1
|
Zhao Y, Zhang J, Yan W, Jiang P, Li J, He H, Ma H, Zhang Y, Yang K, Jiang M, Xi X. Folium Hibisci Mutabilis extract suppresses M1 macrophage polarization through mitochondrial function enhancement in murine acute gouty arthritis. Chin Med 2025; 20:28. [PMID: 40022141 PMCID: PMC11869456 DOI: 10.1186/s13020-025-01081-6] [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: 11/10/2024] [Accepted: 02/18/2025] [Indexed: 03/03/2025] Open
Abstract
BACKGROUND Acute gout arthritis (AGA) is a common metabolic joint disease and urgently needs a safer alternative therapy due to the significant side effects from long-term use of primary medications. Folium Hibisci Mutabilis, a traditional medicinal herb, has demonstrated promising therapeutic efficacy in the clinical management of AGA, but its pharmacological mechanisms remain to be elucidated. METHODS Folium Hibisci Mutabili was isolated and refined into the Folium Hibisci Mutabilis Extract (FHME). Then, monosodium urate-induced AGA animal models were applied to identify the anti-inflammatory and analgesic effects of FHME in vivo through various techniques, including ultrasonography, Paw withdrawal thresholds, histological staining, etc. We used RNA-seq, qRT-PCR, ELISA, and flow cytometry to evaluate the efficacy of FHME on M1 polarization. Utilizing transmission electron microscope and oxygen consumption rate examinations in conjunction with Mito-Tracker staining, we observed the effects of FHME on mitochondrial morphology and function. Finally, we employed proteomics analysis, siRNA, qRT-PCR, western blot and other techniques to investigate the underlying mechanism of FHME's actions between the two phenotypes and the key targets. RESULTS We observed a notable reduction in inflammation and pain, as well as the decreased infiltration of inflammatory cells and expression of IL-1β in synovial tissue of AGA mice upon treatment with FHME. FHME suppressed TNF-α, IL-1β, iNOS, and IL-18 expression in BMDM-derived macrophages and inhibited the formation of F4/80+CD86+ cells. Mechanically, FHME protected mitochondrial morphology and stimulated the expression of key oxidative phosphorylation proteins, such as Ubiquinol Cytochrome c Reductase Core Protein I (UQCRC1), UQCRC2, CYCS, and NDUFA4. Additionally, it enhanced the activity of respiratory complex III, recovered cellular aerobic respiration under LPS and MSU induction. FHME lost its effect to downregulate M1 macrophage polarization with the presence of rotenone or si-UQCRC1. Finally, 10 compounds were identified from FHME having potential binding affinity with the UQCRC1 protein. CONCLUSIONS The therapeutic potential of FHME for AGA is associated with the maintenance of mitochondrial function to inhibit M1 macrophage polarization, which is intimately linked to the UQCRC1. Our findings highlight the potential of Folium Hibisci Mutabilis as a safe and effective approach for AGA.
Collapse
Affiliation(s)
- Yichen Zhao
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Jiahui Zhang
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
- Department of Clinical Laboratory, Wuxi Branch of Ruijin Hospital Shanghai Jiao Tong University School of Medicine, Wuxi, 214111, China
| | - Wei Yan
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Ping Jiang
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Juncheng Li
- Wenzhou Medical University, Wenzhou, 325035, China
| | - Haojun He
- Wenzhou Medical University, Wenzhou, 325035, China
| | - Honghong Ma
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Yuxin Zhang
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Kai Yang
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
| | - Min Jiang
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
| | - Xiaobing Xi
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
| |
Collapse
|
2
|
Wang S, Liu W, Wei B, Wang A, Wang Y, Wang W, Gao J, Jin Y, Lu H, Ka Y, Yue Q. Traditional herbal medicine: Therapeutic potential in acute gouty arthritis. JOURNAL OF ETHNOPHARMACOLOGY 2024; 330:118182. [PMID: 38621464 DOI: 10.1016/j.jep.2024.118182] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Revised: 03/27/2024] [Accepted: 04/08/2024] [Indexed: 04/17/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Acute gouty arthritis (AGA) is characterized by a rapid inflammatory reaction caused by the build-up of monosodium urate (MSU) crystals in the tissues surrounding the joints. This condition often associated with hyperuricemia (HUA), is distinguished by its symptoms of intense pain, active inflammation, and swelling of the joints. Traditional approaches in AGA management often fall short of desired outcomes in clinical settings. However, recent ethnopharmacological investigations have been focusing on the potential of Traditional Herbal Medicine (THM) in various forms, exploring their therapeutic impact and targets in AGA treatment. AIM OF THE REVIEW This review briefly summarizes the current potential pharmacological mechanisms of THMs - including active ingredients, extracts, and prescriptions -in the treatment of AGA, and discusses the relevant potential mechanisms and molecular targets in depth. The objective of this study is to offer extensive information and a reference point for the exploration of targeted AGA treatment using THMs. MATERIALS AND METHODS This review obtained scientific publications focused on in vitro and in vivo studies of anti-AGA THMs conducted between 2013 and 2023. The literature was collected from various journals and electronic databases, including PubMed, Elsevier, ScienceDirect, Web of Science, and Google Scholar. The retrieval and analysis of relevant articles were guided by keywords such as "acute gouty arthritis and Chinese herbal medicine," "acute gouty arthritis herbal prescription," "acute gouty arthritis and immune cells," "acute gouty arthritis and inflammation," "acute gouty arthritis and NOD-like receptor thermoprotein domain associated protein 3 (NLRP3)," "acute gouty arthritis and miRNA," and "acute gouty arthritis and oxidative stress." RESULTS We found that AGA has a large number of therapeutic targets, highlighting the effectiveness the potential of THMs in AGA treatment through in vitro and in vivo studies. THMs and their active ingredients can mitigate AGA symptoms through a variety of therapeutic targets, such as influencing macrophage polarization, neutrophils, T cells, natural killer (NK) cells, and addressing factors like inflammation, NLRP3 inflammasome, signaling pathways, oxidative stress, and miRNA multi-target interactions. The anti-AGA properties of THMs, including their active components and prescriptions, were systematically summarized and categorized based on their respective therapeutic targets. CONCLUSION phenolic, flavonoid, terpenoid and alkaloid compounds in THMs are considered the key ingredients to improve AGA. THMs and their active ingredients achieve enhanced efficacy through interactions with multiple targets, of which NLRP3 is a main therapeutic target. Nonetheless, given the intricate composition of traditional Chinese medicine (TCM), additional research is required to unravel the underlying mechanisms and molecular targets through which THMs alleviate AGA.
Collapse
Affiliation(s)
- Siwei Wang
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, 300193, China; National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, 300381, China
| | - Wei Liu
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, 300193, China; National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, 300381, China.
| | - Bowen Wei
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, 300193, China; National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, 300381, China
| | - Aihua Wang
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, 300193, China; National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, 300381, China
| | - Yiwen Wang
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, 300193, China; National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, 300381, China
| | - Wen Wang
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, 300193, China; National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, 300381, China
| | - Jingyue Gao
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, 300193, China; National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, 300381, China
| | - Yue Jin
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, 300193, China; National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, 300381, China
| | - Hang Lu
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, 300193, China; National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, 300381, China
| | - Yuxiu Ka
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, 300193, China; National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, 300381, China
| | - Qingyun Yue
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, 300193, China; National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, 300381, China
| |
Collapse
|
3
|
Zhang S, Li D, Fan M, Yuan J, Xie C, Yuan H, Xie H, Gao H. Mechanism of Reactive Oxygen Species-Guided Immune Responses in Gouty Arthritis and Potential Therapeutic Targets. Biomolecules 2024; 14:978. [PMID: 39199366 PMCID: PMC11353092 DOI: 10.3390/biom14080978] [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: 07/04/2024] [Revised: 08/02/2024] [Accepted: 08/06/2024] [Indexed: 09/01/2024] Open
Abstract
Gouty arthritis (GA) is an inflammatory disease caused by monosodium urate (MSU) crystals deposited in the joint tissues causing severe pain. The disease can recur frequently and tends to form tophus in the joints. Current therapeutic drugs for the acute phase of GA have many side effects and limitations, are unable to prevent recurrent GA attacks and tophus formation, and overall efficacy is unsatisfactory. Therefore, we need to advance research on the microscopic mechanism of GA and seek safer and more effective drugs through relevant targets to block the GA disease process. Current research shows that the pathogenesis of GA is closely related to NLRP3 inflammation, oxidative stress, MAPK, NET, autophagy, and Ferroptosis. However, after synthesizing and sorting out the above mechanisms, it is found that the presence of ROS is throughout almost the entire spectrum of micro-mechanisms of the gout disease process, which combines multiple immune responses to form a large network diagram of complex and tight connections involved in the GA disease process. Current studies have shown that inflammation, oxidative stress, cell necrosis, and pathological signs of GA in GA joint tissues can be effectively suppressed by modulating ROS network-related targets. In this article, on the one hand, we investigated the generative mechanism of ROS network generation and its association with GA. On the other hand, we explored the potential of related targets for the treatment of gout and the prevention of tophus formation, which can provide effective reference ideas for the development of highly effective drugs for the treatment of GA.
