1
|
Liu Y, Wang Y, Cheng S, Mu J, Yin G, Gao H. Pantothenic acid alleviates osteoarthritis progression by inhibiting inflammatory response and ferroptosis through the SIRT1/Nrf2 signaling pathway. Chem Biol Interact 2025; 413:111494. [PMID: 40157627 DOI: 10.1016/j.cbi.2025.111494] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2025] [Revised: 03/03/2025] [Accepted: 03/26/2025] [Indexed: 04/01/2025]
Abstract
Osteoarthritis (OA) is a degenerative joint disease that is a major cause of deformity, swelling, pain and even loss of function in the knee joints of the elderly. Pantothenic acid (PA) plays a protective role in many organs due to its antioxidant and anti-inflammatory properties. Herein, we aimed to assess the protective role of PA on osteoarthritis and investigate the underlying molecular mechanism. The levels of inflammatory factors (IL-1β and TNF-α) in knee tissues were measured by ELISA. The Safranin O-Fast Green staining was used to assess the severity of OA and the H&E staining was used to assess the degree of synovitis. In vitro, the levels of iron, MDA, GSH were measured by the detection kits. Western blotting was used to assess the levels of signaling-related proteins. Our results showed that PA significantly attenuated the degree of cartilage degeneration in the MIA-induced osteoarthritis model. PA also reduced the expression of IL-1β, TNF-α, MMP1 and MMP3. In vitro, PA effectively reduced the concentrations of MMP1 and MMP3 in IL-1β-stimulated chondrocytes. PA decreased the levels of Fe2+ and MDA, while increasing GSH production and GPX4 and SLC7A11 expression in IL-1β-induced chondrocytes. Meanwhile, we found that PA was able to inhibit the phosphorylation level of p65, IκB protein in chondrocytes, which effectively blocked the NF-κB signaling pathway. Furthermore, PA also increased the level of SIRT1, Nrf2, and HO-1 protein expression. In addition, the inhibition of PA on IL-1β-induced MMPs production and ferroptosis were inhibited by the SIRT1 inhibitor EX-527. In conclusion, PA inhibited chondrocyte ferroptosis and cartilage destruction in osteoarthritis. The mechanism was through activating SIRT1/Nrf2 signaling pathway.
Collapse
Affiliation(s)
- Yi Liu
- Department of Bone and Joint Surgery, Orthopaedic Surgery Center, The First Hospital of Jilin University, Changchun, 130021, China
| | - Yang Wang
- The First Operation Room, The First Hospital of Jilin University, Changchun, 130021, China
| | - Shengqi Cheng
- Department of Bone and Joint Surgery, Orthopaedic Surgery Center, The First Hospital of Jilin University, Changchun, 130021, China
| | - Jie Mu
- Department of Bone and Joint Surgery, Orthopaedic Surgery Center, The First Hospital of Jilin University, Changchun, 130021, China
| | - Guanchen Yin
- Department of Bone and Joint Surgery, Orthopaedic Surgery Center, The First Hospital of Jilin University, Changchun, 130021, China
| | - Hang Gao
- Department of Bone and Joint Surgery, Orthopaedic Surgery Center, The First Hospital of Jilin University, Changchun, 130021, China.
| |
Collapse
|
2
|
Chen H, Zhao D, Liu S, Zhong Y, Wen Y, Chen L. Ginsenoside Rh1 attenuates chondrocyte senescence and osteoarthritis via AMPK/PINK1/Parkin-mediated mitophagy. Int Immunopharmacol 2025; 159:114911. [PMID: 40409109 DOI: 10.1016/j.intimp.2025.114911] [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: 03/13/2025] [Revised: 05/16/2025] [Accepted: 05/17/2025] [Indexed: 05/25/2025]
Abstract
Osteoarthritis (OA) is the most common joint disease characterized by disruption of extracellular matrix (ECM) homeostasis, chronic inflammation, and upregulation of senescent phenotypes. Ginsenoside Rh1 (Rh1) exerted various pharmacological activities, including anti-inflammatory, anti-cancer, and neuroprotective effects. Herein, we aimed to explore the role and mechanism of Rh1 in OA. In IL-1β-induced OA chondrocytes, Rh1 alleviated the imbalance of ECM and senescence phenotypes. Furthermore, we found that Rh1 mitigated mitochondrial damage and the impaired mitophagy of chondrocytes induced by IL-1β, and these effects could be prevented by Mdivi-1 (a mitophagy inhibitor). Knockdown of PINK1 or Parkin partially abolished Rh1-mediated chondroprotection, indicating that Rh1 exerts its therapeutic effects via PINK1/Parkin-dependent mitophagy. Based on molecular docking, Compound C (an AMPK inhibitor), and AMPK siRNA, we found that Rh1 regulated PINK1/Parkin-mediated mitophagy through AMPK. Besides, Rh1 alleviated OA by promoting AMPK-mediated mitophagy in the anterior cruciate ligament transection (ACLT) rats. In conclusion, Rh1 alleviated OA progress by regulating AMPK/PINK1/Parkin-mediated mitophagy and is a potentially effective therapeutic target for age-related OA.
Collapse
Affiliation(s)
- Haitao Chen
- Division of Joint Surgery and Sports Medicine, Department of Orthopedic Surgery, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China.
| | - Danyang Zhao
- Division of Joint Surgery and Sports Medicine, Department of Orthopedic Surgery, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China.
| | - Siyi Liu
- Division of Joint Surgery and Sports Medicine, Department of Orthopedic Surgery, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China.
| | - Yongkang Zhong
- Division of Joint Surgery and Sports Medicine, Department of Orthopedic Surgery, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China.
| | - Yinxian Wen
- Division of Joint Surgery and Sports Medicine, Department of Orthopedic Surgery, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China.
| | - Liaobin Chen
- Division of Joint Surgery and Sports Medicine, Department of Orthopedic Surgery, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China.
| |
Collapse
|
3
|
Liao Z, Liu X, Li L, Li S, Xing X, Zheng X, Song W, Gui P, Liu Q, Rong G, Shao Y, Zou M, Liao H, Wu X. Mechanism of the Proprietary Chinese Medicine "JiuLiWan" to Treat Ulcerative Colitis Revealed by Network Pharmacology, Molecular Docking, and Experimental Verification In Vitro. ACS OMEGA 2025; 10:19598-19613. [PMID: 40415848 PMCID: PMC12096223 DOI: 10.1021/acsomega.5c00261] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/09/2025] [Revised: 03/14/2025] [Accepted: 03/25/2025] [Indexed: 05/27/2025]
Abstract
JiuLiWan (JLW), as a classic traditional Chinese medicine formula, has been clinically used against ulcerative colitis (UC). However, the exact mechanism of its therapeutic effect remains unclear. This study aims to explore and validate the main components and pharmacological mechanism of JLW in the treatment of UC through network pharmacology, molecular docking, and cell experiments. Network pharmacology analyses indicated a total of 107 main components and 286 core targets of JLW against UC. Pathway enrichment analysis demonstrated the involvement of PI3K-AKT, MAPK, Ras, Rap1, TNF, T cell receptor, HIF-1, C-type lectin receptor, VEGF, and Th17 cell differentiation signal pathways in the efficacy of the formula. The molecular docking results indicated that the prominent components (ailanthone (AIL), butylidenephthalide, honokiol, dehydrocostuslactone, ganoderic acid A, atractylenolide I, neokurarinol, glycyrrhetinic acid, palmatine, tangeretin, and bruceine A) could bind to core targets AKT1, P53, STAT3, c-JUN, and ERK1. Subsequently, AIL was used as a representative compound to conduct cell experiments to verify its role and mechanism in anti-inflammation and immunomodulation. Interestingly, AIL could switch Jurkat T cells into a quiescence state without activating the inflammatory and immune status. However, AIL could significantly decrease the levels of interleukin-2 (IL-2) and interferon-gamma (IFN-γ), as well as the expression of surface activation markers CD69 and CD25, in PMA/ionomycin-activated Jurkat T cells by suppressing the RAF/ERK/STAT3 signaling pathway and increasing the phosphorylation of p53. This study combines network pharmacology prediction with experimental verification in vitro to demonstrate the mechanism of JLW in treating UC and provides an effective, safe, and inexpensive strategy for UC treatment.
Collapse
Affiliation(s)
- Zhifang Liao
- Dongguan
Key Laboratory of Characteristic Research and Achievement Transformation
of Integrated Chinese and Western Medicine for Prevention and Treatment
to Common Diseases, The First Dongguan Affiliated
Hospital of Guangdong Medical University, Guangdong Medical University, Dongguan, Guangdong Province523000, P. R. of
China
| | - Xiao Liu
- Dongguan
Key Laboratory of Characteristic Research and Achievement Transformation
of Integrated Chinese and Western Medicine for Prevention and Treatment
to Common Diseases, The First Dongguan Affiliated
Hospital of Guangdong Medical University, Guangdong Medical University, Dongguan, Guangdong Province523000, P. R. of
China
| | - Linxuan Li
- Dongguan
Key Laboratory of Characteristic Research and Achievement Transformation
of Integrated Chinese and Western Medicine for Prevention and Treatment
to Common Diseases, The First Dongguan Affiliated
Hospital of Guangdong Medical University, Guangdong Medical University, Dongguan, Guangdong Province523000, P. R. of
China
- The Key Laboratory
of Sepsis Translational Medicine, The Second
Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong Province524003, P.R. of
China
- Interdisciplinary
Science Research Center of Western Guangdong, The Second Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong Province524003, P.R. of
China
| | - Sikai Li
- Dongguan
Key Laboratory of Characteristic Research and Achievement Transformation
of Integrated Chinese and Western Medicine for Prevention and Treatment
to Common Diseases, The First Dongguan Affiliated
Hospital of Guangdong Medical University, Guangdong Medical University, Dongguan, Guangdong Province523000, P. R. of
China
| | - Xingxing Xing
- Dongguan
Key Laboratory of Characteristic Research and Achievement Transformation
of Integrated Chinese and Western Medicine for Prevention and Treatment
to Common Diseases, The First Dongguan Affiliated
Hospital of Guangdong Medical University, Guangdong Medical University, Dongguan, Guangdong Province523000, P. R. of
China
| | - Xiwen Zheng
- Dongguan
Key Laboratory of Characteristic Research and Achievement Transformation
of Integrated Chinese and Western Medicine for Prevention and Treatment
to Common Diseases, The First Dongguan Affiliated
Hospital of Guangdong Medical University, Guangdong Medical University, Dongguan, Guangdong Province523000, P. R. of
China
| | - Wenyu Song
- Dongguan
Key Laboratory of Characteristic Research and Achievement Transformation
of Integrated Chinese and Western Medicine for Prevention and Treatment
to Common Diseases, The First Dongguan Affiliated
Hospital of Guangdong Medical University, Guangdong Medical University, Dongguan, Guangdong Province523000, P. R. of
China
| | - Pin Gui
- Dongguan
Key Laboratory of Characteristic Research and Achievement Transformation
of Integrated Chinese and Western Medicine for Prevention and Treatment
to Common Diseases, The First Dongguan Affiliated
Hospital of Guangdong Medical University, Guangdong Medical University, Dongguan, Guangdong Province523000, P. R. of
China
| | - Qi Liu
- Dongguan
Key Laboratory of Characteristic Research and Achievement Transformation
of Integrated Chinese and Western Medicine for Prevention and Treatment
to Common Diseases, The First Dongguan Affiliated
Hospital of Guangdong Medical University, Guangdong Medical University, Dongguan, Guangdong Province523000, P. R. of
China
| | - Guanghong Rong
- Dongguan
Key Laboratory of Characteristic Research and Achievement Transformation
of Integrated Chinese and Western Medicine for Prevention and Treatment
to Common Diseases, The First Dongguan Affiliated
Hospital of Guangdong Medical University, Guangdong Medical University, Dongguan, Guangdong Province523000, P. R. of
China
| | - Yiming Shao
- Dongguan
Key Laboratory of Characteristic Research and Achievement Transformation
of Integrated Chinese and Western Medicine for Prevention and Treatment
to Common Diseases, The First Dongguan Affiliated
Hospital of Guangdong Medical University, Guangdong Medical University, Dongguan, Guangdong Province523000, P. R. of
China
- The Key Laboratory
of Sepsis Translational Medicine, The Second
Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong Province524003, P.R. of
China
| | - Mingzhi Zou
- The Key Laboratory
of Sepsis Translational Medicine, The Second
Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong Province524003, P.R. of
China
- Interdisciplinary
Science Research Center of Western Guangdong, The Second Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong Province524003, P.R. of
China
| | - Hongbo Liao
- Guangdong
Provincial Key Laboratory of Research and Development of Natural Drugs,
School of Pharmacy, Guangdong Medical University, Zhanjiang, Guangdong Province524023, P.R. of
China
| | - Xin Wu
- Dongguan
Key Laboratory of Characteristic Research and Achievement Transformation
of Integrated Chinese and Western Medicine for Prevention and Treatment
to Common Diseases, The First Dongguan Affiliated
Hospital of Guangdong Medical University, Guangdong Medical University, Dongguan, Guangdong Province523000, P. R. of
China
- The Key Laboratory
of Sepsis Translational Medicine, The Second
Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong Province524003, P.R. of
China
| |
Collapse
|
4
|
He Y, He MH, Jin T, Siju-Li, Wang HQ, He F. Tangeretin protects against Aβ1-42-induced toxicity and exploring mitochondria-lysosome interactions in HT22 cells. Biochem Biophys Res Commun 2025; 762:151769. [PMID: 40220719 DOI: 10.1016/j.bbrc.2025.151769] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2025] [Revised: 03/31/2025] [Accepted: 04/05/2025] [Indexed: 04/14/2025]
Abstract
Tangeretin, a flavonoid from Citri Reticulatae Pericarpium, is known for its neuroprotective effects, but the mechanisms are not fully understood. Alzheimer's disease, a leading neurodegenerative disorder, characterized by amyloid-beta (Aβ) accumulation, represents a significant therapeutic challenge. This study investigates the protective effects of tangeretin against Aβ1-42-induced neurotoxicity using HT22 cells and zebrafish larvae as experimental models. Tangeretin mitigated Aβ1-42-induced cytotoxicity, as evidenced by enhanced cell viability and reduced apoptosis. Tangeretin treatment mitigated Aβ1-42-induced cytotoxicity in HT22 cells, as evidenced by enhanced cell viability and reduced apoptosis. Mechanistically, tangeretin ameliorated mitochondrial dysfunction by reducing mitochondrial fragmentation, decreasing donut-shaped mitochondria, restoring mitochondrial membrane potential, and attenuating reactive oxygen species (ROS) production. Moreover, tangeretin modulated mitochondria-lysosome interactions by promoting mitophagy and normalizing the prolonged mitochondria-lysosome contact induced by Aβ1-42. In zebrafish larvae, Aβ1-42 exposure resulted in developmental malformations, including pericardial and yolk sac edema, elevated ROS levels, increased apoptosis, and impaired neurodevelopment. Tangeretin effectively counteracted these deficits, as revealed by live imaging, supporting its neuroprotective role observed in cellular models. Collectively, our study suggests that tangeretin may serve as a promising protective agent against Aβ1-42-induced neurotoxicity.
Collapse
Affiliation(s)
- Ying He
- School of Pharmaceutical Science, Sun Yat-sen University, Guangzhou, 510006, China.
| | - Meng-Hui He
- School of Pharmaceutical Science, Sun Yat-sen University, Guangzhou, 510006, China.
| | - Tingting Jin
- School of Pharmaceutical Science, Sun Yat-sen University, Guangzhou, 510006, China.
| | - Siju-Li
- School of Pharmaceutical Science, Sun Yat-sen University, Guangzhou, 510006, China.
| | - Hua-Qiao Wang
- School of Pharmaceutical Science, Sun Yat-sen University, Guangzhou, 510006, China.
| | - Feng He
- School of Pharmaceutical Science, Sun Yat-sen University, Guangzhou, 510006, China.
| |
Collapse
|
5
|
Shen C, Li H, Xiao M, Jiang X, Jin J, Zhou J, Xiong B, Chen Y, Zhao M. Study on the mechanism of the Chinese herbal pair Banxia-Chenpi in ameliorating polycystic ovary syndrome based on the CYP17A1 gene. JOURNAL OF ETHNOPHARMACOLOGY 2025; 344:119503. [PMID: 39961422 DOI: 10.1016/j.jep.2025.119503] [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: 10/29/2024] [Revised: 01/24/2025] [Accepted: 02/13/2025] [Indexed: 03/03/2025]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE As a typical Traditional Chinese Medicine (TCM) couplet medicine, Arum Ternatum Thunb. (Pinellia ternata (Thunb.) Makino, known as Banxia in Chinese) and Citrus Reticulata (pericarps of Citrus reticulata Blanco, known as Chenpi in Chinese) has been widely used in clinical practice for their properties of drying dampness, resolving phlegm, relieving oppression and masses. According to the TCM theories, the imbalance in fluid metabolism could lead to the accumulation of the excess dampness and phlegm, resulting in the pathological phenotype as 'damp-phlegm syndrome'. It can further lead to polycystic ovary syndrome (PCOS) when this accumulation of the excess fluid presents in uterus, affecting women's fertility and endocrine function. Recent studies have indicated that Banxia-Chenpi herbal pair (BXCP) exhibits significant therapeutic effects on damp-phlegm syndrome, yet the precise mechanisms underlying its anti-PCOS actions remain to be fully elucidated. AIM OF THE STUDY The objective was to investigate the signaling pathway involved in steroid biosynthesis, particularly the cytochrome P450 family 17, subfamily A, member 1 (CYP17A1), and to evaluate the effects and mechanisms of BXCP in ameliorating PCOS through both in vivo and in vitro experiments. MATERIALS AND METHODS A systematic evaluation was conducted to assess BXCP's effects on serum biochemical indicators and ovarian tissue pathology in a PCOS rat model (induced by high-fat diet + letrozole) and a DHT-induced human granulosa cells (KGN) model. Core targets were screened using absorbed components analysis, bioinformatics, metabolomics, and network analysis. RT-qPCR and Western blot techniques were employed to confirm the expression of CYP17A1 and related signaling molecule expression during BXCP's amelioration of PCOS, both in vivo and in vitro. RESULTS BXCP significantly ameliorated PCOS in vivo by mitigating weight gain, regulating estrus cycles, and normalizing sex hormone levels in rats. It upregulated metabolites related to steroid biosynthesis, including cortolone and progesterone, with CYP19A1, AKR1C3, and HSD17B1 as key regulators of CYP17A1. The main BXCP components, Naringenin and Nobiletin, increased CYP17A1 and CYP19A1 protein expression while decreased AKR1C3 and HSD17B1. CONCLUSION In conclusion, BXCP ameliorates PCOS by activating the CYP17A1-centered steroid biosynthesis pathway. These findings provide new insights into BXCP's clinical potentials in the management of patients with PCOS, highlighting the importance of TCM in modern medicine.
Collapse
Affiliation(s)
- Can Shen
- School of Traditional Chinese Medicine, Hubei University of Chinese Medicine, Wuhan, 430065, Hubei Province, China
| | - Haotian Li
- School of Basic Medical Sciences, Hubei University of Chinese Medicine, Wuhan, 430065, Hubei Province, China
| | - Min Xiao
- Experimental Center of Traditional Chinese Medicine, Hubei University of Chinese Medicine, Wuhan, 430065, Hubei Province, China; Hubei Shizhen Laboratory, Wuhan, 430061, Hubei Province, China
| | - Xiaocui Jiang
- Experimental Center of Traditional Chinese Medicine, Hubei University of Chinese Medicine, Wuhan, 430065, Hubei Province, China; Hubei Shizhen Laboratory, Wuhan, 430061, Hubei Province, China
| | - Jing Jin
- Hubei Shizhen Laboratory, Wuhan, 430061, Hubei Province, China; The First Clinical Medical School, Hubei University of Chinese Medicine, Wuhan, 430065, Hubei Province, China
| | - Jiayan Zhou
- School of Medicine, Stanford University, Stanford, CA, 94305, USA
| | - Bin Xiong
- School of Traditional Chinese Medicine, Hubei University of Chinese Medicine, Wuhan, 430065, Hubei Province, China; Hubei Shizhen Laboratory, Wuhan, 430061, Hubei Province, China.
| | - Yu Chen
- School of Traditional Chinese Medicine, Hubei University of Chinese Medicine, Wuhan, 430065, Hubei Province, China; Hubei Shizhen Laboratory, Wuhan, 430061, Hubei Province, China.
| | - Min Zhao
- School of Basic Medical Sciences, Hubei University of Chinese Medicine, Wuhan, 430065, Hubei Province, China; Hubei Shizhen Laboratory, Wuhan, 430061, Hubei Province, China.
| |
Collapse
|
6
|
Liu K, Liu R, Zhang C, Huang D, Wei B, Song Y, Wang C, Zhang X, Zheng M, Yan G. Suzi Daotan Decoction alleviates asthmatic airway remodeling through the AMPK/SIRT1/PGC-1α signaling pathway and PI3K/AKT signaling pathway. Sci Rep 2025; 15:6690. [PMID: 39994309 PMCID: PMC11850920 DOI: 10.1038/s41598-025-90870-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2024] [Accepted: 02/17/2025] [Indexed: 02/26/2025] Open
Abstract
Suzi Daotan Decoction (SZDTD), recorded in the "New Edition of the Sasang of Eastern Medicine", serves as a prominent formula for managing asthma in Shao-Yin individuals in Korean traditional medicine. This prescription demonstrates clinical efficacy in asthma treatment and is associated with anti-inflammatory and antioxidant properties. Nonetheless, the precise underlying mechanism remains incompletely understood. This study aims to elucidate the impact of SZDTD in ameliorating asthmatic airway remodeling and investigate whether its mechanism is related to the AMPK/SIRT1/PGC-1α and PI3K/AKT signaling pathways. Through network pharmacology analysis, the components and putative targets of SZDTD were investigated, along with the target genes associated with allergic asthma. Enrichment analysis identified the AMPK/SIRT1/PGC-1α and PI3K/AKT signaling pathways as relevant pathways. Subsequently, in an allergic asthma mouse model sensitized and challenged with ovalbumin (OVA), mice were orally administered a low dose of SZDTD, a high dose of SZDTD, or dexamethasone before the challenge. The control group received 0.9% NaCl only. The number of inflammatory cells was assessed using Diff-Quik staining. The levels of interleukin-4(IL-4), IL-5, IL-13 in broncho-alveolar lavage fluid (BALF), total immunoglobulin E(IgE), and OVA-specific IgE in serum were detected by Enzyme-linked immunosorbent assay. IL-4 and interferon γ (IFN-γ) in spleen and lymph were detected by flow cytometry. Histological staining was employed to observe lung tissue pathology. Protein levels were evaluated using Immunohistochemistry(IHC), Western blotting (WB), and immunofluorescence (IF). Furthermore, BEAS-2B human bronchial epithelial cells stimulated with LPS were treated with varying concentrations of SZDTD, and WB analysis was conducted to determine associated protein levels. SZDTD demonstrated a significant reduction in inflammatory cell infiltration, as well as decreased levels of IL-4, IL-5, and IL-13 in BALF, and total IgE and ovalbumin-specific IgE levels in serum. Flow cytometry analysis revealed that SZDTD treatment led to decreased levels of IFN-γ and IL-4 in the lymph nodes and spleen, with a more pronounced effect observed on IL-4 level. Moreover, results from MASSON staining indicated that SZDTD treatment markedly reduced the expression of α-SMA (α-smooth muscle actin) and mitigated collagen deposition symptoms. Furthermore, SZDTD stimulated the phosphorylation of Adenosine 5'-monophosphate-activated protein kinase (AMPK) and enhanced the expression of Silent information regulator 1 (SIRT1) and Peroxisome proliferator-activated receptor gamma coactivator 1α (PGC-1α), while inhibiting the expression of P-PI3K, P-AKT. In vitro experiments showed that SZDTD promoted the phosphorylation of AMPK, increased the expression of SIRT1 and PGC-1α, and suppressed the expression of P-PI3K, P-AKT. SZDTD can alleviate airway remodeling in allergic asthma by a mechanism related to activation of AMPK/SIRT1/PGC-1α and inhibition of PI3K/AKT signaling pathways.
Collapse
Affiliation(s)
- Kaiyue Liu
- Jilin Key Laboratory for Immune and Targeting Research On Common Allergic Diseases, Yanbian University, Yanji, 133002, Jilin, People's Republic of China
- Department of Integrated Chinese and Western Medicine, Yanbian University Medical College, No. 977, Gongyuan Road, Yanji, 133002, Jilin Province, People's Republic of China
| | - Ruobai Liu
- Jilin Key Laboratory for Immune and Targeting Research On Common Allergic Diseases, Yanbian University, Yanji, 133002, Jilin, People's Republic of China
- Department of Anatomy, Histology and Embryology, Yanbian University Medical College, No. 977, Gongyuan Road, Yanji, 133002, Jilin Province, People's Republic of China
| | - Chenghao Zhang
- Jilin Key Laboratory for Immune and Targeting Research On Common Allergic Diseases, Yanbian University, Yanji, 133002, Jilin, People's Republic of China
- Department of Oral Teaching and Research, Yanbian University Medical College, Yanji, 133000, Jilin Province, China
| | - Dandan Huang
- Jilin Key Laboratory for Immune and Targeting Research On Common Allergic Diseases, Yanbian University, Yanji, 133002, Jilin, People's Republic of China
- Department of Integrated Chinese and Western Medicine, Yanbian University Medical College, No. 977, Gongyuan Road, Yanji, 133002, Jilin Province, People's Republic of China
| | - Bowen Wei
- Jilin Key Laboratory for Immune and Targeting Research On Common Allergic Diseases, Yanbian University, Yanji, 133002, Jilin, People's Republic of China
- Department of Integrated Chinese and Western Medicine, Yanbian University Medical College, No. 977, Gongyuan Road, Yanji, 133002, Jilin Province, People's Republic of China
| | - Yilan Song
- Jilin Key Laboratory for Immune and Targeting Research On Common Allergic Diseases, Yanbian University, Yanji, 133002, Jilin, People's Republic of China
- Department of Anatomy, Histology and Embryology, Yanbian University Medical College, No. 977, Gongyuan Road, Yanji, 133002, Jilin Province, People's Republic of China
| | - Chongyang Wang
- Jilin Key Laboratory for Immune and Targeting Research On Common Allergic Diseases, Yanbian University, Yanji, 133002, Jilin, People's Republic of China
- Department of Anatomy, Histology and Embryology, Yanbian University Medical College, No. 977, Gongyuan Road, Yanji, 133002, Jilin Province, People's Republic of China
| | - Xin Zhang
- Changbai Mountain Protection Development Zone Central Hospital, Antu, 133600, People's Republic of China
| | - Mingyu Zheng
- Jilin Key Laboratory for Immune and Targeting Research On Common Allergic Diseases, Yanbian University, Yanji, 133002, Jilin, People's Republic of China.
- Department of Integrated Chinese and Western Medicine, Yanbian University Medical College, No. 977, Gongyuan Road, Yanji, 133002, Jilin Province, People's Republic of China.
- Key Laboratory of Natural Medicines of the Changbai Mountain, Ministry of Education, Yanbian University, Yanji, 133002, People's Republic of China.
| | - Guanghai Yan
- Jilin Key Laboratory for Immune and Targeting Research On Common Allergic Diseases, Yanbian University, Yanji, 133002, Jilin, People's Republic of China.
- Department of Anatomy, Histology and Embryology, Yanbian University Medical College, No. 977, Gongyuan Road, Yanji, 133002, Jilin Province, People's Republic of China.
- Key Laboratory of Natural Medicines of the Changbai Mountain, Ministry of Education, Yanbian University, Yanji, 133002, People's Republic of China.
| |
Collapse
|
7
|
Huang JX, Xu SZ, Tian T, Wang J, Jiang LQ, He T, Meng SY, Ni J, Pan HF. Genetic Links Between Metabolic Syndrome and Osteoarthritis: Insights From Cross-Trait Analysis. J Clin Endocrinol Metab 2025; 110:e461-e469. [PMID: 38482593 DOI: 10.1210/clinem/dgae169] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/12/2023] [Indexed: 01/22/2025]
Abstract
CONTEXT Previous observational studies have indicated a bidirectional association between metabolic syndrome (MetS) and osteoarthritis (OA). However, it remains unclear whether these bidirectional associations reflect causal relationships or shared genetic factors, and the underlying biological mechanisms of this association are not fully understood. OBJECTIVE We aimed to explore the genetic connection between MetS and OA using genome-wide association study (GWAS) summary data. METHODS Leveraging summary statistics from GWAS conducted by the UK Biobank and the Glucose and Insulin-related Traits Consortium (MAGIC), we performed global genetic correlation analyses, genome-wide cross-trait meta-analyses, and a bidirectional two-sample Mendelian randomization analyses using summary statistics from GWAS to comprehensively assess the relationship of MetS and OA. RESULTS We first detected an extensive genetic correlation between MetS and OA (rg = 0.393, P = 1.52 × 10-18), which was consistent in 4 MetS components, including waist circumference, triglycerides, hypertension, and high-density lipoprotein cholesterol and OA with rg ranging from -0.229 to 0.490. We then discovered 32 variants jointly associated with MetS and OA through Multi-Trait Analysis of GWAS (MTAG). Co-localization analysis found 12 genes shared between MetS and OA, with functional implications in several biological pathways. Finally, Mendelian randomization analysis suggested genetic liability to MetS significantly increased the risk of OA, but no reverse causality was found. CONCLUSION Our results illustrate a common genetic architecture, pleiotropic loci, as well as causality between MetS and OA, potentially enhancing our knowledge of high comorbidity and genetic processes that overlap between the 2 disorders.
Collapse
Affiliation(s)
- Ji-Xiang Huang
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, Hefei, Anhui 230032, China
- Institute of Kidney Disease, Inflammation & Immunity Mediated Diseases, The Second Hospital of Anhui Medical University, Hefei, Anhui 230032, China
| | - Shu-Zhen Xu
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, Hefei, Anhui 230032, China
- Institute of Kidney Disease, Inflammation & Immunity Mediated Diseases, The Second Hospital of Anhui Medical University, Hefei, Anhui 230032, China
| | - Tian Tian
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, Hefei, Anhui 230032, China
- Institute of Kidney Disease, Inflammation & Immunity Mediated Diseases, The Second Hospital of Anhui Medical University, Hefei, Anhui 230032, China
| | - Jing Wang
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, Hefei, Anhui 230032, China
- Institute of Kidney Disease, Inflammation & Immunity Mediated Diseases, The Second Hospital of Anhui Medical University, Hefei, Anhui 230032, China
| | - Ling-Qiong Jiang
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, Hefei, Anhui 230032, China
- Institute of Kidney Disease, Inflammation & Immunity Mediated Diseases, The Second Hospital of Anhui Medical University, Hefei, Anhui 230032, China
| | - Tian He
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, Hefei, Anhui 230032, China
- Institute of Kidney Disease, Inflammation & Immunity Mediated Diseases, The Second Hospital of Anhui Medical University, Hefei, Anhui 230032, China
| | - Shi-Yin Meng
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, Hefei, Anhui 230032, China
- Institute of Kidney Disease, Inflammation & Immunity Mediated Diseases, The Second Hospital of Anhui Medical University, Hefei, Anhui 230032, China
| | - Jing Ni
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, Hefei, Anhui 230032, China
- Institute of Kidney Disease, Inflammation & Immunity Mediated Diseases, The Second Hospital of Anhui Medical University, Hefei, Anhui 230032, China
| | - Hai-Feng Pan
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, Hefei, Anhui 230032, China
- Institute of Kidney Disease, Inflammation & Immunity Mediated Diseases, The Second Hospital of Anhui Medical University, Hefei, Anhui 230032, China
| |
Collapse
|
8
|
Zhang H, Wang Y, Wang S, Xue X, Huang K, Xu D, Jiang L, Li S, Zhang Y. Tangeretin alleviates sepsis-induced acute lung injury by inhibiting ferroptosis of macrophage via Nrf2 signaling pathway. Chin Med 2025; 20:11. [PMID: 39815349 PMCID: PMC11734455 DOI: 10.1186/s13020-025-01063-8] [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: 08/13/2024] [Accepted: 01/07/2025] [Indexed: 01/18/2025] Open
Abstract
BACKGROUND Sepsis-induced acute lung injury (ALI) is a severe clinical condition accompanied with high mortality. Tangeretin, which is widely found in citrus fruits, has been reported to exert antioxidant and anti-inflammatory properties. However, whether tangeretin protects against sepsis-induced ALI and the potential mechanisms remain unclear. METHODS We established an ALI model via intraperitoneally injected with 5 mg/kg lipopolysaccharides (LPS) for 12 h. Tangeretin was applied intraperitoneally 30 min before LPS treatment. Dexamethasone (Dex) was used as a positive control. Hematoxylin and eosin (HE) staining and protein content in bronchoalveolar lavage fluid (BALF) were determined to detect the degree of lung injury. RNA-seq was also applied to explore the effect of tangeretin on ALI. In vitro, RAW264.7 were treated with Nrf2 siRNA, the expression of ferroptosis-associated biomarkers, including glutathione peroxidase 4 (GPX4) and prostaglandin-endoperoxide synthase 2 (PTGS2) were assessed. Glutathione (GSH), malondialdehyde (MDA) levels, reactive oxygen species (ROS) and inflammatory factors were also determined both in vivo and in vitro. Furthermore, mice were treated with an Nrf2 inhibitor (ML385) to verify the mechanism of tangeretin in inhibiting sepsis-induced lung injury and ferroptosis. Data were analyzed using one way analysis of variance or two-tailed unpaired t tests. RESULTS Our study demonstrated that tangeretin significantly alleviated lung injury, reversed the LPS-induced reduction in GPX4 and GSH, and mitigates the elevation of PTGS2 and MDA levels. Tangeretin also reduced 4-HNE and iron levels. Besides, the levels of LPS-stimulated inflammatory factors IL-6, IL-1β and TNF-α were also decreased by tangeretin. RNA-seq and bioinformatics analysis demonstrated that tangeretin inhibited inflammatory response. Mechanistically, we identified that tangeretin inhibited the GPX4-dependent lipid peroxidation through activation of Nrf2. The silence of Nrf2 abolished the inhibitory effect of tangeretin on oxidative stress, inflammatory response and ferroptosis in RAW264.7 cells. Additionally, all the protective effects of tangeretin on ALI were abolished in Nrf2 inhibitor-treated mice. CONCLUSION We identified that ferroptosis as a critical mechanism contributing to sepsis-induced ALI. Tangeretin, a promising therapeutic candidate, effectively mitigates ALI through inhibiting ferroptosis via upregulating Nrf2 signaling pathway.
Collapse
Affiliation(s)
- Hui Zhang
- Department of Anesthesiology and Surgical Intensive Care Unit, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, 1665 Kongjiang Road, Shanghai, 200092, China
| | - Yan Wang
- Department of Anesthesiology and Surgical Intensive Care Unit, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, 1665 Kongjiang Road, Shanghai, 200092, China
| | - Shenghua Wang
- Department of Anesthesiology and Surgical Intensive Care Unit, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, 1665 Kongjiang Road, Shanghai, 200092, China
| | - Xiaomei Xue
- Department of Anesthesiology and Surgical Intensive Care Unit, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, 1665 Kongjiang Road, Shanghai, 200092, China
| | - Kai Huang
- Department of Anesthesiology and Surgical Intensive Care Unit, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, 1665 Kongjiang Road, Shanghai, 200092, China
| | - Dunfeng Xu
- Department of Anesthesiology and Surgical Intensive Care Unit, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, 1665 Kongjiang Road, Shanghai, 200092, China
| | - Lai Jiang
- Department of Anesthesiology and Surgical Intensive Care Unit, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, 1665 Kongjiang Road, Shanghai, 200092, China
| | - Siyuan Li
- Department of Anesthesiology and Surgical Intensive Care Unit, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, 1665 Kongjiang Road, Shanghai, 200092, China.
| | - Yunqian Zhang
- Department of Anesthesiology and Surgical Intensive Care Unit, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, 1665 Kongjiang Road, Shanghai, 200092, China.
| |
Collapse
|
9
|
Li P, Tang W, Wen H, Zhou S, Cao H. Senkyunolide I prevent chondrocytes from oxidative stress through Nrf2/HO-1 signaling pathway. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025:10.1007/s00210-024-03776-3. [PMID: 39779606 DOI: 10.1007/s00210-024-03776-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Accepted: 12/28/2024] [Indexed: 01/11/2025]
Abstract
Osteoarthritis (OA) is a degenerative musculoskeletal disease, featured by the destruction of articular cartilage. Oxidative stress, one of the drivers of the extracellular matrix degradation in cartilage, plays a vital role in OA pathogenesis. Senkyunolide I (SEI) is a natural compound with a prominent anti-oxidative stress property against multiple diseases. However, the protective effect of SEI on OA has not been explored. Here, we aimed to elucidate the effect of SEI on OA in vitro. Our results showed that SEI suppressed the expression of senescence-related markers such as P16 and P21 in IL-1β-induced chondrocytes. Besides, SEI alleviated IL-1β-induced the degradation of extracellular matrix (ECM) by suppressing the matrix proteinase like MMP13 and ATAMDS5 while promoting matrix synthesis regulated biomarkers like COL2A1 and ACAN in chondrocytes. Mechanically, the mitochondrial dysfunction and overproduction of intracellular reactive oxygen species (ROS) in chondrocytes induced by IL-1β were reversed by SEI. Additionally, the ROS inhibitor N-acetylcysteine (NAC) synergistically enhanced the biological effect of SEI in IL-1β-induced chondrocytes. Moreover, it was also found that the expression of Nrf2 and HO-1 was increased by the treatment of SEI in IL-1β-stimulated chondrocytes, while the Nrf2 inhibitor ML385 reversed the protective effect of SEI on OA chondrocytes. In conclusion, SEI could inhibit senescence, the degradation of ECM, and the production of ROS through activating Nrf2/ HO-1 signaling pathway, which provide a novel candidate for OA treatment.
Collapse
Affiliation(s)
- Pengbin Li
- Department of Orthopedics, General Hospital of The Yangtze River Shipping, Wuhan Brain Hospital, Wuhan, China
| | - Wenjuan Tang
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, Hubei, China
| | - Haiyan Wen
- Department of Pharmacy, Renmin Hospital of Wuhan University, Wuhan, 430060, China.
| | - Siqi Zhou
- Department of Orthopedics, Renmin Hospital of Wuhan University, Wuhan, 430060, China.
| | - Hui Cao
- Department of Orthopedics, Renmin Hospital of Wuhan University, Wuhan, 430060, China.
| |
Collapse
|
10
|
Yue L, Li N, Ye X, Xiu Y, Wang B. Polymethoxylated flavones for modulating signaling pathways in inflammation. Int Immunopharmacol 2024; 143:113522. [PMID: 39515044 DOI: 10.1016/j.intimp.2024.113522] [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: 04/16/2024] [Revised: 09/18/2024] [Accepted: 10/25/2024] [Indexed: 11/16/2024]
Abstract
Aberrant signaling pathways play a crucial role in the pathogenesis of various diseases, including inflammatory disorders and autoimmune conditions. Polymethoxylated flavones (PMFs), a class of natural compounds found in citrus fruits, have obtained increasing attention for their potential therapeutic effects in modulating inflammatory responses. Although significant progress has been made in the pharmacological research of PMFs, the mechanisms by which they modulate signaling pathways to treat inflammation have not been systematically reviewed or analyzed. To address this gap in the literature, this review explores the mechanisms underlying the anti-inflammatory properties of PMFs and their prospects as drugs for treating inflammatory diseases. We discuss the molecular targets and signaling pathways through which PMFs exert their anti-inflammatory effects, including NF-κB pathway, PI3K/Akt pathway, MAPK pathway, Nrf2 pathway, and regulation of inflammatory cytokine production. Furthermore, we highlight preclinical studies evaluating the efficacy of PMFs in various inflammatory conditions, such as rheumatoid arthritis (RA), inflammatory bowel disease (IBD), and osteoarthritis (OA). Despite promising findings, challenges remain in optimizing the pharmacokinetic properties and therapeutic efficacy of PMFs for clinical use. Future research directions include elucidating the structure-activity relationships of PMFs, developing novel delivery strategies, and conducting large-scale clinical trials to validate their efficacy and safety profiles. Overall, PMFs represent a promising class of natural compounds with potential applications as anti-inflammatory drugs, offering novel therapeutic opportunities for managing inflammatory diseases.
Collapse
Affiliation(s)
- Lixia Yue
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; Center for Pharmaceutics Research, Shanghai Institute of Materia Medica Chinese Academy of Sciences, Shanghai 201203, China
| | - Ning Li
- Shenzhen Research Institute, the Hong Kong University of Science and Technology, Shenzhen 518054, China
| | - Xianglu Ye
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Yanfeng Xiu
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China.
| | - Bing Wang
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.
| |
Collapse
|
11
|
Liang L, He C, Han X, Liu J, Yang L, Chang F, Zhang Y, Lin J. Zuojin Pill Alleviates Precancerous Lesions of Gastric Cancer by Modulating the MEK/ERK/c-Myc Pathway: An Integrated Approach of Network Pharmacology, Molecular Dynamics Simulation, and Experimental Validation. Drug Des Devel Ther 2024; 18:5905-5929. [PMID: 39679136 PMCID: PMC11646374 DOI: 10.2147/dddt.s487371] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2024] [Accepted: 11/21/2024] [Indexed: 12/17/2024] Open
Abstract
Background Precancerous lesions of gastric cancer (PLGC) represent critical stages in gastric cancer progression, with a high risk of malignancy. Current treatments, such as Helicobacter pylori eradication, show limited efficacy in reversing precancerous molecular changes. Zuojin Pill (ZJP), a traditional Chinese medicine, has demonstrated potential for treating digestive disorders and may offer a promising approach for PLGC intervention. Objective This study aims to investigate the therapeutic effects and mechanisms of ZJP in treating PLGC, focusing on its active components, target pathways, and molecular interactions. By using advanced analytical techniques, we provide a scientific foundation for ZJP's potential application in early gastric cancer intervention. Methods Using ultra-high performance liquid chromatography-quadrupole orbitrap high-resolution mass spectrometry (UPLC-Q-Orbitrap HRMS), we identified active components in ZJP. A network pharmacology approach was then applied to construct a "ZJP-compound-target-disease" network. Molecular docking and molecular dynamics simulations were conducted to analyze the stability and interactions of the main active components of ZJP with core protein targets in PLGC. Animal experiments were used to validate significant targets and pathways in vivo. Results Tangeritin, Isorhamnetin, Caffeic Acid, Azelaic Acid, and Adenosine were identified as the main active components of ZJP in the treatment of PLGC, with key targets including PIK3R1, MAPK3, SRC, JAK2, STAT3, and PIK3CA. Molecular docking and molecular dynamics simulations further confirmed the relationship between compounds and target proteins. The potential molecular mechanism of ZJP predicted by network pharmacology analysis was confirmed in PLGC rats. ZJP downregulated IL-6, TNF-α, c-myc, p-MEK1 and p-ERK1/2, effectively reversing the progression of PLGC. Conclusion ZJP can reverse MNNG-induced PLGC, potentially through inhibition of the MEK/ERK/c-myc pathway and regulation of cellular proliferation and apoptosis.
Collapse
Affiliation(s)
- Lan Liang
- The First Clinical Medical School, Shaanxi University of Chinese Medicine, Xianyang, People’s Republic of China
- College of Nursing, Shaanxi Energy Institute, Xianyang, People’s Republic of China
| | - Chenming He
- LongHua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, People’s Republic of China
| | - Xue Han
- Xijing 986 Hospital Department, Air Force Medical University, Xian, People’s Republic of China
| | - Jia Liu
- School of Basic Medical Sciences, Shaanxi University of Chinese Medicine, Xianyang, People’s Republic of China
| | - Liuhong Yang
- School of Basic Medical Sciences, Shaanxi University of Chinese Medicine, Xianyang, People’s Republic of China
| | - Fengjiao Chang
- School of Nursing, Shaanxi University of Chinese Medicine, Xianyang, People’s Republic of China
| | - Yami Zhang
- The Fifth Oncology Department, Affiliated Hospital of Shaanxi University of Chinese Medicine, Xianyang, People’s Republic of China
| | - Jie Lin
- School of Basic Medical Sciences, Shaanxi University of Chinese Medicine, Xianyang, People’s Republic of China
- Shaanxi Provincial Key Laboratory of TCM Constitution and Disease Prevention, Xianyang, People’s Republic of China
| |
Collapse
|
12
|
Li M, Xiao J, Chen B, Pan Z, Wang F, Chen W, He Q, Li J, Li S, Wang T, Zhang G, Wang H, Chen J. Loganin inhibits the ROS-NLRP3-IL-1β axis by activating the NRF2/HO-1 pathway against osteoarthritis. Chin J Nat Med 2024; 22:977-990. [PMID: 39510640 DOI: 10.1016/s1875-5364(24)60555-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2024] [Indexed: 11/15/2024]
Abstract
Loganin (LOG), a bioactive compound derived from Cornus officinalis Siebold & Zucc, has been understudied in the context of osteoarthritis (OA) treatment. In this study, we induced an inflammatory response in chondrocytes using lipopolysaccharide (LPS) and subsequently treated these cells with LOG. We employed fluorescence analysis to quantify reactive oxygen species (ROS) levels and measured the expression of NLRP3 and nuclear factor erythropoietin-2-related factor 2 (NRF2) using real-time quantitative polymerase chain reaction (qRT-PCR), Western blotting, and immunofluorescence (IF) techniques. Additionally, we developed an OA mouse model by performing medial meniscus destabilization (DMM) surgery and monitored disease progression through micro-computed tomography (micro-CT), hematoxylin and eosin (H&E) staining, safranin O and fast green (S&F) staining, and immunohistochemical (IHC) analysis. Our results indicate that LOG significantly reduced LPS-induced ROS levels in chondrocytes, inhibited the activation of the NLRP3 inflammasome, and enhanced NRF2/heme oxygenase 1 (HO-1) signaling. In vivo, LOG treatment mitigated cartilage degradation and osteophyte formation triggered by DMM surgery, decreased NLRP3 expression, and increased NRF2 expression. These findings suggest that LOG has a protective effect against OA, potentially delaying disease progression by inhibiting the ROS-NLRP3-IL-1β axis and activating the NRF2/HO-1 pathway.
Collapse
Affiliation(s)
- Miao Li
- 1(st) School of Medicine, Guangzhou University of Chinese Medicine, Guangzhou 510405, China; The Laboratory of Orthopaedics and Traumatology of Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| | - Jiacong Xiao
- 1(st) School of Medicine, Guangzhou University of Chinese Medicine, Guangzhou 510405, China; The Laboratory of Orthopaedics and Traumatology of Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| | - Baihao Chen
- 1(st) School of Medicine, Guangzhou University of Chinese Medicine, Guangzhou 510405, China; The Laboratory of Orthopaedics and Traumatology of Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| | - Zhaofeng Pan
- 1(st) School of Medicine, Guangzhou University of Chinese Medicine, Guangzhou 510405, China; The Laboratory of Orthopaedics and Traumatology of Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| | - Fanchen Wang
- 1(st) School of Medicine, Guangzhou University of Chinese Medicine, Guangzhou 510405, China; The Laboratory of Orthopaedics and Traumatology of Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| | - Weijian Chen
- 1(st) School of Medicine, Guangzhou University of Chinese Medicine, Guangzhou 510405, China; The Laboratory of Orthopaedics and Traumatology of Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| | - Qi He
- 1(st) School of Medicine, Guangzhou University of Chinese Medicine, Guangzhou 510405, China; The Laboratory of Orthopaedics and Traumatology of Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| | - Jianliang Li
- 1(st) School of Medicine, Guangzhou University of Chinese Medicine, Guangzhou 510405, China; The Laboratory of Orthopaedics and Traumatology of Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| | - Shaocong Li
- 1(st) School of Medicine, Guangzhou University of Chinese Medicine, Guangzhou 510405, China; The Laboratory of Orthopaedics and Traumatology of Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| | - Ting Wang
- 1(st) School of Medicine, Guangzhou University of Chinese Medicine, Guangzhou 510405, China; The Laboratory of Orthopaedics and Traumatology of Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| | - Gangyu Zhang
- 1(st) School of Medicine, Guangzhou University of Chinese Medicine, Guangzhou 510405, China; Department of Biomedicine, University of Basel, Basel, Switzerland.
| | - Haibin Wang
- Department of Orthopaedics, First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou 510405, China.
| | - Jianfa Chen
- Department of Orthopaedics, First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou 510405, China.
| |
Collapse
|
13
|
Zhang X, Jiang Y, Zeng J, Li X, Xie H, Yang R, Qi H, Zeng N. Phytochemistry, pharmacological properties and pharmacokinetics of Citri Reticulatae Pericarpium: A systematic review. JOURNAL OF ETHNOPHARMACOLOGY 2024; 333:118503. [PMID: 38942157 DOI: 10.1016/j.jep.2024.118503] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/28/2024] [Revised: 06/11/2024] [Accepted: 06/25/2024] [Indexed: 06/30/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Citri Reticulatae Pericarpium (CRP), known as Chen Pi in China, is the most commonly used medicine for regulating qi. As a traditional medicine, CRP has been extensively used in the clinical treatment of nausea, vomiting, cough and phlegm for thousands of years. It is mainly distributed in Guangdong, Sichuan, Fujian and Zhejiang in China. Due to its high frequency of use, many scholars have conducted a lot of research on it and the related chemical constituents it contains. In this review, the research progress on phytochemistry, pharmacology, pharmacokinetics and toxicology of CRP are summarized. AIM OF THE REVIEW The review aims to sort out the methods of extraction and purification, pharmacological activities and mechanisms of action, pharmacokinetics and toxicology of the chemical constituents in CRP, in order to elaborate the future research directions and challenges for the study of CRP and related chemical constituents. MATERIALS AND METHODS Valid and comprehensive relevant information was collected from China National Knowledge Infrastructure, Web of Science, PubMed and so on. RESULTS CRP contains a variety of compounds, of which terpenes, flavonoids and alkaloids are the main components, and they are also the primary bioactive components that play a pharmacological role. Flavonoids and terpenes are extracted and purified by aqueous and alcoholic extraction methods, assisted by ultrasonic and microwave extraction, in order to achieve higher yields with less resources. Pharmacological studies have shown that CRP possesses a variety of highly active chemical components and a wide range of pharmacological activities, including anti-tumor, anti-inflammatory, immunomodulatory, hepatoprotective, therapeutic for cardiovascular-related disorders, antioxidant, antibacterial, and neuroprotective effects. CONCLUSIONS There is a diversity in the chemical compositions of CRP, which have multiple biological activities and promising applications. However, the pharmacological activities of CRP are mainly dependent on the action of its chemical components, but the relationship between the structure of chemical components and the biological effects has not been thoroughly investigated, and therefore, the structure-activity relationship is an issue that needs to be elucidated urgently. In addition, the pharmacokinetic studies of the relevant components can be further deepened and the correlation studies between pharmacological effects and syndromes of TCM can be expanded to ensure the effectiveness and rationality of CRP for human use.
Collapse
Affiliation(s)
- Xiongwei Zhang
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of TCM, Chengdu, 611137, China; School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, 611137, China
| | - Yanning Jiang
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of TCM, Chengdu, 611137, China; School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, 611137, China
| | - Jiuseng Zeng
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of TCM, Chengdu, 611137, China; School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, 611137, China
| | - Xiangyu Li
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of TCM, Chengdu, 611137, China; School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, 611137, China
| | - Hongxiao Xie
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of TCM, Chengdu, 611137, China; School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, 611137, China
| | - Ruocong Yang
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of TCM, Chengdu, 611137, China; School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, 611137, China.
| | - Hu Qi
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of TCM, Chengdu, 611137, China; School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, 611137, China.
| | - Nan Zeng
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of TCM, Chengdu, 611137, China; School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, 611137, China.
| |
Collapse
|
14
|
Sheng W, Yue Y, Qi T, Qin H, Liu P, Wang D, Zeng H, Yu F. The Multifaceted Protective Role of Nuclear Factor Erythroid 2-Related Factor 2 in Osteoarthritis: Regulation of Oxidative Stress and Inflammation. J Inflamm Res 2024; 17:6619-6633. [PMID: 39329083 PMCID: PMC11424688 DOI: 10.2147/jir.s479186] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Accepted: 09/05/2024] [Indexed: 09/28/2024] Open
Abstract
Osteoarthritis (OA) is a chronic degenerative joint disease characterized by the degradation of joint cartilage, subchondral bone sclerosis, synovitis, and structural changes in the joint. Recent research has highlighted the role of various genes in the pathogenesis and progression of OA, with nuclear factor erythroid 2-related factor 2 (NRF2) emerging as a critical player. NRF2, a vital transcription factor, plays a key role in regulating the OA microenvironment and slowing the disease's progression. It modulates the expression of several antioxidant enzymes, such as Heme oxygenase-1 (HO-1) and NAD(P)H oxidoreductase 1 (NQO1), among others, which help reduce oxidative stress. Furthermore, NRF2 inhibits the nuclear factor kappa-B (NF-κB) signaling pathway, thereby decreasing inflammation, joint pain, and the breakdown of cartilage extracellular matrix, while also mitigating cell aging and death. This review discusses NRF2's impact on oxidative stress, inflammation, cell aging, and various cell death modes (such as apoptosis, necroptosis, and ferroptosis) in OA-affected chondrocytes. The role of NRF2 in OA macrophages, and synovial fibroblasts was also discussed. It also covers NRF2's role in preserving the cartilage extracellular matrix and alleviating joint pain. The purpose of this review is to provide a comprehensive understanding of NRF2's protective mechanisms in OA, highlighting its potential as a therapeutic target and underscoring its significance in the development of novel treatment strategies for OA.
Collapse
Affiliation(s)
- Weibei Sheng
- Department of Bone & Joint Surgery, National & Local Joint Engineering Research Center of Orthopaedic Biomaterials, Shenzhen Key Laboratory of Orthopaedic Diseases and Biomaterials Research, Peking University Shenzhen Hospital, Shenzhen, 518036, People’s Republic of China
| | - Yaohang Yue
- Department of Bone & Joint Surgery, National & Local Joint Engineering Research Center of Orthopaedic Biomaterials, Shenzhen Key Laboratory of Orthopaedic Diseases and Biomaterials Research, Peking University Shenzhen Hospital, Shenzhen, 518036, People’s Republic of China
| | - Tiantian Qi
- Department of Bone & Joint Surgery, National & Local Joint Engineering Research Center of Orthopaedic Biomaterials, Shenzhen Key Laboratory of Orthopaedic Diseases and Biomaterials Research, Peking University Shenzhen Hospital, Shenzhen, 518036, People’s Republic of China
| | - Haotian Qin
- Department of Bone & Joint Surgery, National & Local Joint Engineering Research Center of Orthopaedic Biomaterials, Shenzhen Key Laboratory of Orthopaedic Diseases and Biomaterials Research, Peking University Shenzhen Hospital, Shenzhen, 518036, People’s Republic of China
| | - Peng Liu
- Department of Bone & Joint Surgery, National & Local Joint Engineering Research Center of Orthopaedic Biomaterials, Shenzhen Key Laboratory of Orthopaedic Diseases and Biomaterials Research, Peking University Shenzhen Hospital, Shenzhen, 518036, People’s Republic of China
| | - Deli Wang
- Department of Bone & Joint Surgery, National & Local Joint Engineering Research Center of Orthopaedic Biomaterials, Shenzhen Key Laboratory of Orthopaedic Diseases and Biomaterials Research, Peking University Shenzhen Hospital, Shenzhen, 518036, People’s Republic of China
| | - Hui Zeng
- Shenzhen Second People’s Hospital, The First Affiliated Hospital of Shenzhen University, Shenzhen, 518035, People’s Republic of China
| | - Fei Yu
- Department of Bone & Joint Surgery, National & Local Joint Engineering Research Center of Orthopaedic Biomaterials, Shenzhen Key Laboratory of Orthopaedic Diseases and Biomaterials Research, Peking University Shenzhen Hospital, Shenzhen, 518036, People’s Republic of China
| |
Collapse
|
15
|
Wang L, Shao T, Liu C, Han Z, Zhang S, Dong Y, Han T, Cheng B, Ren W. Liensinine inhibits IL-1β-stimulated inflammatory response in chondrocytes and attenuates papain-induced osteoarthritis in rats. Int Immunopharmacol 2024; 138:112601. [PMID: 38971106 DOI: 10.1016/j.intimp.2024.112601] [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: 12/12/2023] [Revised: 06/26/2024] [Accepted: 06/28/2024] [Indexed: 07/08/2024]
Abstract
Osteoarthritis (OA) is a joint disease caused by inflammation of cartilage and synovial tissue. Suppressing the process of inflammatory reaction and the generation of oxidative stress is an effective strategy to alleviate the progression of OA. Liensinine is one of the main components of lotus seeds, which has anti-hypertensive and anti-arrhythmia activities. In this study, we aimed to determine the anti-inflammatory effect of liensinine in an OA. Here, we found that liensinine significantly inhibited the inflammatory response of SW1353 cells and primary chondrocytes by inhibiting the release of inflammatory cytokines and oxidative stress. Moreover, we showed that liensinine was able to inhibit the activation of the NF-κB signaling pathway in IL-1β-induced SW1353 cells. Lastly, we found that liensinine significantly ameliorated cartilage damage and inflammatory response in papain-induced rats. Our study demonstrated a significant protective effect of liensinine against OA, which might be by inhibiting the activation of the NF-κB signaling pathway, and provide a new insight for the treatment of OA using liensinine.
Collapse
Affiliation(s)
- Lei Wang
- The Third Affiliated Hospital of Xinxiang Medical University, Xinxiang 453003, Henan, China; Clinical Medical Center of Tissue Engineering and Regeneration, Xinxiang Medical University, Xinxiang 453003, Henan, China; Xinxiang Key Laboratory of Cellular Stress Biology, School of Life Sciences and Technology, Xinxiang Medical University, Xinxiang 453003, Henan, China
| | - Tianci Shao
- Xinxiang Key Laboratory of Cellular Stress Biology, School of Life Sciences and Technology, Xinxiang Medical University, Xinxiang 453003, Henan, China
| | - Chen Liu
- Xinxiang Key Laboratory of Cellular Stress Biology, School of Life Sciences and Technology, Xinxiang Medical University, Xinxiang 453003, Henan, China
| | - Ziyu Han
- Institutes of Health Central Plains, Xinxiang Medical University, Xinxiang 453003, Henan, China
| | - Shenghui Zhang
- Xinxiang Key Laboratory of Cellular Stress Biology, School of Life Sciences and Technology, Xinxiang Medical University, Xinxiang 453003, Henan, China
| | - Yuqian Dong
- Xinxiang Key Laboratory of Cellular Stress Biology, School of Life Sciences and Technology, Xinxiang Medical University, Xinxiang 453003, Henan, China
| | - Tao Han
- Institutes of Health Central Plains, Xinxiang Medical University, Xinxiang 453003, Henan, China
| | - Binfeng Cheng
- The Third Affiliated Hospital of Xinxiang Medical University, Xinxiang 453003, Henan, China; Xinxiang Key Laboratory of Cellular Stress Biology, School of Life Sciences and Technology, Xinxiang Medical University, Xinxiang 453003, Henan, China.
| | - Wenjie Ren
- The Third Affiliated Hospital of Xinxiang Medical University, Xinxiang 453003, Henan, China; Clinical Medical Center of Tissue Engineering and Regeneration, Xinxiang Medical University, Xinxiang 453003, Henan, China.
| |
Collapse
|
16
|
Peng B, Hu J, Sun Y, Huang Y, Peng Q, Zhao W, Xu W, Zhu L. Tangeretin alleviates inflammation and oxidative response induced by spinal cord injury by activating the Sesn2/Keap1/Nrf2 pathway. Phytother Res 2024; 38:4555-4569. [PMID: 39054118 DOI: 10.1002/ptr.8294] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2023] [Revised: 04/29/2024] [Accepted: 07/07/2024] [Indexed: 07/27/2024]
Abstract
Spinal cord injury (SCI) is a severe disabling disease that is characterized by inflammation and oxidative reactions. Tangeretin has been shown to possess significant antioxidant and anti-inflammatory activities. The Keap1/Nrf2 pathway, downstream of the Sesn2 gene, is involved in regulating the inflammation and oxidative response. The main objective of this study was to investigate the effect of tangeretin on SCI and its possible mechanism through cell and animal models. A T9 clamp injury was used for the mouse model and the LPS-induced stimulation of BV-2 cells was used for the cell model. The improvement of motor function after SCI was assessed by open field, swimming, and footprint experiments. The morphological characteristics of mouse spinal cord tissue and the levels of INOS, Sesn2, TNF-α, Keap1, Nrf2, IL-10, and reactive oxygen species (ROS) in vivo and in vitro were measured by several methods including western blotting, qPCR, immunofluorescence, HE, and Nissl staining. In vivo data showed that tangeretin can improve motor function recovery and reduce neuron loss and injury size in mice with SCI. Simultaneously, the in vitro findings suggested that treatment of BV-2 cells with tangeretin after LPS stimulation reduced the production of inflammatory factors and ROS, and could convert BV-2 cells from the M1 to the M2 type. Furthermore, Sesn2 knockout suppressed Keap1/Nrf2, inflammatory factors, ROS levels, and the M1 to M2 transition. Tangeretin can alleviate the inflammation and oxidative response induced by SCI by activating the Sesn2/Keap1/Nrf2 pathway.
Collapse
Affiliation(s)
- Birong Peng
- Department of Spinal Surgery, Orthopedic Medical Center, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Jinwei Hu
- Department of Spinal Surgery, Orthopedic Medical Center, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Yuanfang Sun
- Department of Spinal Surgery, Orthopedic Medical Center, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Yating Huang
- Department of Endocrinology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Qingshan Peng
- Department of Spinal Surgery, Orthopedic Medical Center, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Weiwen Zhao
- Department of Orthopedics, Hunan Provincial People's Hospital, Hunan Normal University, Changsha, Hunan Province, China
| | - Wenning Xu
- Department of Spinal Surgery, Orthopedic Medical Center, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Lixin Zhu
- Department of Spinal Surgery, Orthopedic Medical Center, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| |
Collapse
|
17
|
Wang F, Xiao J, Li M, He Q, Wang X, Pan Z, Li S, Wang H, Zhou C. Picroside II suppresses chondrocyte pyroptosis through MAPK/NF-κB/NLRP3 signaling pathway alleviates osteoarthritis. PLoS One 2024; 19:e0308731. [PMID: 39208260 PMCID: PMC11361613 DOI: 10.1371/journal.pone.0308731] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Accepted: 07/29/2024] [Indexed: 09/04/2024] Open
Abstract
BACKGROUND Picroside II (P-II) is the main bioactive constituent of Picrorhiza Kurroa, a traditional Chinese herb of interest for its proven anti-inflammatory properties. Its beneficial effects have been noted across several physiological systems, including the nervous, circulatory, and digestive, capable of treating a wide range of diseases. Nevertheless, the potential of Picroside II to treat osteoarthritis (OA) and the mechanisms behind its efficacy remain largely unexplored. AIM This study aims to evaluate the efficacy of Picroside II in the treatment of osteoarthritis and its potential molecular mechanisms. METHODS In vitro, we induced cellular inflammation in chondrocytes with lipopolysaccharide (LPS) and subsequently treated with Picroside II to assess protective effect on chondrocyte. We employed the Cell Counting Kit-8 (CCK-8) assay to assess the impact of Picroside II on cell viability and select the optimal Picroside II concentration for subsequent experiments. We explored the effect of Picroside II on chondrocyte pyroptosis and its underlying molecular mechanisms by qRT-PCR, Western blot (WB) and immunofluorescence. In vivo, we established the destabilization of the medial meniscus surgery to create an OA mouse model. The therapeutic effects of Picroside II were then assessed through Micro-CT scanning, Hematoxylin-eosin (H&E) staining, Safranin O-Fast Green (S&F) staining, immunohistochemistry and immunofluorescence. RESULTS In in vitro studies, toluidine blue and CCK-8 results showed that a certain concentration of Picroside II had a restorative effect on the viability of chondrocytes inhibited by LPS. Picroside II notably suppressed the expression levels of caspase-1, IL-18, and IL-1β, which consequently led to the reduction of pyroptosis. Moreover, Picroside II was shown to decrease NLRP3 inflammasome activation, via the MAPK/NF-κB signaling pathway. In vivo studies have shown that Picroside II can effectively reduce subchondral bone destruction and osteophyte formation in the knee joint of mice after DMM surgery. CONCLUSIONS Our research suggests that Picroside II can inhibit chondrocyte pyroptosis and ameliorate osteoarthritis progression by modulating the MAPK/NF-κB signaling pathway.
Collapse
Affiliation(s)
- Fanchen Wang
- First School of Clinical Medicine, Guangzhou University of Chinese Medicine, Guangzhou, China
- The Laboratory of Orthopedics and Traumatology of Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Jiacong Xiao
- First School of Clinical Medicine, Guangzhou University of Chinese Medicine, Guangzhou, China
- The Laboratory of Orthopedics and Traumatology of Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Miao Li
- First School of Clinical Medicine, Guangzhou University of Chinese Medicine, Guangzhou, China
- The Laboratory of Orthopedics and Traumatology of Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Qi He
- First School of Clinical Medicine, Guangzhou University of Chinese Medicine, Guangzhou, China
- The Laboratory of Orthopedics and Traumatology of Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Xintian Wang
- First School of Clinical Medicine, Guangzhou University of Chinese Medicine, Guangzhou, China
- The Laboratory of Orthopedics and Traumatology of Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Zhaofeng Pan
- The Laboratory of Orthopedics and Traumatology of Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Shaocong Li
- First School of Clinical Medicine, Guangzhou University of Chinese Medicine, Guangzhou, China
- The Laboratory of Orthopedics and Traumatology of Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Haibin Wang
- First School of Clinical Medicine, Guangzhou University of Chinese Medicine, Guangzhou, China
- Department of Orthopedic Surgery, The First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Chi Zhou
- Department of Orthopedic Surgery, The First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, China
- Maoming Hospital of Guangzhou University of Chinese Medicine, Maoming, China
| |
Collapse
|
18
|
Sai Priya T, Ramalingam V, Suresh Babu K. Natural products: A potential immunomodulators against inflammatory-related diseases. Inflammopharmacology 2024:10.1007/s10787-024-01562-4. [PMID: 39196458 DOI: 10.1007/s10787-024-01562-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Accepted: 08/14/2024] [Indexed: 08/29/2024]
Abstract
The incidence and prevalence of inflammatory-related diseases (IRDs) are increasing worldwide. Current approved treatments for IRDs in the clinic are combat against inhibiting the pro-inflammatory cytokines. Though significant development in the treatment in the IRDs has been achieved, the severe side effects and inefficiency of currently practicing treatments are endless challenge. Drug discovery from natural sources is efficacious over a resurgence and also natural products are leading than the synthetic molecules in both clinical trials and market. The use of natural products against IRDs is a conventional therapeutic approach since it is a reservoir of unique structural chemistry, accessibility and bioactivities with reduced side effects and low toxicity. In this review, we discuss the cause of IRDs, treatment of options for IRDs and the impact and adverse effects of currently practicing clinical drugs. As well, the significant role of natural products against various IRDs, the limitations in the clinical development of natural products and thus pave the way for development of natural products as immunomodulators against IRDs are also discussed.
Collapse
Affiliation(s)
- Telukuntla Sai Priya
- Department of Natural Products & Medicinal Chemistry, CSIR-Indian Institute of Chemical Technology, Hyderabad, 500 007, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Vaikundamoorthy Ramalingam
- Department of Natural Products & Medicinal Chemistry, CSIR-Indian Institute of Chemical Technology, Hyderabad, 500 007, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Katragadda Suresh Babu
- Department of Natural Products & Medicinal Chemistry, CSIR-Indian Institute of Chemical Technology, Hyderabad, 500 007, India.
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India.
| |
Collapse
|
19
|
Yao Z, Gan F, Zeng Y, Ren L, Zeng Y. Elucidating Cyathula Officinals' mechanism in osteoarthritis treatment: Network pharmacology and empirical evidence on anti-inflammatory actions. Heliyon 2024; 10:e27999. [PMID: 38524622 PMCID: PMC10958415 DOI: 10.1016/j.heliyon.2024.e27999] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2023] [Revised: 03/06/2024] [Accepted: 03/11/2024] [Indexed: 03/26/2024] Open
Abstract
In this study, we explored the therapeutic potential of Cyathula Officinals (CNX) in Knee Osteoarthritis (KOA) treatment. Utilizing network pharmacology and in vitro experiments, we identified active ingredients, action targets and pathways in CNX. Our analysis, integrating databases like TCMSP, SwissTarget Prediction, Genecards, CTD, STRING, and DAVID, highlighted 396 action targets and 283 disease targets, pinpointing 64 intersection genes linked to KOA. The significant involvement of the MAPK and NF-κB pathways in CNX's anti-inflammatory action was validated through qPCR, which might underlie CNX's efficacy in inhibiting chondrocyte apoptosis and IL-6 expression. These findings suggest CNX's potential in KOA management, offering insights for its clinical application.
Collapse
Affiliation(s)
- Zhicheng Yao
- The First Clinical Medical School, Guangzhou University of Chinese Medicine, China
- Shenzhen Hospital, Beijing University of Chinese Medicine, China
| | - Fengping Gan
- The First Clinical Medical School, Guangzhou University of Chinese Medicine, China
| | - Yuqing Zeng
- The First Clinical Medical School, Guangzhou University of Chinese Medicine, China
| | - Litong Ren
- Shenzhen Hospital, Beijing University of Chinese Medicine, China
| | - Yirong Zeng
- The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Department of Orthopedics, China
| |
Collapse
|
20
|
Wang X, Li X, Zhou J, Lei Z, Yang X. Fisetin suppresses chondrocyte senescence and attenuates osteoarthritis progression by targeting sirtuin 6. Chem Biol Interact 2024; 390:110890. [PMID: 38278314 DOI: 10.1016/j.cbi.2024.110890] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2023] [Revised: 01/07/2024] [Accepted: 01/23/2024] [Indexed: 01/28/2024]
Abstract
Osteoarthritis (OA) is the most common type of arthritis and is an age-related joint disease that is particularly prevalent in subjects over 65 years old. The chronic rise of senescent cells has a close correlation with age-related diseases such as OA, and the senescence-associated secretory phenotype (SASP) is implicated in OA cartilage degeneration pathogenesis. Sirtuin 6 (SIRT6) is likely to be a key senescence-related regulator. Fisetin (FST) is a natural flavonol of the flavonoid family that is recommended as a senolytic drug to extend health and lifespan. However, the potential chondroprotective effects of FST on OA rats are largely unclarified. The aim of this study is to investigate the ameliorative effects of FST on OA joint cartilage and the relationship with SIRT6 and the detailed mechanisms from anti-inflammatory and anti-senescent perspectives. Rats were subjected to destabilization of the medial meniscus (DMM) surgery as a means of inducing the experimental OA model in vivo. Chondrocytes treated with IL-1β were utilized for mimicking the OA cell model in vitro. Intra-articular injection of FST, OSS_128,167 (OSS, SIRT6 inhibitor), and MDL800 (MDL, SIRT6 agonist) in vivo or administering them in IL-1β-induced rat chondrocytes in vitro were performed in order to determine the effects FST has on OA and the link with SIRT6. This study found SIRT6 level to be negatively correlated with OA severity. SIRT6 downregulation was validated in the joint cartilages of DMM rats and IL-1β-treated chondrocytes. It was also notably demonstrated that FST can activate SIRT6. Both the administration of FST and activation of SIRT6 using MDL were found to rescue cartilage erosion, decrease extracellular matrix (ECM) degradation, prevent cartilage from apoptosis, and improve detrimental senescence-related phenotype. The alleviative effects of FST against inflammation, ECM degradation, apoptosis, and senescence in IL-1β-stimulated chondrocytes were also confirmed. SIRT6 loss occurs in articular cartilage in OA pathogenesis, which is linked to aging. FST attenuates injury-induced aging-related phenotype changes in chondrocytes through the targeting of SIRT6.
Collapse
Affiliation(s)
- Xuezhong Wang
- Department of Orthopedics, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Xuyang Li
- Department of Orthopedics, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Jianlin Zhou
- Department of Orthopedics, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Zheng Lei
- Department of Emergency Medicine, China-Japan Union Hospital of Jilin University, Changchun, 130033, China
| | - Xiaoming Yang
- Department of Orthopedics, Renmin Hospital of Wuhan University, Wuhan, 430060, China.
| |
Collapse
|
21
|
You G, Zheng L, Zhang Y, Zhang Y, Wang Y, Guo W, Liu H, Tatiana P, Vladimir K, Zan J. Tangeretin Attenuates Cerebral Ischemia-Reperfusion-Induced Neuronal Pyroptosis by Inhibiting AIM2 Inflammasome Activation via Regulating NRF2. Inflammation 2024; 47:145-158. [PMID: 37725272 DOI: 10.1007/s10753-023-01900-8] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2023] [Revised: 08/27/2023] [Accepted: 09/05/2023] [Indexed: 09/21/2023]
Abstract
Pyroptosis is closely involved in the pathopoiesis of cerebral ischemia and reperfusion (I/R) injury which seriously dangers human's life. Studies report that tangeretin (TANG), which is enriched in the peel of Citrus reticulata, has neuroprotective effects. Here, we explored whether absent in melanoma 2 (AIM2) inflammasome-mediated pyroptosis is involved in the cerebral I/R injury and the protective mechanism of TANG against cerebral I/R injury. In this study, we found that TANG treatment effectively alleviated I/R-induced brain injury and inhibited neuronal pyroptosis in an in vivo mice model with middle cerebral artery occlusion/reperfusion (MCAO/R) injury and in an in vitro hippocampal HT22 cell model with oxygen-glucose deprivation and reoxygenation (OGD/R) injury. Furthermore, we found TANG inhibited cerebral I/R-induced neuronal AIM2 inflammasome activation in vivo and in vitro via regulating nuclear factor E2-related factor 2 (NRF2). Moreover, administration of ML385, a chemical inhibitor of NRF2, notably blocked the neuroprotective effects of TANG against cerebral I/R injury. In conclusion, TANG attenuates cerebral I/R-induced neuronal pyroptosis by inhibiting AIM2 inflammasome activation via regulating NRF2. These findings indicate TANG is a potential therapeutic agent for cerebral I/R injury.
Collapse
Affiliation(s)
- Guoxing You
- School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou, 510006, China
| | - Linbo Zheng
- Department of Traditional Chinese Medicine, Guangdong Second Provincial General Hospital, Guangzhou, 510310, China
| | - Yuanyuan Zhang
- The Affiliated Traditional Chinese Medicine Hospital of Guangzhou Medical University, Guangzhou, 510130, China
| | - Yuting Zhang
- School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou, 510006, China
| | - Yupeng Wang
- School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou, 510006, China
| | - Wenjie Guo
- School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou, 510006, China
| | - Hao Liu
- Department of Traditional Chinese Medicine, Guangdong Second Provincial General Hospital, Guangzhou, 510310, China
| | - Philipovich Tatiana
- Institute of Physiology, National Academy of Sciences of Belarus, Minsk, 220072, Republic of Belarus
| | - Kulchitsky Vladimir
- Institute of Physiology, National Academy of Sciences of Belarus, Minsk, 220072, Republic of Belarus
| | - Jie Zan
- School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou, 510006, China.
| |
Collapse
|
22
|
Liu Y, Zhang Y, You G, Zheng D, He Z, Guo W, Antonina K, Shukhrat Z, Ding B, Zan J, Zhang Z. Tangeretin attenuates acute lung injury in septic mice by inhibiting ROS-mediated NLRP3 inflammasome activation via regulating PLK1/AMPK/DRP1 signaling axis. Inflamm Res 2024; 73:47-63. [PMID: 38147126 DOI: 10.1007/s00011-023-01819-8] [Citation(s) in RCA: 16] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Revised: 10/05/2023] [Accepted: 11/06/2023] [Indexed: 12/27/2023] Open
Abstract
OBJECTIVE NLRP3 inflammasome-mediated pyroptosis of macrophage acts essential roles in the progression of sepsis-induced acute lung injury (ALI). Tangeretin (TAN), enriched in citrus fruit peel, presents anti-oxidative and anti-inflammatory effects. