic disorder-induced immune senescence has been implicated in multi-organ/tissue damage. Herein, we sought to determine the role of immunosenescence in diabetic vascular aging and to investigate the underlying mechanisms. METHODS AND RESULTS Aging hallmarks of the immune system appear prior to the vasculature in streptozotocin (STZ)/high-fat diet (HFD)-induced T2DM mice or db/db mice. Transplantation of aged splenocytes or diabetic splenocytes into young mice triggered vascular senescence and injury compared with normal control splenocyte transfer. RNA sequencing profile and validation in immune tissues revealed that the toll-like receptor 4-nuclear factor-kappa B-NLRP3 axis might be the mediator of diabetic premature immunosenescence. The absence of Nlrp3 attenuated immune senescence and vascular aging during T2DM. Importantly, senescent immune cells, particularly T cells, provoked perivascular adipose tissue (PVAT) dysfunction and alternations in its secretome, which in turn impair vascular biology. In addition, senescent immune cells may uniquely affect vasoconstriction via influencing PVAT. Lastly, rapamycin alleviated diabetic immune senescence and vascular aging, which may be partly due to NLRP3 signalling inhibition. CONCLUSION These results indicated that NLRP3 inflammasome-mediated immunosenescence precedes and drives diabetic vascular aging. The contribution of senescent immune cells to vascular aging is a combined effect of their direct effects and induction of PVAT dysfunction, the latter of which can uniquely affect vasoconstriction. We further demonstrated that infiltration of senescent T cells in PVAT was increased and associated with PVAT secretome alterations. Our findings suggest that blocking the NLRP3 pathway may prevent early immunosenescence and thus mitigate diabetic vascular aging and damage, and targeting senescent T cells or PVAT might also be the potential therapeutic approach.
Collapse
MESH Headings
- Animals
- NLR Family, Pyrin Domain-Containing 3 Protein/metabolism
- NLR Family, Pyrin Domain-Containing 3 Protein/genetics
- NLR Family, Pyrin Domain-Containing 3 Protein/deficiency
- Inflammasomes/metabolism
- Inflammasomes/genetics
- Inflammasomes/immunology
- Signal Transduction
- Diabetes Mellitus, Experimental/metabolism
- Diabetes Mellitus, Experimental/immunology
- Diabetes Mellitus, Experimental/genetics
- Diabetes Mellitus, Experimental/physiopathology
- Diabetes Mellitus, Experimental/pathology
- Male
- Adipose Tissue/metabolism
- Adipose Tissue/immunology
- Adipose Tissue/physiopathology
- Adipose Tissue/pathology
- Mice, Inbred C57BL
- Diabetes Mellitus, Type 2/metabolism
- Diabetes Mellitus, Type 2/immunology
- Diabetes Mellitus, Type 2/genetics
- Diabetes Mellitus, Type 2/physiopathology
- Diabetes Mellitus, Type 2/pathology
- Immunosenescence
- Diabetic Angiopathies/metabolism
- Diabetic Angiopathies/immunology
- Diabetic Angiopathies/physiopathology
- Diabetic Angiopathies/genetics
- Diabetic Angiopathies/pathology
- Diabetic Angiopathies/prevention & control
- Cellular Senescence
- Mice, Knockout
- Vasoconstriction
- T-Lymphocytes/metabolism
- T-Lymphocytes/immunology
- T-Lymphocytes/transplantation
- T-Lymphocytes/pathology
- NF-kappa B/metabolism
- Mice
- Spleen/metabolism
- Spleen/transplantation
- Toll-Like Receptor 4
Collapse
Affiliation(s)
- Guang-Jie Tai
- Department of Clinical Pharmacy, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, 24 Tong jia Lane, Nanjing 210009, China
| | - Yan-Jie Ma
- Department of Clinical Pharmacy, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, 24 Tong jia Lane, Nanjing 210009, China
| | - Jun-Lin Feng
- Department of Clinical Pharmacy, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, 24 Tong jia Lane, Nanjing 210009, China
| | - Jia-Peng Li
- Department of Clinical Pharmacy, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, 24 Tong jia Lane, Nanjing 210009, China
| | - Shu Qiu
- Department of Clinical Pharmacy, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, 24 Tong jia Lane, Nanjing 210009, China
| | - Qing-Qing Yu
- Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, Nanjing, China
| | - Ren-Hua Liu
- Department of Clinical Pharmacy, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, 24 Tong jia Lane, Nanjing 210009, China
| | - Silumbwe Ceaser Wankumbu
- Department of Clinical Pharmacy, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, 24 Tong jia Lane, Nanjing 210009, China
| | - Xin Wang
- Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, UK
| | - Xiao-Xue Li
- Department of Cardiology, Zhongda Hospital, School of Medicine, Southeast University, No. 87 Dingjiaqiao, Nanjing 210009, China
| | - Ming Xu
- Department of Clinical Pharmacy, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, 24 Tong jia Lane, Nanjing 210009, China
| |
Collapse
|
3
|
Cao P, Chen S, Wang H, Chen Y. Taraxasterol mediated autophagy inhibition in pancreatic encephalopathy involves its regulation on L1 cell adhesion molecule. Cytotechnology 2025; 77:72. [PMID: 40051886 PMCID: PMC11880456 DOI: 10.1007/s10616-025-00721-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2024] [Accepted: 01/27/2025] [Indexed: 03/09/2025] Open
Abstract
Pancreatic encephalopathy (PE) is a frequent complication of acute pancreatitis. This study explored the mechanism of taraxasterol (TAS) in PE treatment by inhibiting pyroptosis via L1 cell adhesion molecule (L1CAM) up-regulation. PE rat models were established and treated with TAS, NLRP3 activator, and sh-L1CAM lentivirus. Serum amylase and lipase activities and Serum, hippocampus, and amygdala IL-18 and IL-1β levels were determined by ELISA, followed by TUNEL and HE staining. Rat nerve injury was evaluated by modified Neurological Severity Score (mNSS). Spontaneous behaviors, learning, memory, and emotions in rats were separately assessed by open field, new object recognition, tail suspension, and forced swimming tests. Microstructures of hippocampal CA1 region and amygdala were observed. NLRP3 + GSDMD + cells, pyroptosis markers, L1CAM, and myelin basic protein (MBP) were detected. PE rat model displayed elevated serum amylase and lipase activities and IL-18 and IL-1β levels, increased mNSS, shortened moving distance, reduced discrimination rate, prolonged immobility time, pathological damage in hippocampal CA1 region and amygdala, increased TUNEL-positive and NLRP3 + GSDMD + cells, raised NLRP3, cleaved caspase-1, GSDMD-N, IL-1β and IL-18 levels, and reduced L1CAM and MBP levels. TAS mitigated behavioral deficits and brain injury and curbed NLRP3-mediated pyroptosis in hippocampal CA1 region and amygdala in PE rats. NLRP3 activation partly averted the beneficial impacts of TAS on PE rats. TAS suppressed nerve cell pyroptosis and facilitated myelin regeneration by up-regulating L1CAM. L1CAM silencing partially abrogated TAS's effect on behavioral deficits and brain injury in PE rats. TAS treated PE by inhibiting pyroptosis via L1CAM up-regulation. Supplementary Information The online version contains supplementary material available at 10.1007/s10616-025-00721-x.
Collapse
Affiliation(s)
- Peng Cao
- Department of Hepatopancreatobiliary Surgery, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, China
| | - Shuangxi Chen
- Department of Neurology, The First Affiliated Hospital, Hengyang Medical School, University of South China, No.69, Chuanshan Road, Shigu District, Hengyang, 421001 Hunan Province China
| | - Huiqing Wang
- Department of Hepatopancreatobiliary Surgery, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, China
| | - Yanfang Chen
- Department of Neurology, The First Affiliated Hospital, Hengyang Medical School, University of South China, No.69, Chuanshan Road, Shigu District, Hengyang, 421001 Hunan Province China
| |
Collapse
|
4
|
Liu Q, Wu Z, Yu C, Qi X, Fang H, Yu X, Li L, Bai Y, Liu D, Chen Z, Kai G, Liang C. Identification and characterization of the TmSnRK2 family proteins related to chicoric acid biosynthesis in Taraxacum mongolicum. BMC Genomics 2025; 26:276. [PMID: 40114043 PMCID: PMC11927344 DOI: 10.1186/s12864-025-11460-w] [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: 09/20/2023] [Accepted: 03/07/2025] [Indexed: 03/22/2025] Open
Abstract
BACKGROUND Taraxacum mongolicum is rich in phenolic acids and is widely utilized in food and medicine globally. Our previous research demonstrated that the abscisic acid (ABA) hormone significantly enhances chicoric acid accumulation in T. mongolicum. SNF1-related protein kinase 2s (SnRK2s) are extensively involved in ABA signaling and have the potential to regulate the biosynthesis of phenolic acids. RESULTS In this study, liquid chromatography-mass spectrometry (LC-MS) and transcriptomic analyses revealed that the TmbZIP1-Tm4CL1 pathway plays a crucial role in the transcriptional regulation of chicoric acid biosynthesis. Seven TmSnRK2s were identified in T. mongolicum and classified into three groups. Analysis of the TmSnRK2s promoters (2000 bp in length) indicated that the three most prevalent stress-related elements were ABA, methyl jasmonate (MeJA), and light. ABA treatments (0 h, 2 h, 4 h, 8 h, and 24 h) showed that all seven TmSnRK2s were significantly modulated by ABA, with the exception of SnRK2.7. TmSnRK2.2, TmSnRK2.3, TmSnRK2.6, and TmSnRK2.7 were localized in both the cytoplasm and nucleus, whereas TmSnRK2.1 and TmSnRK2.5 were exclusively observed in the cytoplasm. Yeast two-hybrid (Y2H) and bimolecular fluorescence complementation (BiFC) assays indicated that TmSnRK2.1, TmSnRK2.3, TmSnRK2.6, and TmSnRK2.7 interact with TmbZIP1. The motifs 'Q(S/G)(V/D)(D/E)(I/L)××I(I/V)×EA' and 'D×(D/ED××D)' are identified as the core sites that facilitate the binding of TmSnRK2s to TmbZIP1. Dual-luciferase reporter assays demonstrated that TmSnRK2.3 and TmSnRK2.6 enhance the stability of TmbZIP1 binding to proTm4CL1. CONCLUSION These findings enhance our understanding of the specific roles of certain members of the TmSnRK2 family in the biosynthesis pathway of chicoric acid.
Collapse
Affiliation(s)
- Qun Liu
- Institute of Botany, Jiangsu Province and Chinese Academy of Sciences (Nanjing Botanical Garden Mem. Sun Yat-Sen), Nanjing, 210014, China
- Jinhua Academy, Zhejiang International Science and Technology Cooperation Base for Active Ingredients of Medicinal and Edible Plants and Health, College of Pharmacy, Zhejiang Chinese Medical University, Hangzhou, 310053, Zhejiang, China
- Jiangsu Key Laboratory for the Research and Utilization of Plant Resources, Nanjing, 210014, China
| | - Zhiqing Wu
- Institute of Botany, Jiangsu Province and Chinese Academy of Sciences (Nanjing Botanical Garden Mem. Sun Yat-Sen), Nanjing, 210014, China
- Jiangsu Key Laboratory for the Research and Utilization of Plant Resources, Nanjing, 210014, China
| | - Changyang Yu
- Institute of Botany, Jiangsu Province and Chinese Academy of Sciences (Nanjing Botanical Garden Mem. Sun Yat-Sen), Nanjing, 210014, China
- Jiangsu Key Laboratory for the Research and Utilization of Plant Resources, Nanjing, 210014, China
| | - Xiwu Qi
- Institute of Botany, Jiangsu Province and Chinese Academy of Sciences (Nanjing Botanical Garden Mem. Sun Yat-Sen), Nanjing, 210014, China
- Jiangsu Key Laboratory for the Research and Utilization of Plant Resources, Nanjing, 210014, China
| | - Hailing Fang
- Institute of Botany, Jiangsu Province and Chinese Academy of Sciences (Nanjing Botanical Garden Mem. Sun Yat-Sen), Nanjing, 210014, China
- Jiangsu Key Laboratory for the Research and Utilization of Plant Resources, Nanjing, 210014, China
| | - Xu Yu
- Institute of Botany, Jiangsu Province and Chinese Academy of Sciences (Nanjing Botanical Garden Mem. Sun Yat-Sen), Nanjing, 210014, China
- Jiangsu Key Laboratory for the Research and Utilization of Plant Resources, Nanjing, 210014, China
| | - Li Li
- Institute of Botany, Jiangsu Province and Chinese Academy of Sciences (Nanjing Botanical Garden Mem. Sun Yat-Sen), Nanjing, 210014, China
- Jiangsu Key Laboratory for the Research and Utilization of Plant Resources, Nanjing, 210014, China
| | - Yang Bai
- Institute of Botany, Jiangsu Province and Chinese Academy of Sciences (Nanjing Botanical Garden Mem. Sun Yat-Sen), Nanjing, 210014, China
- Jiangsu Key Laboratory for the Research and Utilization of Plant Resources, Nanjing, 210014, China
| | - Dongmei Liu
- Institute of Botany, Jiangsu Province and Chinese Academy of Sciences (Nanjing Botanical Garden Mem. Sun Yat-Sen), Nanjing, 210014, China
- Jiangsu Key Laboratory for the Research and Utilization of Plant Resources, Nanjing, 210014, China
| | - Zequn Chen
- Institute of Botany, Jiangsu Province and Chinese Academy of Sciences (Nanjing Botanical Garden Mem. Sun Yat-Sen), Nanjing, 210014, China
- Jiangsu Key Laboratory for the Research and Utilization of Plant Resources, Nanjing, 210014, China
| | - Guoyin Kai
- Jinhua Academy, Zhejiang International Science and Technology Cooperation Base for Active Ingredients of Medicinal and Edible Plants and Health, College of Pharmacy, Zhejiang Chinese Medical University, Hangzhou, 310053, Zhejiang, China.
| | - Chengyuan Liang
- Institute of Botany, Jiangsu Province and Chinese Academy of Sciences (Nanjing Botanical Garden Mem. Sun Yat-Sen), Nanjing, 210014, China.
- Jiangsu Key Laboratory for the Research and Utilization of Plant Resources, Nanjing, 210014, China.
| |
Collapse
|
5
|
Shackebaei D, Yari K, Rahimi N, Gorgani S, Yarmohammadi F. Targeting the NLRP3 by Natural Compounds: Therapeutic Strategies to Mitigate Doxorubicin-Induced Cardiotoxicity. Cell Biochem Biophys 2025:10.1007/s12013-025-01723-4. [PMID: 40100343 DOI: 10.1007/s12013-025-01723-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/08/2025] [Indexed: 03/20/2025]
Abstract
Doxorubicin (DOX), a widely utilized anthracycline chemotherapy agent, is known for its potent anticancer efficacy across various malignancies. However, its clinical use is considerably restricted due to the risk of dose-dependent cardiotoxicity, which can lead to long-term heart dysfunction. The underlying mechanism of DOX-induced cardiotoxicity has been associated with the formation of reactive oxygen species (ROS) and disrupting cellular signaling pathways. This is particularly relevant to the activation of the NLRP3 inflammasome, which triggers inflammation and pyroptosis in cardiac cells. In recent years, there has been growing interest in natural compounds that exhibit potential cardioprotective effects against the adverse cardiac effects of DOX. The present study showed that specific natural compounds, such as honokiol, resveratrol, cynaroside, and curcumin, can confer significant protection against DOX-induced cardiotoxicity through the modulation of NLRP3 inflammasome signaling pathways. In summary, incorporating natural compounds into treatment plans could be a practical approach to improve the safety profile of DOX, thereby protecting cardiac health through the regulation of the NLRP3 pathway.
Collapse
Affiliation(s)
- Dareuosh Shackebaei
- Medical Biology Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
- Department of Physiology, School of Medicine, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Kheirollah Yari
- Medical Biology Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Nader Rahimi
- Department of Occupational Health and Safety Engineering, Faculty of Health, Ilam University of Medical Sciences, Ilam, Iran
| | - Sara Gorgani
- Neuroscience Research Center, Department of Anatomy, Faculty of Medicine, Golestan University of Medical Sciences, Gorgan, Iran
| | - Fatemeh Yarmohammadi
- Medical Biology Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran.
| |
Collapse
|
6
|
Yang B, Qi Z, Armas Diaz Y, Cassotta M, Grosso G, Cianciosi D, Zhang D, Zou X, Quiles JL, Battino M, Giampieri F. Pyroptosis: A Novel Therapeutic Target for Bioactive Compounds in Human Disease Treatment? A Narrative Review. Nutrients 2025; 17:461. [PMID: 39940319 PMCID: PMC11820709 DOI: 10.3390/nu17030461] [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: 12/31/2024] [Revised: 01/16/2025] [Accepted: 01/25/2025] [Indexed: 02/14/2025] Open
Abstract
Background/Objectives: Bioactive compounds possess the ability to maintain health and improve diseases by regulating inflammation and cell death processes. Pyroptosis is programmed cell death related to inflammation and exerts a critical role in the development and progression of different types of diseases. This narrative review aims to investigate and discuss the effects of dietary bioactive compounds on pyroptosis in different common human pathologies, such as inflammatory disease, bacterial infection, injury disease, cancer, diabetes and heart disease, etc. Method: Studies published in the major databases until December 2024 in English were considered, for a total of 50 papers. Results: The current evidence demonstrated that the bioactive compounds are able to regulate the pyroptosis process by modulating different inflammasome sensors (NLRP1, NLRP3, and AIM2), caspase family proteins (caspase-1, caspase-3, and caspase-11), and gasdermins (GSDMD and GSDME) in many pathological conditions related to inflammation, including cancer and cardiovascular diseases. Conclusions: Bioactive compounds have powerful potential to be the candidate drug for pyroptosis modulation in inflammatory diseases, even if more clinical studies are needed to confirm the effects and establish efficient doses for humans.
Collapse
Affiliation(s)
- Bei Yang
- Joint Laboratory on Food Science, Nutrition, and Intelligent Processing of Foods, Polytechnic University of Marche, Italy, Universidad Europea del Atlántico Spain and Jiangsu University, China at Polytechnic University of Marche, 60130 Ancona, Italy
- Dipartimento di Scienze Cliniche Specialistiche e Odontostomatologiche, Università Politecnica delle Marche, Via Ranieri 65, 60130 Ancona, Italy
| | - Zexiu Qi
- Joint Laboratory on Food Science, Nutrition, and Intelligent Processing of Foods, Polytechnic University of Marche, Italy, Universidad Europea del Atlántico Spain and Jiangsu University, China at Polytechnic University of Marche, 60130 Ancona, Italy
- Dipartimento di Scienze Cliniche Specialistiche e Odontostomatologiche, Università Politecnica delle Marche, Via Ranieri 65, 60130 Ancona, Italy
| | - Yasmany Armas Diaz
- Joint Laboratory on Food Science, Nutrition, and Intelligent Processing of Foods, Polytechnic University of Marche, Italy, Universidad Europea del Atlántico Spain and Jiangsu University, China at Polytechnic University of Marche, 60130 Ancona, Italy
- Dipartimento di Scienze Cliniche Specialistiche e Odontostomatologiche, Università Politecnica delle Marche, Via Ranieri 65, 60130 Ancona, Italy
| | - Manuela Cassotta
- Joint Laboratory on Food Science, Nutrition, and Intelligent Processing of Foods, Polytechnic University of Marche, Italy, Universidad Europea del Atlántico Spain and Jiangsu University, China at Polytechnic University of Marche, 60130 Ancona, Italy
- Research Group on Food, Nutritional Biochemistry and Health, Universidad Europea del Atlántico, Isabel Torres 21, 39011 Santander, Spain
| | - Giuseppe Grosso
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania, Italy
- Center for Human Nutrition and Mediterranean Foods (NUTREA), University of Catania, 95123 Catania, Italy
| | - Danila Cianciosi
- Joint Laboratory on Food Science, Nutrition, and Intelligent Processing of Foods, Polytechnic University of Marche, Italy, Universidad Europea del Atlántico Spain and Jiangsu University, China at Polytechnic University of Marche, 60130 Ancona, Italy
- Dipartimento di Scienze Cliniche Specialistiche e Odontostomatologiche, Università Politecnica delle Marche, Via Ranieri 65, 60130 Ancona, Italy
| | - Di Zhang
- Joint Laboratory on Food Science, Nutrition, and Intelligent Processing of Foods, Polytechnic University of Marche, Italy, Universidad Europea del Atlántico Spain and Jiangsu University, China at Jiangsu University, Zhenjiang 212013, China
- School of Food and Biological Engineering, Jiangsu University, Zhenjiang 212013, China
| | - Xiaobo Zou
- Joint Laboratory on Food Science, Nutrition, and Intelligent Processing of Foods, Polytechnic University of Marche, Italy, Universidad Europea del Atlántico Spain and Jiangsu University, China at Jiangsu University, Zhenjiang 212013, China
- School of Food and Biological Engineering, Jiangsu University, Zhenjiang 212013, China
| | - José L. Quiles
- Department of Physiology, Institute of Nutrition and Food Technology “Jose Mataix”, Biomedical Research Center, University of Granada, 18016 Granada, Spain
| | - Maurizio Battino
- Joint Laboratory on Food Science, Nutrition, and Intelligent Processing of Foods, Polytechnic University of Marche, Italy, Universidad Europea del Atlántico Spain and Jiangsu University, China at Polytechnic University of Marche, 60130 Ancona, Italy
- Dipartimento di Scienze Cliniche Specialistiche e Odontostomatologiche, Università Politecnica delle Marche, Via Ranieri 65, 60130 Ancona, Italy
- Research Group on Food, Nutritional Biochemistry and Health, Universidad Europea del Atlántico, Isabel Torres 21, 39011 Santander, Spain
- International Joint Research Laboratory of Intelligent Agriculture and Agri-Products Processing, Jiangsu University, Zhenjiang 212013, China
| | - Francesca Giampieri
- Joint Laboratory on Food Science, Nutrition, and Intelligent Processing of Foods, Polytechnic University of Marche, Italy, Universidad Europea del Atlántico Spain and Jiangsu University, China at Polytechnic University of Marche, 60130 Ancona, Italy
- Dipartimento di Scienze Cliniche Specialistiche e Odontostomatologiche, Università Politecnica delle Marche, Via Ranieri 65, 60130 Ancona, Italy
- Research Group on Food, Nutritional Biochemistry and Health, Universidad Europea del Atlántico, Isabel Torres 21, 39011 Santander, Spain
- International Research Center for Food Nutrition and Safety, Jiangsu University, Zhenjiang 212013, China
| |
Collapse
|
7
|
Li J, Ma W, Tang Z, Li Y, Zheng R, Xie Y, Li G. Macrophage‑driven pathogenesis in acute lung injury/acute respiratory disease syndrome: Harnessing natural products for therapeutic interventions (Review). Mol Med Rep 2025; 31:16. [PMID: 39513609 PMCID: PMC11551695 DOI: 10.3892/mmr.2024.13381] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Accepted: 09/27/2024] [Indexed: 11/15/2024] Open
Abstract
Acute lung injury (ALI) or acute respiratory distress syndrome (ARDS) is a common respiratory disease characterized by hypoxemia and respiratory distress. It is associated with high morbidity and mortality. Due to the complex pathogenesis of ALI, the clinical management of patients with ALI/ARDS is challenging, resulting in numerous post‑treatment sequelae and compromising the quality of life of patients. Macrophages, as a class of innate immune cells, play an important role in ALI/ARDS. In recent years, the functions and phenotypes of macrophages have been better understood due to the development of flow cytometry, immunofluorescence, single‑cell sequencing and spatial genomics. However, no macrophage‑targeted drugs for the treatment of ALI/ARDS currently exist in clinical practice. Natural products are important for drug development, and it has been shown that numerous natural compounds from herbal medicine can alleviate ALI/ARDS caused by various factors by modulating macrophage abnormalities. In the present review, the natural products from herbal medicine that can modulate macrophage abnormalities in ALI/ARDS to treat ALI/ARDS are introduced, and their mechanisms of action, discovered in the previous five years (2019‑2024), are presented. This will provide novel ideas and directions for further research, to develop new drugs for the treatment of ALI/ARDS.
Collapse
Affiliation(s)
- Jincun Li
- College of Traditional Chinese Medicine, Yunnan University of Chinese Medicine, Kunming, Yunnan 650500, P.R. China
| | - Wenyu Ma
- College of Traditional Chinese Medicine, Yunnan University of Chinese Medicine, Kunming, Yunnan 650500, P.R. China
| | - Zilei Tang
- College of Traditional Chinese Medicine, Yunnan University of Chinese Medicine, Kunming, Yunnan 650500, P.R. China
| | - Yingming Li
- College of Traditional Chinese Medicine, Yunnan University of Chinese Medicine, Kunming, Yunnan 650500, P.R. China
| | - Ruiyu Zheng
- College of Traditional Chinese Medicine, Yunnan University of Chinese Medicine, Kunming, Yunnan 650500, P.R. China
| | - Yuhuan Xie
- Yunnan Innovation Team of Application Research on Traditional Chinese Medicine Theory of Disease Prevention, Yunnan University of Chinese Medicine, Kunming, Yunnan 650500, P.R. China
- Yunnan Provincial University Key Laboratory of Aromatic Chinese Herb Research, Basic Medical School, Yunnan University of Chinese Medicine, Kunming, Yunnan 650500, P.R. China
| | - Gang Li
- Yunnan Provincial University Key Laboratory of Aromatic Chinese Herb Research, Basic Medical School, Yunnan University of Chinese Medicine, Kunming, Yunnan 650500, P.R. China
- Basic Medical School, Yunnan University of Chinese Medicine, Kunming, Yunnan 650500, P.R. China
| |
Collapse
|
8
|
Wang X, Liu Y, Zhou Y, Li M, Mo T, Xu X, Chen Z, Zhang Y, Yang L. mTORC2 knockdown mediates lipid metabolism to alleviate hyperlipidemic pancreatitis through PPARα. J Biochem Mol Toxicol 2024; 38:e23802. [PMID: 39132808 DOI: 10.1002/jbt.23802] [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: 07/04/2024] [Accepted: 07/30/2024] [Indexed: 08/13/2024]
Abstract
Hyperlipidemic pancreatitis (HP) is an inflammatory injury of the pancreas triggered by elevated serum triglyceride (TG) levels. The mechanistic target of rapamycin (mTOR) signaling pathway plays a crucial role in regulating lipid homeostasis and inflammation. This study aimed to investigate whether the activity of mTOR complex 2 (mTORC2) affects the progression of HP and its underlying mechanisms. In vivo, a high-fat diet and retrograde administration of sodium taurocholate were employed to establish the HP models in rats, with pancreatic tissue pathology evaluated. The expression of Rictor and peroxisome proliferator-activator receptor (PPAR) was examined. The serum levels of TG, fatty acid metabolites, inflammatory and lipid metabolism-related factors were determined. In vitro, pancreatic acinar cells (PACs) were exposed to palmitic acid and cholecystokinin-8. PAC apoptosis, pyroptosis, and ferroptosis were assessed. In the HP models, rats and PACs exhibited upregulated Rictor and downregulated PPARα, and Rictor knockdown promoted PPARα expression. In vivo, Rictor knockdown decreased the serum levels of TG, α-amylase, total cholesterol, low-density lipoprotein cholesterol, lactate dehydrogenase, and inflammatory factors, while increasing high-density lipoprotein cholesterol levels. Rictor knockdown increased ACOX1 and CPT1α and decreased SREBP-1, CD36, SCD1, ACLY, and ACACA. Rictor knockdown reduced damage to pancreatic tissue structure. In vitro, Rictor knockdown inhibited PAC apoptosis, pyroptosis, and ferroptosis. Treatment with the PPARα antagonist GW6471 abolished the beneficial effects of Rictor knockdown. Rictor/mTORC2 deficiency reduces serum TG levels, maintains lipid homeostasis, and suppresses inflammation by inhibiting PPARα expression. Weakening mTORC2 activity holds promise as a novel therapeutic strategy for HP.
Collapse
Affiliation(s)
- Xiangyang Wang
- Department of Gastroenterology, Hunan Provincial People's Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha, China
| | - Yilei Liu
- Department of Gastroenterology, Hunan Provincial People's Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha, China
| | - Yaxiong Zhou
- Department of Gastroenterology, Hunan Provincial People's Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha, China
| | - Min Li
- Department of Gastroenterology, Hunan Provincial People's Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha, China
| | - Tingting Mo
- Department of Gastroenterology, Hunan Provincial People's Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha, China
| | - Xiaoping Xu
- Department of Gastroenterology, Hunan Provincial People's Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha, China
| | - Zhiyuan Chen
- Department of Gastroenterology, Hunan Provincial People's Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha, China
| | - Yu Zhang
- Department of Gastroenterology, Hunan Provincial People's Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha, China
| | - Li Yang
- Department of Gastroenterology, Hunan Provincial People's Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha, China
| |
Collapse
|
9
|
Qi W, Jin L, Huang S, Aikebaier A, Xue S, Wang Q, Chen Q, Lu Y, Ding C. Modulating synovial macrophage pyroptosis and mitophagy interactions to mitigate osteoarthritis progression using functionalized nanoparticles. Acta Biomater 2024; 181:425-439. [PMID: 38729544 DOI: 10.1016/j.actbio.2024.05.014] [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: 02/04/2024] [Revised: 04/29/2024] [Accepted: 05/05/2024] [Indexed: 05/12/2024]
Abstract
Synovial macrophages play an important role in the progression of osteoarthritis (OA). In this study, we noted that synovial macrophages can activate pyroptosis in a gasdermin d-dependent manner and produce reactive oxygen species (ROS), aberrantly activating the mammalian target of rapamycin complex 1 (mTORC1) pathway and matrix metalloproteinase-9 (MMP9) expression in synovial tissue samples collected from both patients with OA and collagen-induced osteoarthritis (CIOA) mouse model. To overcome this, we constructed rapamycin- (RAPA, a mTORC1 inhibitor) loaded mesoporous Prussian blue nanoparticles (MPB NPs, for catalyzing ROS) and modified the NPs with MMP9-targeted peptides (favor macrophage targeting) to develop RAPA@MPB-MMP9 NPs. The inherent enzyme-like activity and RAPA released from RAPA@MPB-MMP9 NPs synergistically impeded the pyroptosis of macrophages and the activation of the mTORC1 pathway. In particular, the NPs decreased pyroptosis-mediated ROS generation, thereby inhibiting cGAS-STING signaling pathway activation caused by the release of mitochondrial DNA. Moreover, the NPs promoted macrophage mitophagy to restore mitochondrial stability, alleviate pyroptosis-related inflammatory responses, and decrease senescent synoviocytes. After the as-prepared NPs were intra-articularly injected into the CIOA mouse model, they efficiently attenuated synovial macrophage pyroptosis and cartilage degradation. In conclusion, our study findings provide a novel therapeutic strategy for OA that modulates the pyroptosis and mitophagy of synovial macrophage by utilizing functionalized NPs. STATEMENT OF SIGNIFICANCE: Osteoarthritis (OA) presents a significant global challenge owing to its complex pathogenesis and finite treatment options. Synovial macrophages have emerged as key players in the progression of OA, managing inflammation and tissue destruction. In this study, we discovered a novel therapeutic strategy in which the pyroptosis and mitophagy of synovial macrophages are targeted to mitigate OA pathology. For this, we designed and prepared rapamycin-loaded mesoporous Prussian blue nanoparticles (RAPA@MPB-MMP9 NPs) to specifically target synovial macrophages and modulate their inflammatory responses. These NPs could efficiently suppress macrophage pyroptosis, diminish reactive oxygen species production, and promote mitophagy, thereby alleviating inflammation and protecting cartilage integrity. Our study findings not only clarify the intricate mechanisms underlying OA pathogenesis but also present a promising therapeutic approach for effectively managing OA by targeting dysregulation in synovial macrophages.
Collapse
Affiliation(s)
- Weizhong Qi
- Clinical Research Centre, The Second School of Clinical Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, 510282, China
| | - Li Jin
- Rheumatology and Clinical Immunology, ZhuJiang Hospital, Southern Medical University, Guangzhou, 510282, China
| | - Shiqian Huang
- Clinical Research Centre, The Second School of Clinical Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, 510282, China
| | - Alafate Aikebaier
- Clinical Research Centre, The Second School of Clinical Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, 510282, China
| | - Song Xue
- Clinical Research Centre, The Second School of Clinical Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, 510282, China
| | - QianYi Wang
- Clinical Research Centre, The Second School of Clinical Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, 510282, China
| | - Qiyue Chen
- Stomatological Hospital, Southern Medical University, Guangzhou, 510282, China.
| | - Yao Lu
- Clinical Research Centre, The Second School of Clinical Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, 510282, China; Department of Joint and Orthopedics, Orthopedic Center, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong 510282, China.
| | - Changhai Ding
- Clinical Research Centre, The Second School of Clinical Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, 510282, China; Menzies Institute for Medical Research, University of Tasmania, 7000, Hobart, Tasmania, Australia.
| |
Collapse
|
10
|
Shan M, Wan H, Ran L, Ye J, Xie W, Lu J, Hu X, Deng S, Zhang W, Chen M, Wang F, Guo Z. Dynasore Alleviates LPS-Induced Acute Lung Injury by Inhibiting NLRP3 Inflammasome-Mediated Pyroptosis. Drug Des Devel Ther 2024; 18:1369-1384. [PMID: 38681210 PMCID: PMC11055558 DOI: 10.2147/dddt.s444408] [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: 10/12/2023] [Accepted: 04/12/2024] [Indexed: 05/01/2024] Open
Abstract
Background Acute lung injury (ALI) and acute respiratory distress syndrome (ARDS) are clinically severe respiratory disorders without available pharmacological therapies. Dynasore is a cell-permeable molecule that inhibits GTPase activity and exerts protective effects in several disease models. However, whether dynasore can alleviate lipopolysaccharide (LPS)-induced ALI is unknown. This study investigated the effect of dynasore on macrophage activation and explored its potential mechanisms in LPS-induced ALI in vitro and in vivo. Methods Bone marrow-derived macrophages (BMDMs) were activated classically with LPS or subjected to NLRP3 inflammasome activation with LPS+ATP. A mouse ALI model was established by the intratracheal instillation (i.t.) of LPS. The expression of PYD domains-containing protein 3 (NLRP3), caspase-1, and gasdermin D (GSDMD) protein was detected by Western blots. Inflammatory mediators were analyzed in the cell supernatant, in serum and bronchoalveolar lavage fluid (BALF) by enzyme-linked immunosorbent assays. Morphological changes in lung tissues were evaluated by hematoxylin and eosin staining. F4/80, Caspase-1 and GSDMD distribution in lung tissue was detected by immunofluorescence. Results Dynasore downregulated nuclear factor (NF)-κB signaling and reduced proinflammatory cytokine production in vitro and inhibited the production and release of interleukin (IL)-1β, NLRP3 inflammasome activation, and macrophage pyroptosis through the Drp1/ROS/NLRP3 axis. Dynasore significantly reduced lung injury scores and proinflammatory cytokine levels in both BALF and serum in vivo, including IL-1β and IL-6. Dynasore also downregulated the co-expression of F4/80, caspase-1 and GSDMD in lung tissue. Conclusion Collectively, these findings demonstrated that dynasore could alleviate LPS-induced ALI by regulating macrophage pyroptosis, which might provide a new therapeutic strategy for ALI/ARDS.
Collapse
Affiliation(s)
- Mengtian Shan
- Department of Pulmonary and Critical Care Medicine, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, People’s Republic of China
| | - Huimin Wan
- Department of Pulmonary and Critical Care Medicine, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, People’s Republic of China
| | - Linyu Ran
- Department of Pulmonary and Critical Care Medicine, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, People’s Republic of China
| | - Jihui Ye
- Department of Pulmonary and Critical Care Medicine, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, People’s Republic of China
| | - Wang Xie
- Department of Respiratory Medicine, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, People’s Republic of China
| | - Jingjing Lu
- Department of Pulmonary and Critical Care Medicine, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, People’s Republic of China
| | - Xueping Hu
- Department of Pulmonary and Critical Care Medicine, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, People’s Republic of China
| | - Shengjie Deng
- Department of Pulmonary and Critical Care Medicine, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, People’s Republic of China
| | - Wenyu Zhang
- Department of Pulmonary and Critical Care Medicine, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, People’s Republic of China
| | - Miao Chen
- Department of Emergency, The First Affiliated Hospital of Hainan Medical University, Haikou, Hainan, People’s Republic of China
| | - Feilong Wang
- Department of Pulmonary and Critical Care Medicine, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, People’s Republic of China
| | - Zhongliang Guo
- Department of Pulmonary and Critical Care Medicine, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, People’s Republic of China
- Department of Respiratory Medicine, Ji’an Hospital, Shanghai East Hospital, Shanghai, Jiangxi, People’s Republic of China
| |
Collapse
|
11
|
Argandona Lopez C, Brown AM. Microglial- neuronal crosstalk in chronic viral infection through mTOR, SPP1/OPN and inflammasome pathway signaling. Front Immunol 2024; 15:1368465. [PMID: 38646526 PMCID: PMC11032048 DOI: 10.3389/fimmu.2024.1368465] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Accepted: 03/25/2024] [Indexed: 04/23/2024] Open
Abstract
HIV-infection of microglia and macrophages (MMs) induces neuronal injury and chronic release of inflammatory stimuli through direct and indirect molecular pathways. A large percentage of people with HIV-associated neurologic and psychiatric co-morbidities have high levels of circulating inflammatory molecules. Microglia, given their susceptibility to HIV infection and long-lived nature, are reservoirs for persistent infection. MMs and neurons possess the molecular machinery to detect pathogen nucleic acids and proteins to activate innate immune signals. Full activation of inflammasome assembly and expression of IL-1β requires a priming event and a second signal. Many studies have demonstrated that HIV infection alone can activate inflammasome activity. Interestingly, secreted phosphoprotein-1 (SPP1/OPN) expression is highly upregulated in the CNS of people infected with HIV and neurologic dysfunction. Interestingly, all evidence thus far suggests a protective function of SPP1 signaling through mammalian target of rapamycin (mTORC1/2) pathway function to counter HIV-neuronal injury. Moreover, HIV-infected mice knocked down for SPP1 show by neuroimaging, increased neuroinflammation compared to controls. This suggests that SPP1 uses unique regulatory mechanisms to control the level of inflammatory signaling. In this mini review, we discuss the known and yet-to-be discovered biological links between SPP1-mediated stimulation of mTOR and inflammasome activity. Additional new mechanistic insights from studies in relevant experimental models will provide a greater understanding of crosstalk between microglia and neurons in the regulation of CNS homeostasis.
Collapse
Affiliation(s)
- Catalina Argandona Lopez
- Division of Neuroimmunology, Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Amanda M. Brown
- Division of Neuroimmunology, Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
- Division of Neuroimmunology, Department of Neurology and Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| |
Collapse
|
12
|
Johri N, Matreja PS, Agarwal S, Nagar P, Kumar D, Maurya A. Unraveling the Molecular Mechanisms of Activated Protein C (APC) in Mitigating Reperfusion Injury and Cardiac Ischemia: a Promising Avenue for Novel Therapeutic Interventions. J Cardiovasc Transl Res 2024; 17:345-355. [PMID: 37851312 DOI: 10.1007/s12265-023-10445-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/04/2023] [Accepted: 10/02/2023] [Indexed: 10/19/2023]
Abstract
Ischemic heart disease, which results from plaque formation in the coronary arteries, hinders the flow of oxygenated blood to the heart, leading to ischemia. Reperfusion injury remains a significant challenge for researchers, and the mechanisms underlying myocardial ischemia-reperfusion injury (MIRI) are not entirely understood. The review directs future research into potential targets in clinical treatment based on our present understanding of the pathophysiological mechanisms of MIRI. The study provides insights into the mechanisms underlying MIRI and offers direction for future research in this area. The use of targeted therapies may hold promise in improving cardiac function in the elderly and minimizing the adverse effects of revascularization therapies. The purpose of this review is to analyze the role of activated protein C (APC) in the pathogenesis of ischemic heart disease, heart failure, and myocardial ischemia-reperfusion injury, and discuss the potential of APC-based therapeutics.
Collapse
Affiliation(s)
- Nishant Johri
- Department of Pharmacy Practice & Pharmacology, Teerthanker Mahaveer College of Pharmacy, Moradabad, Uttar Pradesh, India.
- School of Health & Psychological Sciences, City, University of London, London, United Kingdom.
| | - Prithpal S Matreja
- Department of Pharmacology, Teerthanker Mahaveer Medical College and Research Centre, Moradabad, Uttar Pradesh, India
| | - Shalabh Agarwal
- Department of Cardiology, Teerthanker Mahaveer Hospital & Research Centre, Moradabad, Uttar Pradesh, India
| | - Priya Nagar
- Department of Pharmacy Practice & Pharmacology, Teerthanker Mahaveer College of Pharmacy, Moradabad, Uttar Pradesh, India
| | - Deepanshu Kumar
- Department of Pharmacy Practice & Pharmacology, Teerthanker Mahaveer College of Pharmacy, Moradabad, Uttar Pradesh, India
| | - Aditya Maurya
- Department of Pharmacy Practice & Pharmacology, Teerthanker Mahaveer College of Pharmacy, Moradabad, Uttar Pradesh, India
| |
Collapse
|
13
|
Zhang Y, BinShaari R, Nawi MABA, Bin Hassan A, Cui C. Pharmacological Action and Research Progress of Taraxasterol. Curr Pharm Biotechnol 2024; 25:1767-1777. [PMID: 38178677 DOI: 10.2174/0113892010276692231220103636] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Revised: 10/30/2023] [Accepted: 11/10/2023] [Indexed: 01/06/2024]
Abstract
Primarily sourced from Asteraceae family herbs such as the Dandelion, Taraxasterol is a pentacyclic triterpenoid lauded for its extensive biological functionalities. Its therapeutic potency is demonstrated in various disease models, encompassing enteritis, arthritis, acute hepatic injury, and pneumonia. Scientific literature underscores its anti-inflammatory, antioxidant, and antineoplastic attributes. The primary aim of this study is to thoroughly explore the diseasemodulating mechanisms and effects of taraxasterol. We endeavor to provide an exhaustive review of the experimental subjects, intervention components, distinct action modalities, contributing factors, and protein pathway expressions associated with taraxasterol, systematically represented via diagrams and tables. Such a schematic representation encourages a continued academic dialogue concerning taraxasterol's pharmacological characteristics. This review is envisioned as a practical guide for the selection of experimental subjects and methodologies in prospective research. It is intended to further illuminate taraxasterol's pharmacodynamics, thereby offering theoretical and empirical justification for its clinical application.
Collapse
Affiliation(s)
- Yu Zhang
- School of dental sciences, Universiti Sains Malaysia, Health Campus, Kelantan, Malaysia
- Qilu Medical University, Zibo, China
| | - Ramizu BinShaari
- School of Dental Sciences, Universiti Sains Malaysia, Health Campus, Kelantan, Malaysia
| | | | - Akram Bin Hassan
- School of dental sciences, Universiti Sains Malaysia, Health Campus, Kelantan, Malaysia
| | - Caiyun Cui
- Department of Stomatology, Affiliated Hospital of Binzhou Medical University, Binzhou, China
| |
Collapse
|
14
|
Wang S, Wu P, Fan Z, He X, Liu J, Li M, Chen F. Dandelion polysaccharide treatment protects against dextran sodium sulfate-induced colitis by suppressing NF-κB/NLRP3 inflammasome-mediated inflammation and activating Nrf2 in mouse colon. Food Sci Nutr 2023; 11:7271-7282. [PMID: 37970386 PMCID: PMC10630811 DOI: 10.1002/fsn3.3653] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Revised: 07/26/2023] [Accepted: 08/13/2023] [Indexed: 11/17/2023] Open
Abstract
The treatment of ulcerative colitis (UC) is still an intractable medical problem. Polysaccharides are promising candidates for the treatment of UC and have received widespread attention in recent years. The objective of this study was to explore the protective effect and underlying mechanism of dandelion polysaccharide (DP) on dextran sulfate sodium (DSS)-induced colitis in mice. Our results showed that oral administration of DP could dramatically alleviate colonic lesions, as evidenced by reduced DAI scores, shortening of colon length, and ameliorating pathologic abnormalities in colons. Additionally, the expressions of pro-inflammatory factors (TNF-α, IL-1β, and IL-6) and the infiltration of inflammation-regulation cells, marked by myeloperoxidase and F4/80, were also inhibited after DP treatment. Moreover, DP treatment also markedly suppressed the nuclear translocation of NF-κB-p65 and the activation of the NLRP3 inflammasome. Furthermore, DP also activated the Nrf2/HO-1 pathway and reduced the oxidative stress induced by DSS. Overall, these results suggest that DP could be a promising novel therapeutic approach for the treatment of UC.
Collapse
Affiliation(s)
- Shuo Wang
- School of Pharmaceutical SciencesLiaocheng UniversityLiaochengShandongChina
| | - Ping Wu
- School of Pharmaceutical SciencesLiaocheng UniversityLiaochengShandongChina
| | - Zongqiang Fan
- School of Pharmaceutical SciencesLiaocheng UniversityLiaochengShandongChina
| | - Xingrui He
- School of PharmacyHangzhou Normal UniversityHangzhouZhejiangChina
| | - Jinqian Liu
- School of Pharmaceutical SciencesLiaocheng UniversityLiaochengShandongChina
| | - Ming Li
- Shandong Academy of Occupational Health and Occupational MedicineShandong First Medical University & Shandong Academy of Medical SciencesJinanShandongChina
| | - Fang Chen
- School of Pharmaceutical SciencesLiaocheng UniversityLiaochengShandongChina
| |
Collapse
|
15
|
Chen SY, Li YP, You YP, Zhang HR, Shi ZJ, Liang QQ, Yuan T, Xu R, Xu LH, Zha QB, Ou-Yang DY, He XH. Theaflavin mitigates acute gouty peritonitis and septic organ injury in mice by suppressing NLRP3 inflammasome assembly. Acta Pharmacol Sin 2023; 44:2019-2036. [PMID: 37221235 PMCID: PMC10545837 DOI: 10.1038/s41401-023-01105-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Accepted: 05/03/2023] [Indexed: 05/25/2023]
Abstract
Activation of NLR family pyrin domain-containing 3 (NLRP3) inflammasome plays important role in defending against infections, but its aberrant activation is causally linked to many inflammatory diseases, thus being a therapeutic target for these diseases. Theaflavin, one major ingredient of black tea, exhibits potent anti-inflammatory and anti-oxidative activities. In this study, we investigated the therapeutic effects of theaflavin against NLRP3 inflammasome activation in macrophages in vitro and in animal models of related diseases. We showed that theaflavin (50, 100, 200 μM) dose-dependently inhibited NLRP3 inflammasome activation in LPS-primed macrophages stimulated with ATP, nigericin or monosodium urate crystals (MSU), evidenced by reduced release of caspase-1p10 and mature interleukin-1β (IL-1β). Theaflavin treatment also inhibited pyroptosis as shown by decreased generation of N-terminal fragment of gasdermin D (GSDMD-NT) and propidium iodide incorporation. Consistent with these, theaflavin treatment suppressed ASC speck formation and oligomerization in macrophages stimulated with ATP or nigericin, suggesting reduced inflammasome assembly. We revealed that theaflavin-induced inhibition on NLRP3 inflammasome assembly and pyroptosis resulted from ameliorated mitochondrial dysfunction and reduced mitochondrial ROS production, thereby suppressing interaction between NLRP3 and NEK7 downstream of ROS. Moreover, we showed that oral administration of theaflavin significantly attenuated MSU-induced mouse peritonitis and improved the survival of mice with bacterial sepsis. Consistently, theaflavin administration significantly reduced serum levels of inflammatory cytokines including IL-1β and attenuated liver inflammation and renal injury of mice with sepsis, concomitant with reduced generation of caspase-1p10 and GSDMD-NT in the liver and kidney. Together, we demonstrate that theaflavin suppresses NLRP3 inflammasome activation and pyroptosis by protecting mitochondrial function, thus mitigating acute gouty peritonitis and bacterial sepsis in mice, highlighting a potential application in treating NLRP3 inflammasome-related diseases.
Collapse
Affiliation(s)
- Si-Yuan Chen
- Department of Immunobiology, College of Life Science and Technology, Jinan University, Guangzhou, 510632, China
| | - Ya-Ping Li
- Department of Immunobiology, College of Life Science and Technology, Jinan University, Guangzhou, 510632, China
| | - Yi-Ping You
- Department of Immunobiology, College of Life Science and Technology, Jinan University, Guangzhou, 510632, China
| | - Hong-Rui Zhang
- Department of Immunobiology, College of Life Science and Technology, Jinan University, Guangzhou, 510632, China
| | - Zi-Jian Shi
- Department of Fetal Medicine, the First Affiliated Hospital of Jinan University, Guangzhou, 510630, China
| | - Qi-Qi Liang
- Department of Immunobiology, College of Life Science and Technology, Jinan University, Guangzhou, 510632, China
| | - Tao Yuan
- Department of Immunobiology, College of Life Science and Technology, Jinan University, Guangzhou, 510632, China
| | - Rong Xu
- Department of Immunobiology, College of Life Science and Technology, Jinan University, Guangzhou, 510632, China
| | - Li-Hui Xu
- Department of Cell Biology, College of Life Science and Technology, Jinan University, Guangzhou, 510632, China
| | - Qing-Bing Zha
- Department of Fetal Medicine, the First Affiliated Hospital of Jinan University, Guangzhou, 510630, China.
- Department of Clinical Laboratory, the Fifth Affiliated Hospital of Jinan University, Heyuan, 517000, China.
| | - Dong-Yun Ou-Yang
- Department of Immunobiology, College of Life Science and Technology, Jinan University, Guangzhou, 510632, China.
| | - Xian-Hui He
- Department of Immunobiology, College of Life Science and Technology, Jinan University, Guangzhou, 510632, China.
- Department of Clinical Laboratory, the Fifth Affiliated Hospital of Jinan University, Heyuan, 517000, China.
| |
Collapse
|
16
|
Feng M, Wang X, Zhou S, Li M, Liu T, Wei X, Lin W. CD83 + B cells alleviate uveitis through inhibiting DCs by sCD83. Immunology 2023; 170:134-153. [PMID: 37137669 DOI: 10.1111/imm.13654] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Accepted: 04/18/2023] [Indexed: 05/05/2023] Open
Abstract
Soluble CD83 (sCD83) exerts immunosuppressive functions in many autoimmune diseases, including experimental autoimmune uveitis (EAU), but the cells and mechanisms involved are unclear. This study showed that CD83+ B cells were the main sources of sCD83. They alleviated the symptoms of EAU and decreased the percentage of T cells and DCs in the eyes and lymph nodes. These CD83+ B cells decreased IL-1β, IL-18 and IFN-γ secretion by DCs through sCD83. sCD83 interacted with GTPase Ras-related protein (Rab1a) in DCs to promote Rab1a accumulation in autolysosomes and inhibit mTORC1 phosphorylation and NLRP3 expression. Hence, CD83+ B cells play a regulatory role in EAU by secreting sCD83. The lack of regulation of CD83+ B cells might be an important factor leading to hyperimmune activation in patients with autoimmune uveitis. CD83+ B cells suppress activated DCs in uveitis, indicating the potential therapeutic role of CD83+ B cells in uveitis.
Collapse
Affiliation(s)
- Meng Feng
- Department of Rheumatology and Autoimmunology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, China
- School of Clinical and Basic Medicine, Shandong First Medical University &Shandong Academy of Medical Sciences, Jinan, China
- Shandong Key Laboratory of Rheumatic Disease and Translational Medicine, Shandong Medicine and Health Key Laboratory of Rheumatism, The First Affiliated Hospital of Shandong First Medical University, Jinan, China
| | - Xin Wang
- Department of Clinical Laboratory, Qilu Hospital, Shandong University, Jinan, China
- Shandong Engineering Research Center of Biomarker and Artificial Intelligence Application, Jinan, China
| | - Shuping Zhou
- Department of Rheumatology and Autoimmunology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, China
- School of Clinical and Basic Medicine, Shandong First Medical University &Shandong Academy of Medical Sciences, Jinan, China
- Shandong Key Laboratory of Rheumatic Disease and Translational Medicine, Shandong Medicine and Health Key Laboratory of Rheumatism, The First Affiliated Hospital of Shandong First Medical University, Jinan, China
| | - Minghao Li
- Department of Rheumatology and Autoimmunology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, China
- School of Clinical and Basic Medicine, Shandong First Medical University &Shandong Academy of Medical Sciences, Jinan, China
- Shandong Key Laboratory of Rheumatic Disease and Translational Medicine, Shandong Medicine and Health Key Laboratory of Rheumatism, The First Affiliated Hospital of Shandong First Medical University, Jinan, China
| | - Tingting Liu
- Shandong Eye Hospital, State Key Laboratory Cultivation Base, Shandong Provincial Key Laboratory of Ophthalmology, Shandong Eye Institute, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, China
| | - Xunbin Wei
- Biomedical Engineering Department, Peking University, Beijing, China
- School of Biomedical Engineering, Anhui Medical University, Hefei, China
- Institute of Medical Technology, Peking University Health Science Center, Beijing, China
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital & Institute, Beijing, China
- International Cancer Institute, Peking University, Beijing, China
| | - Wei Lin
- Department of Rheumatology and Autoimmunology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, China
- School of Clinical and Basic Medicine, Shandong First Medical University &Shandong Academy of Medical Sciences, Jinan, China
- Shandong Key Laboratory of Rheumatic Disease and Translational Medicine, Shandong Medicine and Health Key Laboratory of Rheumatism, The First Affiliated Hospital of Shandong First Medical University, Jinan, China
| |
Collapse
|
17
|
Feng WD, Wang Y, Luo T, Jia X, Cheng CQ, Wang HJ, Zhang MQ, Li QQ, Wang XJ, Li YY, Wang JY, Huang GR, Wang T, Xu AL. Scoparone suppresses mitophagy-mediated NLRP3 inflammasome activation in inflammatory diseases. Acta Pharmacol Sin 2023; 44:1238-1251. [PMID: 36522512 PMCID: PMC10203299 DOI: 10.1038/s41401-022-01028-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/18/2022] [Accepted: 11/07/2022] [Indexed: 12/23/2022]
Abstract
Recent evidence shows that targeting NLRP3 inflammasome activation is an important means to treat inflammasome-driven diseases. Scoparone, a natural compound isolated from the Chinese herb Artemisia capillaris Thunb, has anti-inflammatory activity. In this study we investigated the effect of scoparone on NLRP3 inflammasome activation in inflammatory diseases. In LPS-primed, ATP or nigericin-stimulated mouse macrophage J774A.1 cells and bone marrow-derived macrophages (BMDMs), pretreatment with scoparone (50 μM) markedly restrained canonical and noncanonical NLRP3 inflammasome activation, evidenced by suppressed caspase-1 cleavage, GSDMD-mediated pyroptosis, mature IL-1β secretion and the formation of ASC specks. We then conducted a transcriptome analysis in scoparone-pretreated BMDMs, and found that the differentially expressed genes were significantly enriched in mitochondrial reactive oxygen species (ROS) metabolic process, mitochondrial translation and assembly process, as well as in inflammatory response. We demonstrated in J774A.1 cells and BMDMs that scoparone promoted mitophagy, a well-characterized mechanism to control mitochondrial quality and reduce ROS production and subsequent NLRP3 inflammasome activation. Mitophagy blockade by 3-methyladenine (3-MA, 5 mM) reversed the protective effects of scoparone on mitochondrial damage and inflammation in the murine macrophages. Moreover, administration of scoparone (50 mg/kg) exerted significant preventive effects via inhibition of NLRP3 activation in mouse models of bacterial enteritis and septic shock. Collectively, scoparone displays potent anti-inflammatory effects via blocking NLRP3 inflammasome activation through enhancing mitophagy, highlighting a potential action mechanism in treating inflammasome-related diseases for further clinical investigation.
Collapse
Affiliation(s)
- Wan-di Feng
- Beijing Research Institute of Chinese Medicine, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Yao Wang
- Department of Immunology, School of Life Science, Beijing University of Chinese Medicine, Beijing, 100029, China
- National Key Laboratory of Efficacy and Mechanism on Chinese Medicine for Metabolic Diseases, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Tong Luo
- Department of Immunology, School of Life Science, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Xin Jia
- Department of Pharmacology, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Cui-Qin Cheng
- Department of Immunology, School of Life Science, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Hao-Jia Wang
- Department of Immunology, School of Life Science, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Mei-Qi Zhang
- Department of Pharmacology, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Qi-Qi Li
- Department of Pharmacology, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Xue-Jiao Wang
- Department of Immunology, School of Life Science, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Yi-Ying Li
- Department of Immunology, School of Life Science, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Jin-Yong Wang
- Department of Pharmacology, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Guang-Rui Huang
- Department of Immunology, School of Life Science, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Ting Wang
- Beijing Research Institute of Chinese Medicine, Beijing University of Chinese Medicine, Beijing, 100029, China
- National Key Laboratory of Efficacy and Mechanism on Chinese Medicine for Metabolic Diseases, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - An-Long Xu
- Beijing Research Institute of Chinese Medicine, Beijing University of Chinese Medicine, Beijing, 100029, China.
- Department of Immunology, School of Life Science, Beijing University of Chinese Medicine, Beijing, 100029, China.
- National Key Laboratory of Efficacy and Mechanism on Chinese Medicine for Metabolic Diseases, Beijing University of Chinese Medicine, Beijing, 100029, China.
| |
Collapse
|
18
|
Long J, Sun Y, Liu S, Yang S, Chen C, Zhang Z, Chu S, Yang Y, Pei G, Lin M, Yan Q, Yao J, Lin Y, Yi F, Meng L, Tan Y, Ai Q, Chen N. Targeting pyroptosis as a preventive and therapeutic approach for stroke. Cell Death Discov 2023; 9:155. [PMID: 37165005 PMCID: PMC10172388 DOI: 10.1038/s41420-023-01440-y] [Citation(s) in RCA: 53] [Impact Index Per Article: 26.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Revised: 04/18/2023] [Accepted: 04/20/2023] [Indexed: 05/12/2023] Open
Abstract
Stroke has caused tremendous social stress worldwide, yet despite decades of research and development of new stroke drugs, most have failed and rt-PA (Recombinant tissue plasminogen activator) is still the accepted treatment for ischemic stroke. the complexity of the stroke mechanism has led to unsatisfactory efficacy of most drugs in clinical trials, indicating that there are still many gaps in our understanding of stroke. Pyroptosis is a programmed cell death (PCD) with inflammatory properties and are thought to be closely associated with stroke. Pyroptosis is regulated by the GSDMD of the gasdermin family, which when cleaved by Caspase-1/Caspase-11 into N-GSDMD with pore-forming activity can bind to the plasma membrane to form small 10-20 nm pores, which would allow the release of inflammatory factors IL-18 and IL-1β before cell rupture, greatly exacerbating the inflammatory response. The pyroptosis occurs mainly in the border zone of cerebral infarction, and glial cells, neuronal cells and brain microvascular endothelial cells (BMECs) all undergo pyroptosis after stroke, which largely exacerbates the breakdown of the blood-brain barrier (BBB) and thus aggravates brain injury. Therefore, pyroptosis may be a good direction for the treatment of stroke. In this review, we focus on the latest mechanisms of action of pyroptosis and the process by which pyroptosis regulates stroke development. We also suggest potential therapeutic stroke drugs that target the pyroptosis pathway, providing additional therapeutic strategies for the clinical management of stroke. The role of pyroptosis after stroke. After stroke, microglia first rush to the damaged area and polarize into M1 and M2 types. Under the influence of various stimuli, microglia undergo pyroptosis, release pro-inflammatory factors, and are converted to the M1 type; astrocytes and neuronal cells also undergo pyroptosis under the stimulation of various pro-inflammatory factors, leading to astrocyte death due to increased osmotic pressure in the membrane, resulting in water absorption and swelling until rupture. BMECs, the main structural component of the BBB, also undergo pyroptosis when stimulated by pro-inflammatory factors released from microglia and astrocytes, leading to the destruction of the structural integrity of the BBB, ultimately causing more severe brain damage. In addition, GSDMD in neutrophils mainly mediate the release of NETs rather than pyroptosis, which also aggravates brain injury. IL-10=interleukin-10; TGF-β = transforming growth factor-β; IL-18=interleukin-18; IL-1β = interleukin-1β; TNF-α = tumor necrosis factor-α; iNOS=induced nitrogen monoxide synthase; MMPs=Matrix metalloproteinases; GSDMD = gasdermin D; BMECs=brain microvascular endothelial cells; BBB = blood-brain barrier.
Collapse
Affiliation(s)
- Junpeng Long
- Hunan Engineering Technology Center of Standardization and Function of Chinese Herbal Decoction Pieces, College of Pharmacy, Hunan University of Chinese Medicine, Changsha, P. R. China
| | - Yang Sun
- Hunan Engineering Technology Center of Standardization and Function of Chinese Herbal Decoction Pieces, College of Pharmacy, Hunan University of Chinese Medicine, Changsha, P. R. China
| | - Shasha Liu
- Department of Pharmacy, Changsha Hospital for Matemal & Child Health Care, Changsha, P. R. China
| | - Songwei Yang
- Hunan Engineering Technology Center of Standardization and Function of Chinese Herbal Decoction Pieces, College of Pharmacy, Hunan University of Chinese Medicine, Changsha, P. R. China
| | - Chen Chen
- Department of Pharmacy, The First Hospital of Lanzhou University, Lanzhou, P. R. China
| | - Zhao Zhang
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica & Neuroscience Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, P. R. China
| | - Shifeng Chu
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica & Neuroscience Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, P. R. China
| | - Yantao Yang
- Hunan Engineering Technology Center of Standardization and Function of Chinese Herbal Decoction Pieces, College of Pharmacy, Hunan University of Chinese Medicine, Changsha, P. R. China
| | - Gang Pei
- Hunan Engineering Technology Center of Standardization and Function of Chinese Herbal Decoction Pieces, College of Pharmacy, Hunan University of Chinese Medicine, Changsha, P. R. China
| | - Meiyu Lin
- Hunan Engineering Technology Center of Standardization and Function of Chinese Herbal Decoction Pieces, College of Pharmacy, Hunan University of Chinese Medicine, Changsha, P. R. China
| | - Qian Yan
- Hunan Engineering Technology Center of Standardization and Function of Chinese Herbal Decoction Pieces, College of Pharmacy, Hunan University of Chinese Medicine, Changsha, P. R. China
| | - Jiao Yao
- Hunan Engineering Technology Center of Standardization and Function of Chinese Herbal Decoction Pieces, College of Pharmacy, Hunan University of Chinese Medicine, Changsha, P. R. China
| | - Yuting Lin
- Hunan Engineering Technology Center of Standardization and Function of Chinese Herbal Decoction Pieces, College of Pharmacy, Hunan University of Chinese Medicine, Changsha, P. R. China
| | - Fan Yi
- Key Laboratory of Cosmetic, China National Light Industry, Beijing Technology and Business University, Beijing, P. R. China
| | - Lei Meng
- Hunan Engineering Technology Center of Standardization and Function of Chinese Herbal Decoction Pieces, College of Pharmacy, Hunan University of Chinese Medicine, Changsha, P. R. China
| | - Yong Tan
- Department of Nephrology, Xiangtan Central Hospital, Xiangtan, P. R. China
| | - Qidi Ai
- Hunan Engineering Technology Center of Standardization and Function of Chinese Herbal Decoction Pieces, College of Pharmacy, Hunan University of Chinese Medicine, Changsha, P. R. China.
| | - Naihong Chen
- Hunan Engineering Technology Center of Standardization and Function of Chinese Herbal Decoction Pieces, College of Pharmacy, Hunan University of Chinese Medicine, Changsha, P. R. China.
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica & Neuroscience Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, P. R. China.
| |
Collapse
|
19
|
Leng S, Xu W, Wu L, Liu L, Du J, Yang F, Huang D, Zhang L. NLRP3 Disturbs Treg/Th17 Cell Balance to Aggravate Apical Periodontitis. J Dent Res 2023; 102:656-666. [PMID: 36883625 DOI: 10.1177/00220345231151692] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/09/2023] Open
Abstract
Apical periodontitis is an inflammatory condition that is considered an immunological reaction of the periapical tissue to invading bacteria and their pathogenic components. Recent research has revealed that NLR family pyrin domain containing 3 (NLRP3) is crucial to the pathogenesis of apical periodontitis and serves as a link between innate and adaptive immunity. The balance between regulatory T-cell (Treg) and T helper cell 17 (Th17 cell) determines the direction of the inflammatory response. Therefore, this study aimed to investigate whether NLRP3 exacerbated periapical inflammation by disturbing Treg/Th17 balance and the underlying regulatory mechanisms. In the present study, NLRP3 was raised in apical periodontitis tissues as opposed to healthy pulp tissues. Low NLRP3 expression in dendritic cells (DCs) increased transforming growth factor β secretion while decreasing interleukin (IL)-1β and IL-6 production. The Treg ratio and IL-10 secretion rose when CD4+ T cells were cocultured with DCs primed with IL-1β neutralizing antibody (anti-IL-1β) and specific small interfering RNA (siRNA) targeting NLRP3 (siRNA NLRP3), but the proportion of Th17 cells and IL-17 release dropped. Furthermore, siRNA NLRP3-mediated suppression of NLRP3 expression aided Treg differentiation and elevated Foxp3 expression as well as IL-10 production in CD4+ T cells. Inhibition of NLRP3 activity by MCC950 boosted the percentage of Tregs while decreasing the ratio of Th17 cells, leading to reduced periapical inflammation and bone resorption. Nigericin administration, however, exacerbated periapical inflammation and bone destruction with an unbalanced Treg/Th17 response. These findings demonstrate that NLRP3 is a pivotal regulator by regulating the release of inflammatory cytokines from DCs or directly suppressing Foxp3 expression to disturb Treg/Th17 balance, thus exacerbating apical periodontitis.
Collapse
Affiliation(s)
- S Leng
- Department of Operative Dentistry and Endodontics, State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Disease, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - W Xu
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key laboratory of Oral Biomedical Research of Zhejiang Province Cancer Center of Zhejiang University, Hangzhou, China
| | - L Wu
- Department of Geriatric Stomatology, State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - L Liu
- Department of Operative Dentistry and Endodontics, State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Disease, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - J Du
- Department of Health Care (Department of General Dentistry II), School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration & Shandong Provincial Clinical Research Center for Oral Diseases, Jinan, China
| | - F Yang
- Department of Operative Dentistry and Endodontics, State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Disease, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - D Huang
- Department of Operative Dentistry and Endodontics, State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Disease, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - L Zhang
- Department of Operative Dentistry and Endodontics, State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Disease, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| |
Collapse
|
20
|
Moutan Cortex Extract Modulates Macrophage Activation via Lipopolysaccharide-Induced Calcium Signaling and ER Stress-CHOP Pathway. Int J Mol Sci 2023; 24:ijms24032062. [PMID: 36768384 PMCID: PMC9916843 DOI: 10.3390/ijms24032062] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Revised: 01/16/2023] [Accepted: 01/17/2023] [Indexed: 01/21/2023] Open
Abstract
Moutan Cortex, Paeonia suffruticosa root, has long been used as a medicine for the treatment of inflammatory diseases. The aim of this study was to evaluate the modulative properties of Moutan Cortex water extract (CP) on endoplasmic reticulum (ER) stress-related macrophage activation via the calcium-CHOP pathway. RAW 264.7 mouse macrophages were activated by lipopolysaccharide (LPS), and the levels of various inflammatory mediators from RAW 264.7 were evaluated. The multiplex cytokine assay was used to investigate both cytokines and growth factors, and RT-PCR was used to investigate the expressions of inflammation-related genes, such as CHOP. Data represent the levels of NO and cytosolic calcium in LPS-stimulated RAW 264.7 were significantly inhibited by CP as well as hydrogen peroxide (p < 0.05). Minutely, NO production in LPS-stimulated RAW 264.7 incubated with CP at concentrations of 25, 50, 100, and 200 µg/mL for 24 h was 97.32 ± 1.55%, 95.86 ± 2.26%, 94.64 ± 1.83%, and 92.69 ± 2.31% of the control value (LPS only), respectively (p < 0.05). Calcium release in LPS-stimulated RAW 264.7 incubated with CP at concentrations of 25, 50, 100, and 200 µg/mL for 18 h was 95.78 ± 1.64%, 95.41 ± 1.14%, 94.54 ± 2.76%, and 90.89 ± 3.34% of the control value, respectively (p < 0.05). Hydrogen peroxide production in LPS-stimulated RAW 264.7 incubated with CP at concentrations of 25, 50, 100, and 200 µg/mL for 24 h was 79.15 ± 7.16%, 63.83 ± 4.03%, 46.27 ± 4.38%, and 40.66 ± 4.03% of the control value, respectively (p < 0.05). It is interesting that the production of IL-6, TNF-α, G-CSF, MIP-1α, MIP-2, and M-CSF in LPS-stimulated RAW 264.7 were significantly inhibited by CP (p < 0.05), while the production of LIX, LIF, RANTES, and MIP-1β showed a meaningful decrease. CP at concentrations of 25, 50, 100, and 200 µg/mL significantly reduced the transcription of Chop, Camk2α, NOS, STAT1, STAT3, Ptgs2, Jak2, c-Jun, Fas, c-Fos, TLR3, and TLR9 in LPS-stimulated RAW 264.7 (p < 0.05). CP at concentrations of 25, 50, and 100 µg/mL significantly reduced the phosphorylation of STAT3, p38 MAPK, and IκB-α in LPS-stimulated RAW 264.7 (p < 0.05). These results suggest that CP might modulate macrophage activation via LPS-induced calcium signaling and the ER stress-CHOP pathway.
Collapse
|
21
|
Xu J, Gao C, He Y, Fang X, Sun D, Peng Z, Xiao H, Sun M, Zhang P, Zhou T, Yang X, Yu Y, Li R, Zou X, Shu H, Qiu Y, Zhou X, Yuan S, Yao S, Shang Y. NLRC3 expression in macrophage impairs glycolysis and host immune defense by modulating the NF-κB-NFAT5 complex during septic immunosuppression. Mol Ther 2023; 31:154-173. [PMID: 36068919 PMCID: PMC9840117 DOI: 10.1016/j.ymthe.2022.08.023] [Citation(s) in RCA: 38] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Revised: 06/23/2022] [Accepted: 08/30/2022] [Indexed: 01/28/2023] Open
Abstract
Impairment of innate immune cell function and metabolism underlies immunosuppression in sepsis; however, a promising therapy to orchestrate this impairment is currently lacking. In this study, high levels of NOD-like receptor family CARD domain containing-3 (NLRC3) correlated with the glycolytic defects of monocytes/macrophages from septic patients and mice that developed immunosuppression. Myeloid-specific NLRC3 deletion improved macrophage glycolysis and sepsis-induced immunosuppression. Mechanistically, NLRC3 inhibits nuclear factor (NF)-κB p65 binding to nuclear factor of activated T cells 5 (NFAT5), which further controls the expression of glycolytic genes and proinflammatory cytokines of immunosuppressive macrophages. This is achieved by decreasing NF-κB activation-co-induced by TNF-receptor-associated factor 6 (TRAF6) or mammalian target of rapamycin (mTOR)-and decreasing transcriptional co-activator p300 activity by inducing NLRC3 sequestration of mTOR and p300. Genetic inhibition of NLRC3 disrupted the NLRC3-mTOR-p300 complex and enhanced NF-κB binding to the NFAT5 promoter in concert with p300. Furthermore, intrapulmonary delivery of recombinant adeno-associated virus harboring a macrophage-specific NLRC3 deletion vector significantly improved the defense of septic mice that developed immunosuppression upon secondary intratracheal bacterial challenge. Collectively, these findings indicate that NLRC3 mediates critical aspects of innate immunity that contribute to an immunocompromised state during sepsis and identify potential therapeutic targets.
Collapse
Affiliation(s)
- Jiqian Xu
- Department of Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; Institute of Anesthesiology and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Chenggang Gao
- Department of Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Yajun He
- Department of Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Xiangzhi Fang
- Department of Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Deyi Sun
- Department of Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Zhekang Peng
- Department of Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Hairong Xiao
- Institute of Anesthesiology and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Miaomiao Sun
- Department of Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Pei Zhang
- Department of Paediatrics, Jinling Hospital, School of Medicine, Nanjing University, Nanjing 210016, China
| | - Ting Zhou
- Department of Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Xiaobo Yang
- Department of Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Yuan Yu
- Department of Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Ruiting Li
- Department of Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Xiaojing Zou
- Department of Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Huaqing Shu
- Department of Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Yang Qiu
- State Key Laboratory of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan Institute of Virology, Wuhan 43007, China
| | - Xi Zhou
- State Key Laboratory of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan Institute of Virology, Wuhan 43007, China
| | - Shiying Yuan
- Department of Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; Institute of Anesthesiology and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Shanglong Yao
- Institute of Anesthesiology and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - You Shang
- Department of Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; Institute of Anesthesiology and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China.
| |
Collapse
|
22
|
Li M, He Y, Zhang W, Yin Y, Jiang Q, Loor JJ, Wang J, Wen J, Yang W, Xu C, Zhang B. Taraxasterol alleviates fatty acid-induced lipid deposition in calf hepatocytes by decreasing ROS production and endoplasmic reticulum stress. J Anim Sci 2023; 101:skad015. [PMID: 36638067 PMCID: PMC9977349 DOI: 10.1093/jas/skad015] [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: 07/13/2022] [Accepted: 01/09/2023] [Indexed: 01/14/2023] Open
Abstract
Increased concentrations of free fatty acids (FFAs) induce reactive oxygen species (ROSs) generation and endoplasmic reticulum (ER) stress, thus, increasing the risk of fatty liver in dairy cows during the periparturient period. In non-ruminants, Taraxasterol (Tara; a pentacyclic triterpenoid found in medicinal plants) plays an important role in anti-inflammatory and antioxidant reactions. Whether Tara can alleviate or prevent fatty liver in ruminants is unknown. We addressed whether Tara supply could dampen lipid accumulation, ROSs production, and ER stress caused by FFAs in calf hepatocytes. Primary calf hepatocytes were isolated from five healthy calves (1 d old, female, 30-40 kg, fasting, rectal temperature 38.7-39.7 °C). In the first experiment, hepatocytes were incubated with various concentrations of Tara (2.5, 5, and 10 μg/mL) for 12 h prior to the 1.2-mM FFAs challenge. Results indicated that the level of ROSs was lowest with 5 μg/mL Tara. Thus, to further characterize the molecular mechanisms whereby Tara protects from FFAs-induced lipid deposition in calf hepatocytes, we performed incubations with 5 μg/mL Tara for 12 h prior to a 1.2-mM FFAs challenge for an additional 12 h. Results indicated that 1.2-mM FFAs challenge increased mitochondrial membrane potential (MMP), enhanced expression of proteins and mRNA associated with ER stress (PERK, IRE1, GRP78, ATF6, and CHOP) and fatty acid synthesis (FASN, ACC1, and SREBP-1c), and ultimately led to increased lipid droplet synthesis. In contrast, Tara treatment alleviated these negative effects after 1.2-mM FFAs challenge. To determine whether Tara protects against FFAs-induced lipid droplet synthesis by alleviating oxidative stress, hepatocytes were treated with 5 μg/mL Tara for 22 h prior to H2O2 (440 μM) challenge for 2 h. Compared with H2O2 treatment alone, results revealed a marked decrease in ROSs, MMP, and protein abundance of ER stress (GRP78, ATF6, and CHOP) and lipid droplet synthesis in response to Tara prior to H2O2 challenge. Data suggested that the increase in mitochondrial ROSs production contributes to lipid accumulation in calf hepatocytes. Collectively, our in vitro data indicate that Tara alleviates fatty acid-induced lipid deposition. Further research is warranted to ascertain that Tara can be helpful in the therapeutic management of early lactating cows to control or alleviate excessive hepatic lipid deposition.
Collapse
Affiliation(s)
- Ming Li
- College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing163319, Heilongjiang, China
| | - Yuxin He
- College of Life Science and Technology, Heilongjiang Bayi Agricultural University, Daqing163319, Heilongjiang, China
| | - Wei Zhang
- College of Life Science and Technology, Heilongjiang Bayi Agricultural University, Daqing163319, Heilongjiang, China
| | - Yufeng Yin
- College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing163319, Heilongjiang, China
| | - Qianming Jiang
- Mammalian NutriPhysio Genomics, Department of Animal Sciences and Division of Nutritional Sciences, University of Illinois, Urbana, IL 61801, USA
| | - Juan J Loor
- Mammalian NutriPhysio Genomics, Department of Animal Sciences and Division of Nutritional Sciences, University of Illinois, Urbana, IL 61801, USA
| | - Jingjing Wang
- College of Life Science and Technology, Heilongjiang Bayi Agricultural University, Daqing163319, Heilongjiang, China
| | - Jianan Wen
- College of Life Science and Technology, Heilongjiang Bayi Agricultural University, Daqing163319, Heilongjiang, China
| | - Wei Yang
- College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing163319, Heilongjiang, China
| | - Chuang Xu
- College of Veterinary Medicine, China Agricultural University, Beijing 100091, China
| | - Bingbing Zhang
- College of Life Science and Technology, Heilongjiang Bayi Agricultural University, Daqing163319, Heilongjiang, China
| |
Collapse
|
23
|
Peng X, Zhang C, Gao JW, Wang F, Bao JP, Zhou ZM, Sun R, Ji HY, Vlf C, Wu XT. A20 ameliorates disc degeneration by suppressing mTOR/BNIP3 axis-mediated mitophagy. Genes Genomics 2022; 45:657-671. [PMID: 36583816 DOI: 10.1007/s13258-022-01343-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Accepted: 10/27/2022] [Indexed: 12/31/2022]
Abstract
BACKGROUND The pathological mechanism of intervertebral disc degeneration (IDD) is an unanswered question that we are committed to exploring. A20 is an anti-inflammatory protein of nucleus pulposus (NP) cells and plays a protective role in intervertebral disc degeneration. OBJECTIVE This study aims to investigate the molecular mechanism by which A20 attenuates disc degeneration. METHODS The proteins of interest were measured by immunoblotting, immunofluorescence, ELISA assay, and immunohistochemical technique to conduct related experiments. Immunofluorescence assays and mitochondrial membrane potential (JC-1) were used to assess mitophagy and mitochondrial fitness, respectively. RESULTS Here, we demonstrated that A20 promoted mitophagy, attenuated pyroptosis, and inhibited the degradation of the extracellular matrix, consequently significantly ameliorating disc degeneration. Mechanistically, A20 reduces pyroptosis and further suppresses cellular mTOR activity. On the one hand, A20-induced mTOR inhibition triggers BNIP3-mediated mitophagy to ensure mitochondrial fitness under LPS stimulation, as a result of mitigating mitochondrial dysfunction induced by LPS. On the other hand, A20-induced mTOR inhibition reduces the loss of mitochondrial membrane potential and the generation of Mitochondrial ROS. CONCLUSION The study revealed that A20 promotes BNIP3-mediated mitophagy by suppressing mTOR pathway activation against LPS-induced pyroptosis.
Collapse
Affiliation(s)
- Xin Peng
- Medical School of Southeast University, Nanjing, China
| | - Cong Zhang
- Department of Orthopedics, Zhongda Hospital, Southeast University, Nanjing, China
| | - Jia-Wei Gao
- Medical School of Southeast University, Nanjing, China
| | - Feng Wang
- Department of Orthopedics, Zhongda Hospital, Southeast University, Nanjing, China
| | - Jun-Ping Bao
- Department of Orthopedics, Zhongda Hospital, Southeast University, Nanjing, China
| | - Zhi-Min Zhou
- Medical School of Southeast University, Nanjing, China
| | - Rui Sun
- Medical School of Southeast University, Nanjing, China
| | - Hang-Yu Ji
- Department of Orthopedics, Zhongda Hospital, Southeast University, Nanjing, China
| | - Cabral Vlf
- Medical School of Southeast University, Nanjing, China
| | - Xiao-Tao Wu
- Medical School of Southeast University, Nanjing, China.
- Department of Orthopedics, Zhongda Hospital, Southeast University, Nanjing, China.
| |
Collapse
|
24
|
Zhao M, Wei F, Sun G, Wen Y, Xiang J, Su F, Zhan L, Nian Q, Chen Y, Zeng J. Natural compounds targeting glycolysis as promising therapeutics for gastric cancer: A review. Front Pharmacol 2022; 13:1004383. [PMID: 36438836 PMCID: PMC9684197 DOI: 10.3389/fphar.2022.1004383] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Accepted: 10/28/2022] [Indexed: 09/23/2023] Open
Abstract
Gastric cancer, a common malignant disease, seriously endangers human health and life. The high mortality rate due to gastric cancer can be attributed to a lack of effective therapeutic drugs. Cancer cells utilize the glycolytic pathway to produce energy even under aerobic conditions, commonly referred to as the Warburg effect, which is a characteristic of gastric cancer. The identification of new targets based on the glycolytic pathway for the treatment of gastric cancer is a viable option, and accumulating evidence has shown that phytochemicals have extensive anti-glycolytic properties. We reviewed the effects and mechanisms of action of phytochemicals on aerobic glycolysis in gastric cancer cells. Phytochemicals can effectively inhibit aerobic glycolysis in gastric cancer cells, suppress cell proliferation and migration, and promote apoptosis, via the PI3K/Akt, c-Myc, p53, and other signaling pathways. These pathways affect the expressions of HIF-1α, HK2, LDH, and other glycolysis-related proteins. This review further assesses the potential of using plant-derived compounds for the treatment of gastric cancer and sheds insight into the development of new drugs.
Collapse
Affiliation(s)
- Maoyuan Zhao
- Department of Oncology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Feng Wei
- Department of Oncology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Guangwei Sun
- Department of Oncology, Sichuan Integrative Medicine Hospital, Chengdu, China
| | - Yueqiang Wen
- School of Basic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Juyi Xiang
- TCM Regulating Metabolic Diseases Key Laboratory of Sichuan Province, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Fangting Su
- Department of Oncology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Lu Zhan
- Department of Oncology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Qing Nian
- Department of Blood Transfusion, Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Yu Chen
- Department of Oncology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Jinhao Zeng
- TCM Regulating Metabolic Diseases Key Laboratory of Sichuan Province, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
- Geriatric Department, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| |
Collapse
|
25
|
Chen H, Zhou J, Zhang G, Luo Z, Li L, Kang X. Emerging role and therapeutic implication of mTOR signalling in intervertebral disc degeneration. Cell Prolif 2022; 56:e13338. [PMID: 36193577 PMCID: PMC9816935 DOI: 10.1111/cpr.13338] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2022] [Revised: 06/16/2022] [Accepted: 09/01/2022] [Indexed: 01/11/2023] Open
Abstract
Intervertebral disc degeneration (IDD), an important cause of chronic low back pain (LBP), is considered the pathological basis for various spinal degenerative diseases. A series of factors, including inflammatory response, oxidative stress, autophagy, abnormal mechanical stress, nutritional deficiency, and genetics, lead to reduced extracellular matrix (ECM) synthesis by intervertebral disc (IVD) cells and accelerate IDD progression. Mammalian target of rapamycin (mTOR) is an evolutionarily conserved serine/threonine kinase that plays a vital role in diverse degenerative diseases. Recent studies have shown that mTOR signalling is involved in the regulation of autophagy, oxidative stress, inflammatory responses, ECM homeostasis, cellular senescence, and apoptosis in IVD cells. Accordingly, we reviewed the mechanism of mTOR signalling in the pathogenesis of IDD to provide innovative ideas for future research and IDD treatment.
Collapse
Affiliation(s)
- Hai‐Wei Chen
- Department of OrthopaedicsLanzhou University Second HospitalLanzhouGansuPeople's Republic of China,The Second Clinical Medical CollegeLanzhou UniversityLanzhouGansuPeople's Republic of China
| | - Jian‐Wei Zhou
- Department of OrthopaedicsLanzhou University Second HospitalLanzhouGansuPeople's Republic of China,Key Laboratory of Orthopaedics Disease of Gansu ProvinceLanzhou University Second HospitalLanzhouGansu ProvincePeople's Republic of China
| | - Guang‐Zhi Zhang
- Department of OrthopaedicsLanzhou University Second HospitalLanzhouGansuPeople's Republic of China,The Second Clinical Medical CollegeLanzhou UniversityLanzhouGansuPeople's Republic of China
| | - Zhang‐Bin Luo
- Department of OrthopaedicsLanzhou University Second HospitalLanzhouGansuPeople's Republic of China,The Second Clinical Medical CollegeLanzhou UniversityLanzhouGansuPeople's Republic of China
| | - Lei Li
- Department of OrthopaedicsLanzhou University Second HospitalLanzhouGansuPeople's Republic of China,The Second Clinical Medical CollegeLanzhou UniversityLanzhouGansuPeople's Republic of China
| | - Xue‐Wen Kang
- Department of OrthopaedicsLanzhou University Second HospitalLanzhouGansuPeople's Republic of China,The Second Clinical Medical CollegeLanzhou UniversityLanzhouGansuPeople's Republic of China,Key Laboratory of Orthopaedics Disease of Gansu ProvinceLanzhou University Second HospitalLanzhouGansu ProvincePeople's Republic of China
| |
Collapse
|
26
|
Sun J, Li Y. Pyroptosis and respiratory diseases: A review of current knowledge. Front Immunol 2022; 13:920464. [PMID: 36248872 PMCID: PMC9561627 DOI: 10.3389/fimmu.2022.920464] [Citation(s) in RCA: 42] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Accepted: 09/20/2022] [Indexed: 11/13/2022] Open
Abstract
Pyroptosis is a relatively newly discovered programmed cell death accompanied by an inflammatory response. In the classical view, pyroptosis is mediated by caspases-1,-4,-5,-11 and executed by GSDMD, however, recently it was demonstrated that caspase-3 and-8 also participate in the process of pyroptosis, by cleaving GSDMD/E and GSDMD respectively. Different from autophagy and apoptosis, many pores are formed on the cell membrane during pyroptosis, which makes the cell membrane lose its integrity, eventually leading to the release of cytokines interleukin(IL)-1β and IL-18. When the body is infected with pathogens or exposed to some stimulations, pyroptosis could play an immune defense role. It is found that pyroptosis exists widely in infectious and inflammatory respiratory diseases such as acute lung injury, bronchial dysplasia, chronic obstructive pulmonary disease, and asthma. Excessive pyroptosis may accompany airway inflammation, tissue injury, and airway damage, and induce an inflammatory reaction, leading to more serious damage and poor prognosis of respiratory diseases. This review summarizes the relationship between pyroptosis and related respiratory diseases.
Collapse
|
27
|
Kania-Dobrowolska M, Baraniak J. Dandelion (Taraxacum officinale L.) as a Source of Biologically Active Compounds Supporting the Therapy of Co-Existing Diseases in Metabolic Syndrome. Foods 2022; 11:foods11182858. [PMID: 36140985 PMCID: PMC9498421 DOI: 10.3390/foods11182858] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Revised: 09/08/2022] [Accepted: 09/13/2022] [Indexed: 11/21/2022] Open
Abstract
Nowadays, many people are struggling with obesity, type 2 diabetes, and atherosclerosis, which are called the scourge of the 21st century. These illnesses coexist in metabolic syndrome, which is not a separate disease entity because it includes several clinical conditions such as central (abdominal) obesity, elevated blood pressure, and disorders of carbohydrate and fat metabolism. Lifestyle is considered to have an impact on the development of metabolic syndrome. An unbalanced diet, the lack of sufficient physical activity, and genetic factors result in the development of type 2 diabetes and atherosclerosis, which significantly increase the risk of cardiovascular complications. The treatment of metabolic syndrome is aimed primarily at reducing the risk of the development of coexisting diseases, and the appropriate diet is the key factor in the treatment. Plant raw materials containing compounds that regulate lipid and carbohydrate metabolism in the human body are investigated. Dandelion (Taraxacum officinale F.H. Wigg.) is a plant, the consumption of which affects the regulation of lipid and sugar metabolism. The growth of this plant is widely spread in Eurasia, both Americas, Africa, New Zealand, and Australia. The use and potential of this plant that is easily accessible in the world in contributing to the treatment of type 2 diabetes and atherosclerosis have been proved by many studies.
Collapse
|
28
|
miR-99a-5p: A Potential New Therapy for Atherosclerosis by Targeting mTOR and Then Inhibiting NLRP3 Inflammasome Activation and Promoting Macrophage Autophagy. DISEASE MARKERS 2022; 2022:7172583. [PMID: 35968506 PMCID: PMC9374553 DOI: 10.1155/2022/7172583] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Accepted: 07/14/2022] [Indexed: 11/18/2022]
Abstract
Objective MicroRNAs have been revealed to be involved in the development of atherosclerosis. The present study is aimed at exploring the potential of miR-99a-5p as a therapy for atherosclerosis. We suspected that miR-99a-5p might inhibit NLRP3 inflammasome activation and promote macrophage autophagy via constraining mTOR, therefore, alleviating atherosclerosis. Methods The cell viability in ox-LDL-induced THP-1 macrophages was assessed by CCK-8 assay. Bioinformatic analysis was used to predict the target genes of miR-99a-5p. The binding between miR-99a-5p and mTOR was confirmed by luciferase reporter assay. In vivo, a high-fat-diet-induced atherosclerosis model was established in apolipoprotein E knockout mice. Hematoxylin-eosin, oil red O, and Sirius red staining were performed for the determination of atherosclerotic lesions. MTOR and associated protein levels were detected by Western blot analysis. Results miR-99a-5p inhibited NLRP3 inflammasome activation and promoted macrophage autophagy by targeting mTOR. Enforced miR-99a-5p significantly reduced the levels of inflammasome complex and inflammatory cytokines. Furthermore, miR-99a-5p overexpression inhibited the expression of mTOR, whereas mTOR overexpression reversed the trend of the above behaviors. In vivo, the specific overexpression of miR-99a-5p significantly reduced atherosclerotic lesions, accompanied by a significant downregulation of autophagy marker CD68 protein expression. Conclusion We demonstrated for the first time that miR-99a-5p may be considered a therapy for atherosclerosis. The present study has revealed that miR-99a-5p might inhibit NLRP3 inflammasome activation and promote macrophage autophagy by targeting mTOR, therefore, alleviating atherosclerosis.
Collapse
|
29
|
He M, Fan J, Zhou R, Gao G, Li R, Zuo Y, Li B, Li Y, Sun T. NLRP3/Caspase-1-Mediated Pyroptosis of Astrocytes Induced by Antipsychotics Is Inhibited by a Histamine H1 Receptor-Selective Agonist. Front Aging Neurosci 2022; 14:847561. [PMID: 35615587 PMCID: PMC9125084 DOI: 10.3389/fnagi.2022.847561] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2022] [Accepted: 04/15/2022] [Indexed: 11/13/2022] Open
Abstract
Emerging data indicate that antipsychotic treatment causes brain volume loss and astrocyte death, but the mechanisms remain elusive. Pyroptosis, inflammatory cell death characterized by the formation of inflammatory bodies, increased expression of nod-like receptor proteins (NLRPs) such as NLRP3, and activation of caspases and gasdermin D (GSDMD) are largely associated with innate immunity, inflammation, and cell injury/death. However, the main effect of antipsychotics on astrocyte pyroptotic signaling and the molecular mechanisms remain obscure. In the present study, 72-h treatment with olanzapine, quetiapine, risperidone, or haloperidol significantly decreased the viability of astrocytes. Twenty-four hour treatment with olanzapine, quetiapine, risperidone, or haloperidol dose-dependently increased the protein expression of astrocytic NLRP3, NLRP6, caspase-1, caspase-4, and GSDMD. Co-treatment with a histamine H1 receptor agonist, 2-(3-trifluoromethylphenyl) histamine (FMPH), dose-dependently reduced the increased expression of NLRP3, caspase-1 and GSDMD induced by olanzapine, quetiapine, risperidone, or haloperidol. Moreover, olanzapine, quetiapine, risperidone, or haloperidol treatment induced pore formation in the membranes of astrocytes, and these effects were inhibited by FMPH co-treatment. Taken together, antipsychotic treatment activated astrocyte pyroptotic signaling, and these effects may be related to antipsychotic-induced astrocyte death. H1 receptor activation is an effective treatment strategy to suppress antipsychotic-induced astrocyte pyroptosis and inflammation.
Collapse
Affiliation(s)
- Meng He
- School of Chemistry, Chemical Engineering and Life Sciences, Wuhan University of Technology, Wuhan, China
| | - Jun Fan
- School of Chemistry, Chemical Engineering and Life Sciences, Wuhan University of Technology, Wuhan, China
| | - Ruqin Zhou
- School of Chemistry, Chemical Engineering and Life Sciences, Wuhan University of Technology, Wuhan, China
| | - Guanbin Gao
- State Key Laboratory of Advanced Technology for Materials Synthesis and Processing, Wuhan University of Technology, Wuhan, China
| | - Ruoxi Li
- School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - YuFeng Zuo
- School of Chemistry, Chemical Engineering and Life Sciences, Wuhan University of Technology, Wuhan, China
| | - Benben Li
- School of Chemistry, Chemical Engineering and Life Sciences, Wuhan University of Technology, Wuhan, China
| | - Yanmei Li
- Hubei Provincial Key Laboratory for Applied Toxicology, Hubei Provincial Center for Disease Control and Prevention, Wuhan, China
- *Correspondence: Taolei Sun,
| | - Taolei Sun
- School of Chemistry, Chemical Engineering and Life Sciences, Wuhan University of Technology, Wuhan, China
- Yanmei Li,
| |
Collapse
|
30
|
Montaño LM, Sommer B, Gomez-Verjan JC, Morales-Paoli GS, Ramírez-Salinas GL, Solís-Chagoyán H, Sanchez-Florentino ZA, Calixto E, Pérez-Figueroa GE, Carter R, Jaimez-Melgoza R, Romero-Martínez BS, Flores-Soto E. Theophylline: Old Drug in a New Light, Application in COVID-19 through Computational Studies. Int J Mol Sci 2022; 23:ijms23084167. [PMID: 35456985 PMCID: PMC9030606 DOI: 10.3390/ijms23084167] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Revised: 04/04/2022] [Accepted: 04/04/2022] [Indexed: 02/04/2023] Open
Abstract
Theophylline (3-methyxanthine) is a historically prominent drug used to treat respiratory diseases, alone or in combination with other drugs. The rapid onset of the COVID-19 pandemic urged the development of effective pharmacological treatments to directly attack the development of new variants of the SARS-CoV-2 virus and possess a therapeutical battery of compounds that could improve the current management of the disease worldwide. In this context, theophylline, through bronchodilatory, immunomodulatory, and potentially antiviral mechanisms, is an interesting proposal as an adjuvant in the treatment of COVID-19 patients. Nevertheless, it is essential to understand how this compound could behave against such a disease, not only at a pharmacodynamic but also at a pharmacokinetic level. In this sense, the quickest approach in drug discovery is through different computational methods, either from network pharmacology or from quantitative systems pharmacology approaches. In the present review, we explore the possibility of using theophylline in the treatment of COVID-19 patients since it seems to be a relevant candidate by aiming at several immunological targets involved in the pathophysiology of the disease. Theophylline down-regulates the inflammatory processes activated by SARS-CoV-2 through various mechanisms, and herein, they are discussed by reviewing computational simulation studies and their different applications and effects.
Collapse
Affiliation(s)
- Luis M. Montaño
- Departamento de Farmacología, Facultad de Medicina, Universidad Nacional Autónoma de México, Ciudad de México 04510, CP, Mexico; (L.M.M.); (R.J.-M.); (B.S.R.-M.)
| | - Bettina Sommer
- Laboratorio de Hiperreactividad Bronquial, Instituto Nacional de Enfermedades Respiratorias “Ismael Cosío Villegas”, Ciudad de México 14080, CP, Mexico;
| | - Juan C. Gomez-Verjan
- Dirección de Investigación, Instituto Nacional de Geriatría, Ciudad de México 10200, CP, Mexico; (J.C.G.-V.); (G.S.M.-P.)
| | - Genaro S. Morales-Paoli
- Dirección de Investigación, Instituto Nacional de Geriatría, Ciudad de México 10200, CP, Mexico; (J.C.G.-V.); (G.S.M.-P.)
| | - Gema Lizbeth Ramírez-Salinas
- Laboratorio de Diseño y Desarrollo de Nuevos Fármacos e Innovación Biotecnológica (Laboratory for the Design and Development of New Drugs and Biotechnological Innovation), Escuela Superior de Medicina, Instituto Politécnico Nacional, Plan de San Luis y Díaz Mirón S/N, Col. Santo Tomas, Ciudad de México 11340, CP, Mexico;
- Departamento de Inmunología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Circuito Escolar s/n, Ciudad de México 14510, CP, Mexico
| | - Héctor Solís-Chagoyán
- Laboratorio de Neurofarmacología, Instituto Nacional de Psiquiatría “Ramón de la Fuente Muñiz”, Ciudad de México 14370, CP, Mexico; (H.S.-C.); (Z.A.S.-F.)
| | - Zuly A. Sanchez-Florentino
- Laboratorio de Neurofarmacología, Instituto Nacional de Psiquiatría “Ramón de la Fuente Muñiz”, Ciudad de México 14370, CP, Mexico; (H.S.-C.); (Z.A.S.-F.)
| | - Eduardo Calixto
- Departamento de Neurobiología, Dirección de Investigación en Neurociencias, Instituto Nacional de Psiquiatría “Ramón de la Fuente Muñiz”, Ciudad de México 14370, CP, Mexico;
| | - Gloria E. Pérez-Figueroa
- Instituto Nacional de Neurología y Neurocirugía, Unidad Periférica en el Estudio de la Neuroinflamación en Patologías Neurológicas, Ciudad de México 06720, CP, Mexico;
- Laboratorio de Investigación en Inmunología y Proteómica, Hospital Infantil de México Federico Gómez, Ciudad de México 06720, CP, Mexico
| | - Rohan Carter
- FRACGP/MBBS, Murchison Outreach Service Mount Magnet Western Australia, Mount Magnet, WA 6530, Australia;
| | - Ruth Jaimez-Melgoza
- Departamento de Farmacología, Facultad de Medicina, Universidad Nacional Autónoma de México, Ciudad de México 04510, CP, Mexico; (L.M.M.); (R.J.-M.); (B.S.R.-M.)
| | - Bianca S. Romero-Martínez
- Departamento de Farmacología, Facultad de Medicina, Universidad Nacional Autónoma de México, Ciudad de México 04510, CP, Mexico; (L.M.M.); (R.J.-M.); (B.S.R.-M.)
| | - Edgar Flores-Soto
- Departamento de Farmacología, Facultad de Medicina, Universidad Nacional Autónoma de México, Ciudad de México 04510, CP, Mexico; (L.M.M.); (R.J.-M.); (B.S.R.-M.)
- Correspondence: ; Tel.: +52-555-6232279
| |
Collapse
|
31
|
Kang L, Miao MS, Song YG, Fang XY, Zhang J, Zhang YN, Miao JX. Total flavonoids of Taraxacum mongolicum inhibit non-small cell lung cancer by regulating immune function. JOURNAL OF ETHNOPHARMACOLOGY 2021; 281:114514. [PMID: 34384846 DOI: 10.1016/j.jep.2021.114514] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Revised: 07/29/2021] [Accepted: 08/08/2021] [Indexed: 06/13/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Taraxacum mongolicum Hand.-Mazz. has been used in lung cancer treatment in Chinese medicine. However, its specific mechanism of action has not yet been reported, and developing pharmaceutical anti-cancer resources is important. Here, we aimed to elucidate the anti-tumor effects of dandelion in vitro and in vivo and assess its effects on immune function in lung cancer patients. AIM OF THE STUDY In the present study, we mainly observed the therapeutic effects of total flavonoids from Taraxacum mongolicum Hand.-Mazz. (TFTM) on non-small cell lung cancer and its influence on the body's immune function. MATERIALS AND METHODS In vitro experiments on A549 and H1299 cells were performed using the CCK8 method; the proliferation and migration of cells were observed to investigate the wound healing effects of TFTM, and flow cytometry was used to detect the apoptotic rate of TFTM on lung cancer cells. In vivo experiments were preformed to establish a non-small cell lung cancer mouse model using subcutaneously transplanted Lewis cells, and the body weight and tumor growth of the mice were recorded. Hematoxylin and eosin staining was performed for tumor tissue to assess pathological changes. The thymus, spleen, and lungs were isolated for to calculate organ index. The CD4+, CD8+, and CD4+/CD8+ levels were detected in mouse spleen using flow cytometry, and IL-2, IL-3, IFN-γ, and TNF-α levels were determined in serum using enzyme-linked immunosorbent assay. Expressions of IL-2, IL-3, IFN-γ, and TNF-α were detected using quantitative real-time PCR in tumor tissues, and Ki67 expression was observed by immunofluorescence. RESULTS At 24 h, TFTM (100 and 200 μg/mL) had the best inhibitory effect on the proliferation of A549 and H1299 cells. The cell migration rate significantly reduced (P < 0.01), and the tumor inhibition rate increased (P < 0.01) and promoted apoptosis (P < 0.01). The mouse thymus index significantly increased (P < 0.05) and mouse spleen index reduced (P < 0.05). The CD4+, CD8+, and CD4+/CD8+ levels in Lewis lung cancer mouse model increased, as did the levels of IL-2, IL-3, IFN-γ, and TNF-α in the serum and tumor of mice; Ki67 expression in tumor tissues significantly reduced (P < 0.01). CONCLUSION TFTM has an inhibitory effect on lung cancer. The mechanism may be that it improves the host's protective immune response by having a milder tumor growth inhibitory effect than cyclophosphamide.
Collapse
Affiliation(s)
- Le Kang
- National International Cooperation Base of Chinese Medicine, Henan University of Chinese Medicine, Zhengzhou, 450046, China.
| | - Ming-San Miao
- National International Cooperation Base of Chinese Medicine, Henan University of Chinese Medicine, Zhengzhou, 450046, China.
| | - Ya-Gang Song
- National International Cooperation Base of Chinese Medicine, Henan University of Chinese Medicine, Zhengzhou, 450046, China; Academy of Chinese Medicine Science, Henan University of Chinese Medicine, Zhengzhou, 450046, China.
| | - Xiao-Yan Fang
- National International Cooperation Base of Chinese Medicine, Henan University of Chinese Medicine, Zhengzhou, 450046, China.
| | - Jin Zhang
- Academy of Chinese Medicine Science, Henan University of Chinese Medicine, Zhengzhou, 450046, China.
| | - Ya-Nan Zhang
- National International Cooperation Base of Chinese Medicine, Henan University of Chinese Medicine, Zhengzhou, 450046, China.
| | - Jin-Xin Miao
- Academy of Chinese Medicine Science, Henan University of Chinese Medicine, Zhengzhou, 450046, China.
| |
Collapse
|
32
|
Zhao Y, Zhang L, Guo M, Yang H. Taraxasterol suppresses cell proliferation and boosts cell apoptosis via inhibiting GPD2-mediated glycolysis in gastric cancer. Cytotechnology 2021; 73:815-825. [PMID: 34776631 DOI: 10.1007/s10616-021-00499-8] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Accepted: 09/25/2021] [Indexed: 12/26/2022] Open
Abstract
Gastric cancer (GC) is the most common malignant tumor of digestive tract. Taraxasterol (TAX), a kind of phytosterol, has been proved to exert anti-tumor functions in GC. Herein, the current work was carried out to identify the biological role of TAX and molecular mechanisms underlying TAX in the progression of GC. In the present study, CCK-8 assay, Colony formation assay, EDU staining and TUNEL staining were performed to evaluate the malignant behaviors of GC cells. Levels of proliferation and apoptosis-associated proteins were assessed using western blotting analysis. Besides, GPD2 expression in GC cells was presented on CCLE database and the interaction between TAX and GPD2 was obtained from STRING database. The glucose uptake, lactate production, LDH activity, ATP and expressions of glycolysis-associated enzymes were measured to evaluate glycolysis level. Results of the present research revealed that TAX suppressed the proliferative and clone-forming abilities of GC cells and boosted the apoptosis of GC cells. TAX reduced GPD2 expression in GC cells. Furthermore, overexpression of GPD2 reversed the inhibitory effects of TAX on the proliferative and clone-forming abilities of GC cells as well as abolished the promoting effects of TAX on the apoptosis of GC cells. Besides, upregulation of GPD2 abrogated the inhibition of TAX on glycolysis. To conclude, TAX could suppress GC progression via inhibiting GPD2-mediated glycolysis, which helps to develop a promising molecular target for GC therapies.
Collapse
Affiliation(s)
- Yang Zhao
- Department of Surgical Oncology II, General Hospital of Ningxia Medical University, No. 804 Shengli Road, Xingqing District, Yinchuan, 750004 Ningxia China
| | - Li Zhang
- Department of Geriatric Digestive Surgery, The Second Affiliated Hospital of Xi'an Jiaotong University, No. 157 West Five Road, Xi'an, 710004 Shaanxi China
| | - Min Guo
- Health Science Center, Xi'an Jiaotong University, No. 76 West Yanta Road, Xi'an, 710061 Shaanxi China
| | - Haixia Yang
- Department of Traditional Chinese Medicine, The Second Affiliated Hospital of Xi'an Jiaotong University, No. 157 West Five Road, Xi'an, 710004 Shaanxi China
| |
Collapse
|
33
|
Autophagy Regulation on Pyroptosis: Mechanism and Medical Implication in Sepsis. Mediators Inflamm 2021; 2021:9925059. [PMID: 34257519 PMCID: PMC8253640 DOI: 10.1155/2021/9925059] [Citation(s) in RCA: 50] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Accepted: 06/14/2021] [Indexed: 02/07/2023] Open
Abstract
Sepsis is defined as a life-threatening disease involving multiple organ dysfunction caused by dysregulated host responses to infection. To date, sepsis remains a dominant cause of death among critically ill patients. Pyroptosis is a unique form of programmed cell death mediated by the gasdermin family of proteins and causes lytic cell death and release of proinflammatory cytokines. Although there might be some positive aspects to pyroptosis, it is regarded as harmful during sepsis and needs to be restricted. Autophagy was originally characterized as a homeostasis-maintaining mechanism in living cells. In the past decade, its function in negatively modulating pyroptosis and inflammation during sepsis has attracted increased attention. Here, we present a comprehensive review of the regulatory effect of autophagy on pyroptosis during sepsis, including the latest advances in our understanding of the mechanism and signaling pathways involved, as well as the potential therapeutic application in sepsis.
Collapse
|