Collapse
Affiliation(s)
- Sai Zhang
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu 610075, China; (S.Z.)
- TCM Regulating Metabolic Diseases Key Laboratory of Sichuan Province, Chengdu 610072, China
- Department of Endocrinology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu 610032, China
| | - Daocheng Li
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu 610075, China; (S.Z.)
- TCM Regulating Metabolic Diseases Key Laboratory of Sichuan Province, Chengdu 610072, China
- Department of Endocrinology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu 610032, China
| | - Mingyuan Fan
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu 610075, China; (S.Z.)
- TCM Regulating Metabolic Diseases Key Laboratory of Sichuan Province, Chengdu 610072, China
- Department of Endocrinology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu 610032, China
| | - Jiushu Yuan
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu 610075, China; (S.Z.)
- TCM Regulating Metabolic Diseases Key Laboratory of Sichuan Province, Chengdu 610072, China
- Department of Endocrinology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu 610032, China
| | - Chunguang Xie
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu 610075, China; (S.Z.)
- TCM Regulating Metabolic Diseases Key Laboratory of Sichuan Province, Chengdu 610072, China
- Department of Endocrinology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu 610032, China
| | - Haipo Yuan
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu 610075, China; (S.Z.)
- TCM Regulating Metabolic Diseases Key Laboratory of Sichuan Province, Chengdu 610072, China
- Department of Endocrinology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu 610032, China
| | - Hongyan Xie
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu 610075, China; (S.Z.)
- TCM Regulating Metabolic Diseases Key Laboratory of Sichuan Province, Chengdu 610072, China
- Department of Endocrinology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu 610032, China
| | - Hong Gao
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu 610075, China; (S.Z.)
- TCM Regulating Metabolic Diseases Key Laboratory of Sichuan Province, Chengdu 610072, China
- Department of Endocrinology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu 610032, China
| |
Collapse
|
4
|
Chen X, Deng G, Chen K, Chen Y, Ye W, Sun P. Targeting the NLRP3 inflammasome in psoriasis. Int J Dermatol 2024; 63:844-851. [PMID: 38345734 DOI: 10.1111/ijd.17073] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Revised: 01/16/2024] [Accepted: 01/19/2024] [Indexed: 07/05/2024]
Abstract
The NLRP3 inflammasome, a complex consisting of the nucleotide-binding oligomerization domain, leucine-rich repeat, and pyrin domain-containing protein 3, has emerged as a critical mediator of pathological inflammation and a significant therapeutic target for various inflammatory diseases. Psoriasis, a chronic inflammatory skin condition without a definitive cure, has shown promising results in animal models through the inhibition of the NLRP3 inflammasome. This review aims to explore the development of the NLRP3 inflammasome in psoriasis and the molecular mechanisms responsible for its inhibition by natural products and small molecules currently being developed for psoriasis treatment. Furthermore, we are examining clinical trials using agents that block the NLRP3 pathway for the treatment of psoriasis. This study is timely to provide a new perspective on managing psoriasis.
Collapse
Affiliation(s)
- Xiuhui Chen
- Key Laboratory of Precision Pharmacy and Pharmaceutical Basic Research, Dongguan, China
- Department of Pharmacy, Dongguan Eighth People's Hospital, Dongguan Children's Hospital Affiliated to Guangdong Medical University, Dongguan, China
| | - Guoliang Deng
- Key Laboratory of Precision Pharmacy and Pharmaceutical Basic Research, Dongguan, China
- Department of Pharmacy, Dongguan Eighth People's Hospital, Dongguan Children's Hospital Affiliated to Guangdong Medical University, Dongguan, China
| | - Kaifeng Chen
- Key Laboratory of Molecular Target & Clinical Pharmacology and the State & NMPA Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences & the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Yanhong Chen
- Department of Pharmacy, Baoan Central Hospital of Shenzhen, Shenzhen, China
| | - Weijun Ye
- Key Laboratory of Precision Pharmacy and Pharmaceutical Basic Research, Dongguan, China
- Department of Pharmacy, Dongguan Eighth People's Hospital, Dongguan Children's Hospital Affiliated to Guangdong Medical University, Dongguan, China
| | - Ping Sun
- Key Laboratory of Molecular Target & Clinical Pharmacology and the State & NMPA Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences & the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| |
Collapse
|
5
|
Chen H, Yue H, Yan Y, Wu N, Wu D, Sun P, Hu W, Yang Z. Design, synthesis and biological evaluation of tanshinone IIA derivatives as NLRP3 inflammasome inhibitors. Bioorg Med Chem Lett 2024; 104:129725. [PMID: 38555073 DOI: 10.1016/j.bmcl.2024.129725] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Revised: 03/27/2024] [Accepted: 03/28/2024] [Indexed: 04/02/2024]
Abstract
Natural product structures have long provided valuable pharmacophores and even candidates for drug discovery. Tanshinone scaffold showed moderately inhibitory activity in NLRP3 inflammasome/IL-1β pathway. Herein, we designed a series of derivatives on different regions of Tanshinone IIA (TNA) scaffold. The biological evaluation identified compound T10, a scaffold hybrid of TNA and salicylic acid, as a potent NLRP3 inflammasome inhibitor. Mechanistically, T10 inhibits the production of ROS and prevents NLRP3 inflammasome-dependent IL-1β production. In addition, treatment with T10 significantly attenuated inflammatory response in DSS-induced peritonitis. Our work describes a potential tanshinone-based derivative, which needs to be further structurally optimized as NLRP3 inflammasome inhibitors for treating inflammatory disorders.
Collapse
Affiliation(s)
- Hao Chen
- Key Laboratory of Molecular Target, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou, Guangdong 511436, China
| | - Hu Yue
- Key Laboratory of Molecular Target, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou, Guangdong 511436, China
| | - Yuyun Yan
- Key Laboratory of Molecular Target, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou, Guangdong 511436, China
| | - Nannan Wu
- Department of Pharmacy, Central People's Hospital of Zhanjiang, Zhanjiang, Guangdong 524045, China
| | - Dan Wu
- Key Laboratory of Molecular Target, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou, Guangdong 511436, China
| | - Ping Sun
- Key Laboratory of Molecular Target, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou, Guangdong 511436, China.
| | - Wenhui Hu
- Key Laboratory of Molecular Target, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou, Guangdong 511436, China.
| | - Zhongjin Yang
- Key Laboratory of Molecular Target, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou, Guangdong 511436, China.
| |
Collapse
|
6
|
Fan W, Liu C, Chen D, Xu C, Qi X, Zhang A, Zhu X, Liu Y, Wang L, Hao L, Liu WT, Hu L. Ozone alleviates MSU-induced acute gout pain via upregulating AMPK/GAS6/MerTK/SOCS3 signaling pathway. J Transl Med 2023; 21:890. [PMID: 38066599 PMCID: PMC10704676 DOI: 10.1186/s12967-023-04769-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Accepted: 11/28/2023] [Indexed: 12/18/2023] Open
Abstract
BACKGROUND Gout pain seriously affects the quality of patients' life. There is still no effective treatment. The inflammatory response is the main mechanism of gout. Here, we found that ozone can reduce the inflammatory reaction in the joints of gouty mice and relieve gout pain, and we further explore its protective mechanism. METHODS MSU was used to establish the gouty mice model. Nociception was assessed by Von Frey hairs. Cell signaling assays were performed by western blotting and immunohistochemistry. The mouse leukemia cells of monocyte macrophage line RAW264.7 were cultured to investigate the effects of ozone administration on macrophage. RESULTS Ozone reduced inflammation, relieved gout pain and improved the paw mean intensity and duty cycle of the gouty mice. Ozone increased the phosphorylation of AMP-activated protein kinase (AMPK), induced suppressor of cytokine signaling 3 (SOCS3) expression and inhibited metallopeptidase 9 (MMP9) expression. In vivo, ozone activated AMPK to induce Gas6 release, and upregulated MerTK/SOCS3 signaling pathway to reduce inflammation in mouse macrophage line RAW264.7. Inhibitors of AMPK and MerTK, respectively abolished the analgesic and anti-inflammatory effects of ozone in vivo and in vitro. Gas6 knockout cancelled the protectively effects of ozone on gout pain and the paw mean intensity and duty cycle of gouty mice. Additionally, the level of Gas6 and protein S in plasma of patients with hyperuricemia was significantly higher than that of healthy contrast group. CONCLUSION Ozone reduces inflammation and alleviates gout pain by activating AMPK to up-regulate Gas6/MerTK/SOCS3 signaling pathway.
Collapse
Affiliation(s)
- Wen Fan
- Department of Pharmacology, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, 211166, China
| | - Chong Liu
- Department of Pharmacology, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, 211166, China
| | - Dacai Chen
- The Fourth Affiliated Hospital of Nantong University, Jiangsu, China
- Yancheng Ruikang Hospital, Jiangsu, 224000, China
| | - Chenjie Xu
- Department of Anesthesiology and Pain, Nanjing First Hospital, Nanjing Medical University, Nanjing, 210006, Jiangsu, China
| | - Xiuting Qi
- Department of Pharmacology, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, 211166, China
| | - Ailin Zhang
- Nanjing University of Chinese Medicine Institute of Literature in Chinese Medicine, Jiangsu, 224000, China
| | - Xuexian Zhu
- Department of Pharmacology, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, 211166, China
| | - Yujie Liu
- Department of Pharmacology, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, 211166, China
| | - Lei Wang
- Department of Pharmacology, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, 211166, China
| | - Lanxiang Hao
- The Yancheng Clinical College of Xuzhou Medical University, The First People's Hospital of Yancheng, Jiangsu, 224005, China.
| | - Wen-Tao Liu
- Department of Pharmacology, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, 211166, China.
| | - Liang Hu
- Department of Pharmacology, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, 211166, China.
| |
Collapse
|
7
|
Qiu C, Zhang JZ, Wu B, Xu CC, Pang HH, Tu QC, Lu YQ, Guo QY, Xia F, Wang JG. Advanced application of nanotechnology in active constituents of Traditional Chinese Medicines. J Nanobiotechnology 2023; 21:456. [PMID: 38017573 PMCID: PMC10685519 DOI: 10.1186/s12951-023-02165-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Accepted: 10/16/2023] [Indexed: 11/30/2023] Open
Abstract
Traditional Chinese Medicines (TCMs) have been used for centuries for the treatment and management of various diseases. However, their effective delivery to targeted sites may be a major challenge due to their poor water solubility, low bioavailability, and potential toxicity. Nanocarriers, such as liposomes, polymeric nanoparticles, inorganic nanoparticles and organic/inorganic nanohybrids based on active constituents from TCMs have been extensively studied as a promising strategy to improve the delivery of active constituents from TCMs to achieve a higher therapeutic effect with fewer side effects compared to conventional formulations. This review summarizes the recent advances in nanocarrier-based delivery systems for various types of active constituents of TCMs, including terpenoids, polyphenols, alkaloids, flavonoids, and quinones, from different natural sources. This review covers the design and preparation of nanocarriers, their characterization, and in vitro/vivo evaluations. Additionally, this review highlights the challenges and opportunities in the field and suggests future directions for research. Nanocarrier-based delivery systems have shown great potential in improving the therapeutic efficacy of TCMs, and this review may serve as a comprehensive resource to researchers in this field.
Collapse
Affiliation(s)
- Chong Qiu
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao-Di Herbs, Artemisinin Research Center, and Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, China
| | - Jun Zhe Zhang
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao-Di Herbs, Artemisinin Research Center, and Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, China
| | - Bo Wu
- Department of Traditional Chinese Medical Science, Sixth Medical Center of the Chinese PLA General Hospital, Beijing, 100037, China
| | - Cheng Chao Xu
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao-Di Herbs, Artemisinin Research Center, and Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, China
| | - Huan Huan Pang
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao-Di Herbs, Artemisinin Research Center, and Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, China
| | - Qing Chao Tu
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao-Di Herbs, Artemisinin Research Center, and Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, China
| | - Yu Qian Lu
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao-Di Herbs, Artemisinin Research Center, and Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, China
| | - Qiu Yan Guo
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao-Di Herbs, Artemisinin Research Center, and Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, China.
| | - Fei Xia
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao-Di Herbs, Artemisinin Research Center, and Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, China.
| | - Ji Gang Wang
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao-Di Herbs, Artemisinin Research Center, and Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, China.
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117600, Singapore.
| |
Collapse
|
8
|
Zhong C, Yang J, Deng K, Lang X, Zhang J, Li M, Qiu L, Zhong G, Yu J. Tiliroside Attenuates NLRP3 Inflammasome Activation in Macrophages and Protects against Acute Lung Injury in Mice. Molecules 2023; 28:7527. [PMID: 38005247 PMCID: PMC10673355 DOI: 10.3390/molecules28227527] [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: 09/08/2023] [Revised: 10/31/2023] [Accepted: 11/08/2023] [Indexed: 11/26/2023] Open
Abstract
The Nod-like receptor family PYRIN domain containing 3 (NLRP3) inflammasome is a multiprotein signaling complex that plays a pivotal role in innate immunity, and the dysregulated NLRP3 inflammasome activation is implicated in various diseases. Tiliroside is a natural flavonoid in multiple medicinal and dietary plants with known anti-inflammatory activities. However, its role in regulating NLRP3 inflammasome activation and NLRP3-related disease has not been evaluated. Herein, it was demonstrated that tiliroside is inhibitory in activating the NLRP3 inflammasome in macrophages. Mechanistically, tiliroside promotes AMP-activated protein kinase (AMPK) activation, thereby leading to ameliorated mitochondrial damage as evidenced by the reduction of mitochondrial reactive oxygen species (ROS) production and the improvement of mitochondrial membrane potential, which is accompanied by attenuated NLRP3 inflammasome activation in macrophages. Notably, tiliroside potently attenuated lipopolysaccharide (LPS)-induced acute lung injury in mice, which has been known to be NLRP3 inflammasome dependent. For the first time, this study identified that tiliroside is an NLRP3 inflammasome inhibitor and may represent a potential therapeutic agent for managing NLRP3-mediated inflammatory disease.
Collapse
Affiliation(s)
- Chao Zhong
- Center for Translational Medicine, College of Traditional Chinese Medicine, Jiangxi University of Chinese Medicine, Nanchang 330004, China
| | - Jing Yang
- Center for Translational Medicine, College of Traditional Chinese Medicine, Jiangxi University of Chinese Medicine, Nanchang 330004, China
| | - Keke Deng
- Center for Translational Medicine, College of Traditional Chinese Medicine, Jiangxi University of Chinese Medicine, Nanchang 330004, China
| | - Xiaoya Lang
- Center for Translational Medicine, College of Traditional Chinese Medicine, Jiangxi University of Chinese Medicine, Nanchang 330004, China
| | - Jiangtao Zhang
- Center for Translational Medicine, College of Traditional Chinese Medicine, Jiangxi University of Chinese Medicine, Nanchang 330004, China
| | - Min Li
- Center for Traditional Chinese Medicine Resources and Ethnic Medicine, Jiangxi University of Chinese Medicine, Nanchang 330004, China
| | - Liang Qiu
- Center for Translational Medicine, College of Traditional Chinese Medicine, Jiangxi University of Chinese Medicine, Nanchang 330004, China
| | - Guoyue Zhong
- Center for Traditional Chinese Medicine Resources and Ethnic Medicine, Jiangxi University of Chinese Medicine, Nanchang 330004, China
| | - Jun Yu
- Department of Cardiovascular Sciences, Center for Metabolic Disease Research, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA
| |
Collapse
|
9
|
Dang K, Zhang N, Gao H, Wang G, Liang H, Xue M. Influence of intestinal microecology in the development of gout or hyperuricemia and the potential therapeutic targets. Int J Rheum Dis 2023; 26:1911-1922. [PMID: 37606177 DOI: 10.1111/1756-185x.14888] [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: 01/07/2023] [Revised: 08/08/2023] [Accepted: 08/09/2023] [Indexed: 08/23/2023]
Abstract
Gout and hyperuricemia are common metabolic diseases. Patients with purine metabolism disorder and/or decreased uric acid excretion showed increased uric acid levels in the blood. The increase of uric acid in the blood leads to the deposition of urate crystals in tissues, joints, and kidneys, and causes gout. Recent studies have revealed that imbalance of the intestinal microecology is closely related to the occurrence and development of hyperuricemia and gout. Disorder of the intestinal flora often occurs in patients with gout, and high purine and high fructose may induce the disorder of intestinal flora. Short-chain fatty acids and endotoxins produced by intestinal bacteria are closely related to the inflammatory response of gout. This article summarizes the characteristics of intestinal microecology in patients or animal models with hyperuricemia or gout, and explores the relationship between intestinal microecology and gout or hyperuricemia from the aspect of the intestinal barrier, intestinal microorganisms, intestinal metabolites, and intestinal immune system. We also review the current status of hyperuricemia treatment by targeting intestinal microecology.
Collapse
Affiliation(s)
- Kai Dang
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Qingdao University, Qingdao, China
| | - Nan Zhang
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Qingdao University, Qingdao, China
| | - Haiqi Gao
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Qingdao University, Qingdao, China
| | - Guifa Wang
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Qingdao University, Qingdao, China
| | - Hui Liang
- Department of Human Nutrition, College of Public Health, Qingdao University, Qingdao, China
| | - Meilan Xue
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Qingdao University, Qingdao, China
| |
Collapse
|
10
|
Torunoglu ST, Zajda A, Tampio J, Markowicz-Piasecka M, Huttunen KM. Metformin derivatives - Researchers' friends or foes? Biochem Pharmacol 2023; 215:115743. [PMID: 37591450 DOI: 10.1016/j.bcp.2023.115743] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Revised: 08/09/2023] [Accepted: 08/09/2023] [Indexed: 08/19/2023]
Abstract
Metformin has been used for ages to treat diabetes mellitus due to its safety profile and low cost. However, metformin has variable pharmacokinetics in patients, and due to its poor oral absorption, the therapeutic doses are relatively high, causing unpleasant gastrointestinal adverse effects. Therefore, novel derivatives of metformin have been synthesized during the past decades. Particularly, after the mid-2000 s, when organic cation transporters were identified as the main metformin carriers, metformin derivatives have been under intensive investigation. Nevertheless, due to the biguanide structure, derivatives of metformin have been challenging to synthesize. Moreover, the mechanisms of metformin's action are not fully understood to date, and since it has multifunctional properties, the interests have switched to re-purposing for other diseases. Indeed, metformin derivatives have been demonstrated in many cases to be more effective than metformin itself and have the potential to be used in different diseases, including several types of cancers and neurodegenerative diseases. On the other hand, the pleiotropic nature of metformin and its derivatives can also create challenges. Not all properties are fit for all diseases. In this review, the history of the development of metformin-like compounds is summarized, and insights into their potential for future drug discovery are discussed.
Collapse
Affiliation(s)
- Sema Tuna Torunoglu
- School of Pharmacy, Faculty of Health Sciences, University of Eastern Finland, P.O. Box 1627, FI-70211 Kuopio, Finland.
| | - Agnieszka Zajda
- Laboratory of Bioanalysis, Department of Pharmaceutical Chemistry, Drug Analysis and Radiopharmacy, Medical University of Lodz, ul. Muszyńskiego 1, 90-151 Lodz, Poland
| | - Janne Tampio
- School of Pharmacy, Faculty of Health Sciences, University of Eastern Finland, P.O. Box 1627, FI-70211 Kuopio, Finland
| | | | - Kristiina M Huttunen
- School of Pharmacy, Faculty of Health Sciences, University of Eastern Finland, P.O. Box 1627, FI-70211 Kuopio, Finland.
| |
Collapse
|
11
|
Zhao J, Liu H, Hong Z, Luo W, Mu W, Hou X, Xu G, Fang Z, Ren L, Liu T, Wen J, Shi W, Wei Z, Yang Y, Zou W, Zhao J, Xiao X, Bai Z, Zhan X. Tanshinone I specifically suppresses NLRP3 inflammasome activation by disrupting the association of NLRP3 and ASC. Mol Med 2023; 29:84. [PMID: 37400760 DOI: 10.1186/s10020-023-00671-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2022] [Accepted: 05/29/2023] [Indexed: 07/05/2023] Open
Abstract
BACKGROUND Abnormal activation of NLRP3 inflammasome is related to a series of inflammatory diseases, including type 2 diabetes, gouty arthritis, non-alcoholic steatohepatitis (NASH), and neurodegenerative disorders. Therefore, targeting NLRP3 inflammasome is regarded as a potential therapeutic strategy for many inflammatory diseases. A growing number of studies have identified tanshinone I (Tan I) as a potential anti-inflammatory agent because of its good anti-inflammatory activity. However, its specific anti-inflammatory mechanism and direct target are unclear and need further study. METHODS IL-1β and caspase-1 were detected by immunoblotting and ELISA, and mtROS levels were measured by flow cytometry. Immunoprecipitation was used to explore the interaction between NLRP3, NEK7 and ASC. In a mouse model of LPS-induced septic shock, IL-1β levels in peritoneal lavage fluid and serum were measured by ELISA. Liver inflammation and fibrosis in the NASH model were analyzed by HE staining and immunohistochemistry. RESULTS Tan I inhibited the activation of NLRP3 inflammasome in macrophages, but had no effect on the activation of AIM2 or NLRC4 inflammasome. Mechanistically, Tan I inhibited NLRP3 inflammasome assembly and activation by targeting NLRP3-ASC interaction. Furthermore, Tan I exhibited protective effects in mouse models of NLRP3 inflammasome-mediated diseases, including septic shock and NASH. CONCLUSIONS Tan I specifically suppresses NLRP3 inflammasome activation by disrupting the association of NLRP3 and ASC, and exhibits protective effects in mouse models of LPS-induced septic shock and NASH. These findings suggest that Tan I is a specific NLRP3 inhibitor and may be a promising candidate for treating NLRP3 inflammasome-related diseases.
Collapse
Affiliation(s)
- Jia Zhao
- Department of Hepatology, the Fifth Medical Center of Chinese PLA General Hospital, Beijing, 100039, China
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
- School of Pharmacy, North SiChuan Medical College, Nanchong, 637000, China
| | - Hongbin Liu
- Department of Hepatology, the Fifth Medical Center of Chinese PLA General Hospital, Beijing, 100039, China
- Department of Pharmacy, Hebei Key Laboratory of Neuropharmacology, Hebei North University, Zhangjiakou, 075000, China
| | - Zhixian Hong
- Department of Hepatology, the Fifth Medical Center of Chinese PLA General Hospital, Beijing, 100039, China
| | - Wei Luo
- Department of Hepatology, the Fifth Medical Center of Chinese PLA General Hospital, Beijing, 100039, China
| | - Wenqing Mu
- Department of Hepatology, the Fifth Medical Center of Chinese PLA General Hospital, Beijing, 100039, China
| | - Xiaorong Hou
- Department of Hepatology, the Fifth Medical Center of Chinese PLA General Hospital, Beijing, 100039, China
| | - Guang Xu
- Department of Hepatology, the Fifth Medical Center of Chinese PLA General Hospital, Beijing, 100039, China
| | - Zhie Fang
- Department of Hepatology, the Fifth Medical Center of Chinese PLA General Hospital, Beijing, 100039, China
| | - Lutong Ren
- Department of Hepatology, the Fifth Medical Center of Chinese PLA General Hospital, Beijing, 100039, China
| | - Tingting Liu
- Department of Hepatology, the Fifth Medical Center of Chinese PLA General Hospital, Beijing, 100039, China
| | - Jincai Wen
- Department of Hepatology, the Fifth Medical Center of Chinese PLA General Hospital, Beijing, 100039, China
| | - Wei Shi
- Department of Hepatology, the Fifth Medical Center of Chinese PLA General Hospital, Beijing, 100039, China
| | - Ziying Wei
- Department of Hepatology, the Fifth Medical Center of Chinese PLA General Hospital, Beijing, 100039, China
| | - Yongping Yang
- Department of Hepatology, the Fifth Medical Center of Chinese PLA General Hospital, Beijing, 100039, China
| | - Wenjun Zou
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Jun Zhao
- Department of Hepatology, the Fifth Medical Center of Chinese PLA General Hospital, Beijing, 100039, China.
| | - Xiaohe Xiao
- Department of Hepatology, the Fifth Medical Center of Chinese PLA General Hospital, Beijing, 100039, China.
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China.
| | - Zhaofang Bai
- Department of Hepatology, the Fifth Medical Center of Chinese PLA General Hospital, Beijing, 100039, China.
| | - Xiaoyan Zhan
- Department of Hepatology, the Fifth Medical Center of Chinese PLA General Hospital, Beijing, 100039, China.
| |
Collapse
|
12
|
Crocin Attenuates NLRP3 Inflammasome Activation by Inhibiting Mitochondrial Reactive Oxygen Species and Ameliorates Monosodium Urate-Induced Mouse Peritonitis. Curr Issues Mol Biol 2023; 45:2090-2104. [PMID: 36975504 PMCID: PMC10047758 DOI: 10.3390/cimb45030134] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Revised: 02/27/2023] [Accepted: 03/01/2023] [Indexed: 03/08/2023] Open
Abstract
Crocin is a hydrophilic carotenoid pigment found in the stigma of Crocus sativus or the fruit of Gardenia jasminoides. In this study, we investigated the effects of Crocin on the activation of the nucleotide-binding oligomerization domain, leucine-rich repeat, and pyrin domain containing 3 (NLRP3) inflammasome in J774A.1 murine macrophage cells and monosodium urate (MSU)-induced peritonitis. Crocin significantly inhibited Nigericin-, adenosine triphosphate (ATP)-, MSU-induced interleukin (IL)-1β secretion, and caspase-1 cleavage without affecting pro-IL-1β and pro-caspase-1. Crocin also suppressed gasdermin-D cleavage and lactate dehydrogenase release and enhanced cell viability, indicating that Crocin reduces pyroptosis. Similar effects were observed in primary mouse macrophages. However, Crocin did not affect poly(dA:dT)-induced absent in melanoma 2 (AIM2) and muramyl dipeptide-induced NLRP1 inflammasomes. Crocin decreased Nigericin-induced oligimerization and the speck formation of apoptosis-associated speck-like protein containing a caspase recruitment domain (ASC). Crocin also dramatically alleviated the ATP-induced production of mitochondrial reactive oxygen species (mtROS). Finally, Crocin ameliorated the MSU-induced production of IL-1β and IL-18 and the recruitment of neutrophils during peritoneal inflammation. These results suggest that Crocin suppresses NLRP3 inflammasome activation by blocking mtROS production and ameliorates MSU-induced mouse peritonitis. Thus, Crocin may have therapeutic potential in various NLRP3 inflammasome-related inflammatory diseases.
Collapse
|
13
|
Li J, Sheng H, Wang Y, Lai Z, Wang Y, Cui S. Scaffold Hybrid of the Natural Product Tanshinone I with Piperidine for the Discovery of a Potent NLRP3 Inflammasome Inhibitor. J Med Chem 2023; 66:2946-2963. [PMID: 36786612 DOI: 10.1021/acs.jmedchem.2c01967] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/15/2023]
Abstract
Natural products provide inspiration and have proven to be the most valuable source for drug discovery. Herein, we report a scaffold hybrid strategy of Tanshinone I for the discovery of NLRP3 inflammasome inhibitors. 36 compounds were designed and synthesized, and the cheminformatic analyses showed that these compounds occupy a unique chemical space. The biological evaluation identified compounds 5j, 12a, and 12d as NLRP3 inflammasome inhibitors with significant potency, selectivity, and drug-likeness. Mechanistic studies revealed that these Tanshinone I derivatives could inhibit the degradation of the protein NLRP3 and block the oligomerization of NLRP3-induced apoptosis-associated speck-like proteins, thus inhibiting NLRP3 inflammasome activation. In addition, the water solubility, in vitro metabolic stability, and oral bioavailability of these compounds were also greatly improved compared to Tanshinone I. Therefore, this protocol provides a new structural evolution of Tanshinone I and a new class of potent NLRP3 inflammasome inhibitors.
Collapse
Affiliation(s)
- Jiaming Li
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Hongda Sheng
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Yingchao Wang
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Zhencheng Lai
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Yi Wang
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Sunliang Cui
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| |
Collapse
|
14
|
Jiang X, Lu Z, Zhang Q, Yu J, Han D, Liu J, Li P, Li F. Osthole: A potential AMPK agonist that inhibits NLRP3 inflammasome activation by regulating mitochondrial homeostasis for combating rheumatoid arthritis. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2023; 110:154640. [PMID: 36608498 DOI: 10.1016/j.phymed.2022.154640] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Revised: 12/16/2022] [Accepted: 12/31/2022] [Indexed: 06/17/2023]
Abstract
BACKGROUND Osthole (OST), a characteristic coumarin compound in Angelicae pubescentis radix (APR), has shown potent efficacy in the treatment of rheumatoid arthritis (RA), but its specific targets and potential mechanism are limited. PURPOSE This study aimed to explore the potential targets and molecular mechanisms of OST against RA using computer-assisted techniques in combination with RA fibroblast-like synoviocytes (FLS) inflammation model and CIA rat model. METHODS Network pharmacology and molecular docking were applied to initially predict the potential targets of OST for the treatment of RA. Thereafter, TNFα was used to stimulate FLS to build an in vitro model of inflammation, combined with RNA-seq technology and molecular biology such as qPCR to investigate the anti-inflammatory effects and related mechanisms of OST. Finally, the anti-RA effect of OST was demonstrated by establishing a CIA rat model. RESULTS The network model results showed that the anti-RA effect of OST was mainly related to its anti-inflammatory effect, and AMPK was identified as a potential target for the potency of OST. In the TNFα-induced FLS cells, OST inhibited the secretion of FLS inflammatory factors, which was attributed to the ability of OST to activate AMPK to inhibit the activation of the NLRP3 inflammasome. Further, it was observed that the activation of AMPK by OST facilitated mitochondrial biogenesis, and corrected abnormal mitochondrial dynamics in FLS, which was favoured to the restoration of mitochondrial homeostasis, and further promoted the occurrence of apoptosis and the decrease of ROS in FLS. Consistent with in vivo studies, administration of OST significantly improved joint deformity and toe erythema, reduced arthritis index scores and inhibited synovial inflammation in CIA rats. CONCLUSION Our study proposed for the first time that AMPK, served as a potential target of OST, positively participated in the anti-RA therapeutic effect of OST. By regulating mitochondrial homeostasis and function, OST can effectively inhibit the activation of inflammasome and the secretion of inflammatory factors in vitro and in vivo, and finally achieve beneficial effects in the treatment of RA, which provides support and greater possibility to make further efforts on pharmacological research and clinical application of OST.
Collapse
Affiliation(s)
- Xiaoli Jiang
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, No. 24 Tongjia Lane, Nanjing 210009, China
| | - Zhuojian Lu
- College of Pharmacy, Xinjiang Medical University, Urumqi 830011, China
| | - Qian Zhang
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, No. 24 Tongjia Lane, Nanjing 210009, China
| | - Jialin Yu
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, No. 24 Tongjia Lane, Nanjing 210009, China
| | - Dong Han
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, No. 24 Tongjia Lane, Nanjing 210009, China
| | - Jinhong Liu
- College of Pharmacy, Xinjiang Medical University, Urumqi 830011, China
| | - Ping Li
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, No. 24 Tongjia Lane, Nanjing 210009, China.
| | - Fei Li
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, No. 24 Tongjia Lane, Nanjing 210009, China; College of Pharmacy, Xinjiang Medical University, Urumqi 830011, China.
| |
Collapse
|
15
|
Lee JH, Kang HI, Kim S, Ahn YB, Kim H, Hong JK, Baik JY. NAD + supplementation improves mAb productivity in CHO cells via a glucose metabolic shift. Biotechnol J 2023; 18:e2200570. [PMID: 36717516 DOI: 10.1002/biot.202200570] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Revised: 12/22/2022] [Accepted: 01/19/2023] [Indexed: 02/01/2023]
Abstract
Aerobic glycolysis and its by-product lactate accumulation are usually associated with adverse culture phenotypes such as poor cell viability and productivity. Due to the lack of knowledge on underlying mechanisms and accompanying biological processes, the regulation of aerobic glycolysis has been an ongoing challenge in culture process development for therapeutic protein productivity. Nicotinamide adenine dinucleotide (NAD+ ), a coenzyme and co-substrate in energy metabolism, promotes the conversion of inefficient glycolysis into an efficient oxidative phosphorylation (OXPHOS) pathway. However, the effect of NAD+ on Chinese hamster ovary (CHO) cells for biopharmaceutical production has not been reported yet. In this work, we aimed to elucidate the influence of NAD+ on cell culture performance by examining metabolic shifts and mAb productivity. The supplementation of NAD+ increased the intracellular concentration of NAD+ and promoted SIRT3 expression. Antibody titer and the specific productivity in the growth phase were improved by up to 1.82- and 1.88-fold, respectively, with marginal restrictions on cell growth. NAD+ significantly reduced the accumulation of reactive oxygen species (ROS) and the lactate yield from glucose, determined by lactate accumulation versus glucose consumption (YLAC/GLC ). In contrast, OXPHOS capacity and amino acid consumption rate increased substantially. Collectively, these results suggest that NAD+ contributes to improving therapeutic protein productivity in bioprocessing via inducing an energy metabolic shift.
Collapse
Affiliation(s)
- Ji Hwan Lee
- Department of Biological Sciences and Bioengineering, Inha University, Incheon, South Korea
| | - Hye-Im Kang
- Department of Biological Sciences and Bioengineering, Inha University, Incheon, South Korea
| | - Suheon Kim
- Department of Biological Sciences and Bioengineering, Inha University, Incheon, South Korea
| | - Yeong Bin Ahn
- Division of Biological Science and Technology, Yonsei University, Wonju, Gangwon-do, Republic of Korea
| | - Hagyeong Kim
- Department of Biological Sciences and Bioengineering, Inha University, Incheon, South Korea
| | - Jong Kwang Hong
- Division of Biological Science and Technology, Yonsei University, Wonju, Gangwon-do, Republic of Korea
| | - Jong Youn Baik
- Department of Biological Sciences and Bioengineering, Inha University, Incheon, South Korea
| |
Collapse
|
16
|
Chen Y, Chen X, Liang S, Ou Y, Lin G, Hua L, Wu X, Zhou Y, Liu Z, Cai H, Yang Z, Hu W, Sun P. Chlorquinaldol inhibits the activation of nucleotide-binding oligomerization domain-like receptor family pyrin domain-containing protein 3 inflammasome and ameliorates imiquimod-induced psoriasis-like dermatitis in mice. Chem Biol Interact 2022; 365:110122. [PMID: 36002070 DOI: 10.1016/j.cbi.2022.110122] [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: 03/01/2022] [Revised: 07/14/2022] [Accepted: 08/17/2022] [Indexed: 11/03/2022]
Abstract
Psoriasis is a common chronic autoinflammatory/autoimmune skin disease associated with elevated pro-inflammatory cytokines. The pivotal role of interleukin (IL)-1β and nucleotide-binding oligomerization domain-like receptor family pyrin domain-containing protein 3 (NLRP3) inflammasome in the pathogenesis of psoriasis has been widely described. Accordingly, the suppression of NLRP3-dependent IL-1β release is a potential therapy for psoriasis. Repurposing marketed drugs is a strategy for identifying new inhibitors of NLRP3 inflammasome activation. Herein, chlorquinaldol (CQD), a historic antimicrobial agent used as a topical treatment for skin and vaginal infections, was found to have a distinct effect by inhibiting NLRP3 inflammasome activation at concentrations ranging from 2 to 6 μM. CQD significantly suppressed apoptosis-associated speck-like protein containing a caspase-recruitment domain (ASC) oligomerization, NLRP3-ASC interaction, and pyroptosis in macrophages. The levels of cleaved IL-1β and caspase-1 were reduced by CQD in the cell lysates of macrophages, suggesting that CQD acted on upstream of pore formation in the cell membrane. Mechanistically, CQD reduced mitochondrial reactive oxygen species production but did not affect the nuclear factor-κB (NF-κB) pathway. Intraperitoneal administration of CQD (15 mg/kg) for 6 days was found to improve the skin lesions in the imiquimod-induced psoriatic mouse model (male C57BL/6 mice), while secretion of pro-inflammatory cytokines (IL-17 and IL-1β) and keratinocyte proliferation were significantly suppressed by CQD. In conclusion, CQD exerted inhibitory effects on NLRP3 inflammasome activation in macrophages and decreased the severity of psoriatic response in vivo. Such findings indicate that the repurposing of the old drug, CQD, is a potential pharmacological approach for the treatment of psoriasis and other NLRP3-driven diseases.
Collapse
Affiliation(s)
- Yanhong Chen
- Key Laboratory of Molecular Target & Clinical Pharmacology and the State & NMPA Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences & the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, 511436, PR China
| | - Xiuhui Chen
- Key Laboratory of Molecular Target & Clinical Pharmacology and the State & NMPA Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences & the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, 511436, PR China
| | - Shuli Liang
- Key Laboratory of Molecular Target & Clinical Pharmacology and the State & NMPA Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences & the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, 511436, PR China
| | - Yitao Ou
- Key Laboratory of Molecular Target & Clinical Pharmacology and the State & NMPA Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences & the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, 511436, PR China
| | - Geng Lin
- Key Laboratory of Molecular Target & Clinical Pharmacology and the State & NMPA Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences & the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, 511436, PR China
| | - Lei Hua
- Key Laboratory of Molecular Target & Clinical Pharmacology and the State & NMPA Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences & the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, 511436, PR China
| | - Xinyi Wu
- Key Laboratory of Molecular Target & Clinical Pharmacology and the State & NMPA Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences & the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, 511436, PR China
| | - Yinghua Zhou
- Key Laboratory of Molecular Target & Clinical Pharmacology and the State & NMPA Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences & the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, 511436, PR China
| | - Zhuorong Liu
- Key Laboratory of Molecular Target & Clinical Pharmacology and the State & NMPA Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences & the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, 511436, PR China
| | - Haowei Cai
- Key Laboratory of Molecular Target & Clinical Pharmacology and the State & NMPA Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences & the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, 511436, PR China
| | - Zhongjin Yang
- Key Laboratory of Molecular Target & Clinical Pharmacology and the State & NMPA Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences & the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, 511436, PR China.
| | - Wenhui Hu
- Key Laboratory of Molecular Target & Clinical Pharmacology and the State & NMPA Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences & the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, 511436, PR China.
| | - Ping Sun
- Key Laboratory of Molecular Target & Clinical Pharmacology and the State & NMPA Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences & the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, 511436, PR China.
| |
Collapse
|
17
|
Liu F, Zhou Y, Liu L, Pan H, Liu H. Effect of 2-ethylbutyric acid on thermodynamics stability of various nonionic surfactants tanshione-loaded micelles. J Mol Liq 2022. [DOI: 10.1016/j.molliq.2022.119775] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
18
|
Niu X, Song H, Xiao X, Yu J, Yu J, Yang Y, Huang Q, Zang L, Han T, Zhang D, Li W. Tectoridin alleviates lipopolysaccharide -induced inflammaion via inhibiting TLR4-NF-κB/NLRP3 signaling in vivo and in vitro. Immunopharmacol Immunotoxicol 2022; 44:641-655. [PMID: 35506641 DOI: 10.1080/08923973.2022.2073890] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
BACKGROUD Tectoridin, widely extracted and separated from the rhizome of Iris tectorum Maxium, is extensively reported to have affluent bioactivity, but rarely reported to have anti-inflammatory effects. In this study, we aim to investigate the anti-inflammatory effects and the underlying mechanisms of tectoridin. METHODS Here, RAW264.7 macrophages were stimulated with Lipopolysaccharide (LPS) for the inflammation model in vitro. Experimental animals received tectoridin and Dexamethasone (DEX) before LPS injection for endotoxic shock mouse model in vivo. The pro-inflammatory mediators and cytokines in the cell supernatant and serum were detected by ELISA kits. The tissue damages were assessed by biochemical indexes and H&E staining. Immunohistochemistry and Western blot were performed for the detection of proteins. RESULTS Our data showed that tectoridin attenuated the LPS-up-regulated nitric oxide (NO), interleukin-6, (IL-6) and interleukin-18, (IL-18) from macrophages and tumor necrosis factor-α, (TNF-α); (IL-6) and (IL-1β) in the serum levels. Besides, our histopathological study showed that the damages caused by LPS in the lung, liver and kidney tissues were decreased. Furthermore, our results demonstrated that tectoridin inhibited the activation of TLR4-NF-κB/NLRP3 signaling proved by immunohistochemistry assay and Western blot. CONCLUSION Taken all together, tectoridin might have the potential ability of anti-inflammatory effects and the possible mechanism may be relevant to its inhibition of TLR4-NF-κB/NLRP3 signaling.
Collapse
Affiliation(s)
- Xiaofeng Niu
- School of Pharmacy, Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Huixin Song
- School of Pharmacy, Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Xin Xiao
- School of Pharmacy, Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Jinjin Yu
- School of Pharmacy, Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Jiabao Yu
- School of Pharmacy, Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Yajie Yang
- School of Pharmacy, Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Qiuxia Huang
- School of Pharmacy, Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Lulu Zang
- School of Pharmacy, Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Tengfei Han
- Shaanxi Panlong Pharmaceutical Group Limited by Share LTD, Xi'an, Shaanxi, China
| | - Dezhu Zhang
- Shaanxi Panlong Pharmaceutical Group Limited by Share LTD, Xi'an, Shaanxi, China
| | - Weifeng Li
- School of Pharmacy, Xi'an Jiaotong University, Xi'an, Shaanxi, China
| |
Collapse
|
19
|
Molecular Mechanism of Salvia miltiorrhiza Bunge in Treating Cerebral Infarction. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2022; 2022:5992394. [PMID: 35392650 PMCID: PMC8983215 DOI: 10.1155/2022/5992394] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/10/2021] [Accepted: 12/22/2021] [Indexed: 11/18/2022]
Abstract
Background Cerebral infarction (CI) is a common brain disease in clinical practice, which is mainly due to the pathological environment of ischemia and hypoxia caused by difficult cerebral circulation perfusion function, resulting in ischemic necrosis of local brain tissue and neurological impairment. In traditional Chinese medicine (TCM) theory, CI is mainly due to blood stasis in the brain. Therefore, blood-activating and stasis-dissipating drugs are often used to treat CI in clinical practice. Salvia miltiorrhiza Bunge (SMB) is a kind of traditional Chinese medicine with good efficacy in promoting blood circulation and removing blood stasis, and treatment of CI with it is a feasible strategy. Based on the above analysis, we chose network pharmacology to investigate the feasibility of SMB in the treatment of CI and to study the possible molecular mechanisms by providing some reference for the treatment of CI with TCM. Methods The active ingredients and related targets of SMB were obtained through the Traditional Chinese Medicine Systems Pharmacology (TCMSP) database, and CI-related targets were obtained from the GeneCards and DisGeNET databases. The target of SMB for the treatment of CI was obtained using Cytoscape software and visualized. GO and KEGG enrichment analysis was performed based on “clusterProfiler” within R, and the prediction results were validated by molecular docking technique. Results By constructing a compound-target (C-T) network, it was found that the active components in SMB mainly treated CI by regulating key proteins such as AKT1, IL-6, and EGFR. These key proteins mainly involve in pathways such as immune regulation, cancer and lipid metabolism, such as lipid and atherosclerosis, chemical carcinogenesis-receptor activation pathways, and IL-17 signaling pathway. In the GO term, it mainly regulates the response to steroid hormones, membrane rafts, and G protein-amine receptor coupled activity. Eventually, we verified that the luteolin and tanshinone IIA components in SMB have a good possibility of action with AKT1 and IL-6 by in silico techniques, indicating that SMB has some scientificity in the treatment of CI. Conclusion SMB mainly treats CI by regulating 94 proteins involved in lipid and atherosclerosis, chemical carcinogenesis-receptor activation, and IL-17 signaling pathway. Our research strategy provided a template for the drug development of TCM for the treatment of CI.
Collapse
|
20
|
Zhao J, Wei K, Jiang P, Chang C, Xu L, Xu L, Shi Y, Guo S, Xue Y, He D. Inflammatory Response to Regulated Cell Death in Gout and Its Functional Implications. Front Immunol 2022; 13:888306. [PMID: 35464445 PMCID: PMC9020265 DOI: 10.3389/fimmu.2022.888306] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Accepted: 03/17/2022] [Indexed: 02/03/2023] Open
Abstract
Gout, a chronic inflammatory arthritis disease, is characterized by hyperuricemia and caused by interactions between genetic, epigenetic, and metabolic factors. Acute gout symptoms are triggered by the inflammatory response to monosodium urate crystals, which is mediated by the innate immune system and immune cells (e.g., macrophages and neutrophils), the NACHT, LRR, and PYD domains-containing protein 3 (NLRP3) inflammasome activation, and pro-inflammatory cytokine (e.g., IL-1β) release. Recent studies have indicated that the multiple programmed cell death pathways involved in the inflammatory response include pyroptosis, NETosis, necroptosis, and apoptosis, which initiate inflammatory reactions. In this review, we explore the correlation and interactions among these factors and their roles in the pathogenesis of gout to provide future research directions and possibilities for identifying potential novel therapeutic targets and enhancing our understanding of gout pathogenesis.
Collapse
Affiliation(s)
- Jianan Zhao
- Guanghua Clinical Medical College, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Department of Rheumatology, Shanghai Guanghua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Institute of Arthritis Research in Integrative Medicine, Shanghai Academy of Traditional Chinese Medicine, Shanghai, China
| | - Kai Wei
- Guanghua Clinical Medical College, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Department of Rheumatology, Shanghai Guanghua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Institute of Arthritis Research in Integrative Medicine, Shanghai Academy of Traditional Chinese Medicine, Shanghai, China
| | - Ping Jiang
- Guanghua Clinical Medical College, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Department of Rheumatology, Shanghai Guanghua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Institute of Arthritis Research in Integrative Medicine, Shanghai Academy of Traditional Chinese Medicine, Shanghai, China
| | - Cen Chang
- Guanghua Clinical Medical College, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Department of Rheumatology, Shanghai Guanghua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Institute of Arthritis Research in Integrative Medicine, Shanghai Academy of Traditional Chinese Medicine, Shanghai, China
| | - Lingxia Xu
- Guanghua Clinical Medical College, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Department of Rheumatology, Shanghai Guanghua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Institute of Arthritis Research in Integrative Medicine, Shanghai Academy of Traditional Chinese Medicine, Shanghai, China
| | - Linshuai Xu
- Guanghua Clinical Medical College, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Department of Rheumatology, Shanghai Guanghua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Institute of Arthritis Research in Integrative Medicine, Shanghai Academy of Traditional Chinese Medicine, Shanghai, China
| | - Yiming Shi
- Guanghua Clinical Medical College, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Department of Rheumatology, Shanghai Guanghua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Institute of Arthritis Research in Integrative Medicine, Shanghai Academy of Traditional Chinese Medicine, Shanghai, China
| | - Shicheng Guo
- Computation and Informatics in Biology and Medicine, University of Wisconsin-Madison, Madison, WI, United States
- Department of Medical Genetics, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, United States
| | - Yu Xue
- Department of Rheumatology, Huashan Hospital, Institute of Rheumatology, Immunology and Allergy, Fudan University, Shanghai, China
| | - Dongyi He
- Guanghua Clinical Medical College, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Department of Rheumatology, Shanghai Guanghua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Institute of Arthritis Research in Integrative Medicine, Shanghai Academy of Traditional Chinese Medicine, Shanghai, China
- Arthritis Institute of Integrated Traditional and Western Medicine, Shanghai Chinese Medicine Research Institute, Shanghai, China
| |
Collapse
|
21
|
Wei Z, Zhan X, Ding K, Xu G, Shi W, Ren L, Fang Z, Liu T, Hou X, Zhao J, Li H, Li J, Li Z, Li Q, Lin L, Yang Y, Xiao X, Bai Z, Cao J. Dihydrotanshinone I Specifically Inhibits NLRP3 Inflammasome Activation and Protects Against Septic Shock In Vivo. Front Pharmacol 2021; 12:750815. [PMID: 34721038 PMCID: PMC8552015 DOI: 10.3389/fphar.2021.750815] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2021] [Accepted: 09/10/2021] [Indexed: 12/17/2022] Open
Abstract
The abnormal activation of the NLRP3 inflammasome is closely related to the occurrence and development of many inflammatory diseases. Targeting the NLRP3 inflammasome has been considered an efficient therapy to treat infections. We found that dihydrotanshinone I (DHT) specifically blocked the canonical and non-canonical activation of the NLRP3 inflammasome. Nevertheless, DHT had no relation with the activation of AIM2 or the NLRC4 inflammasome. Further study demonstrated that DHT had no influences on potassium efflux, calcium flux, or the production of mitochondrial ROS. We also discovered that DHT suppressed ASC oligomerization induced by NLRP3 agonists, suggesting that DHT inhibited the assembly of the NLRP3 inflammasome. Importantly, DHT possessed a significant therapeutic effect on NLRP3 inflammasome–mediated sepsis in mice. Therefore, our results aimed to clarify DHT as a specific small-molecule inhibitor for the NLRP3 inflammasome and suggested that DHT can be used as a potential drug against NLRP3-mediated diseases.
Collapse
Affiliation(s)
- Ziying Wei
- School of Chinese Meteria Medica, Beijing University of Chinese Medicine, Beijing, China.,Senior Department of Hepatology, the Fifth Medical Center of PLA General Hospital, Bejjing, China
| | - Xiaoyan Zhan
- Senior Department of Hepatology, the Fifth Medical Center of PLA General Hospital, Bejjing, China.,China Military Institute of Chinese Materia, Fifth Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Kaixin Ding
- Senior Department of Hepatology, the Fifth Medical Center of PLA General Hospital, Bejjing, China.,China Military Institute of Chinese Materia, Fifth Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Guang Xu
- Senior Department of Hepatology, the Fifth Medical Center of PLA General Hospital, Bejjing, China
| | - Wei Shi
- Senior Department of Hepatology, the Fifth Medical Center of PLA General Hospital, Bejjing, China
| | - Lutong Ren
- School of Chinese Meteria Medica, Beijing University of Chinese Medicine, Beijing, China.,Senior Department of Hepatology, the Fifth Medical Center of PLA General Hospital, Bejjing, China
| | - Zhie Fang
- Senior Department of Hepatology, the Fifth Medical Center of PLA General Hospital, Bejjing, China
| | - Tingting Liu
- Senior Department of Hepatology, the Fifth Medical Center of PLA General Hospital, Bejjing, China
| | - Xiaorong Hou
- Senior Department of Hepatology, the Fifth Medical Center of PLA General Hospital, Bejjing, China
| | - Jia Zhao
- Senior Department of Hepatology, the Fifth Medical Center of PLA General Hospital, Bejjing, China
| | - Hui Li
- School of Chinese Meteria Medica, Beijing University of Chinese Medicine, Beijing, China.,Senior Department of Hepatology, the Fifth Medical Center of PLA General Hospital, Bejjing, China
| | - Jiayi Li
- School of Chinese Meteria Medica, Beijing University of Chinese Medicine, Beijing, China.,Senior Department of Hepatology, the Fifth Medical Center of PLA General Hospital, Bejjing, China
| | - Zhiyong Li
- Senior Department of Hepatology, the Fifth Medical Center of PLA General Hospital, Bejjing, China
| | - Qiang Li
- Senior Department of Hepatology, the Fifth Medical Center of PLA General Hospital, Bejjing, China
| | - Li Lin
- Senior Department of Hepatology, the Fifth Medical Center of PLA General Hospital, Bejjing, China
| | - Yan Yang
- Senior Department of Hepatology, the Fifth Medical Center of PLA General Hospital, Bejjing, China
| | - Xiaohe Xiao
- Senior Department of Hepatology, the Fifth Medical Center of PLA General Hospital, Bejjing, China.,China Military Institute of Chinese Materia, Fifth Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Zhaofang Bai
- Senior Department of Hepatology, the Fifth Medical Center of PLA General Hospital, Bejjing, China.,China Military Institute of Chinese Materia, Fifth Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Junling Cao
- School of Chinese Meteria Medica, Beijing University of Chinese Medicine, Beijing, China.,Department of Pharmacy, Dongfang Hospital, Beijing University of Chinese Medicine, Beijing, China
| |
Collapse
|
22
|
Ren Y, Feng Y, Xu K, Yue S, Yang T, Nie K, Xu M, Xu H, Xiong X, Körte F, Barbeck M, Zhang P, Liu L. Enhanced Bioavailability of Dihydrotanshinone I-Bovine Serum Albumin Nanoparticles for Stroke Therapy. Front Pharmacol 2021; 12:721988. [PMID: 34531747 PMCID: PMC8438562 DOI: 10.3389/fphar.2021.721988] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Accepted: 07/28/2021] [Indexed: 02/03/2023] Open
Abstract
Dihydrotanshinone I (DHT) is a natural component in Salvia miltiorrhiza and has been widely researched for its multiple bioactivities. However, poor solubility and biocompatibility of DHT limit its desirable application for clinical purposes. Herein, DHT was encapsulated with bovine serum albumin (BSA) to enhance bioavailability. Compared to free DHT, DHT-BSA NPs (nanoparticles) showed an improved solubility in normal saline and increased protection against hydrogen peroxide-induced oxidative damage in PC12 cells. In addition, DHT-BSA NPs administered by intravenous injection displayed a significant efficacy in the middle cerebral artery occlusion/reperfusion models, without any impact on the cerebral blood flow. In summary, DHT-BSA NPs show an enhanced bioavailability compared with free DHT and a successful penetration into the central nervous system for stroke therapy, demonstrating their application potential in cardio-cerebrovascular diseases.
Collapse
Affiliation(s)
- Yanru Ren
- Beijing Advanced Innovation Center for Soft Matter Science and Engineering, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, China
| | - Yicheng Feng
- Beijing Advanced Innovation Center for Soft Matter Science and Engineering, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, China
| | - Kunyao Xu
- Beijing Advanced Innovation Center for Soft Matter Science and Engineering, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, China
| | - Saisai Yue
- Beijing Advanced Innovation Center for Soft Matter Science and Engineering, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, China
| | - Tiantian Yang
- Beijing Advanced Innovation Center for Soft Matter Science and Engineering, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, China
| | - Kaili Nie
- Beijing Advanced Innovation Center for Soft Matter Science and Engineering, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, China
| | - Man Xu
- Beijing Advanced Innovation Center for Soft Matter Science and Engineering, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, China
| | - Haijun Xu
- Beijing Advanced Innovation Center for Soft Matter Science and Engineering, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, China
| | - Xin Xiong
- NMI Natural and Medical Sciences Institute at the University of Tübingen, Reutlingen, Germany
| | - Fabian Körte
- NMI Natural and Medical Sciences Institute at the University of Tübingen, Reutlingen, Germany
| | - Mike Barbeck
- Institute of Material Science and Technology, Technical University of Berlin, Berlin, Germany
| | - Peisen Zhang
- Beijing Advanced Innovation Center for Soft Matter Science and Engineering, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, China
| | - Luo Liu
- Beijing Advanced Innovation Center for Soft Matter Science and Engineering, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, China
| |
Collapse
|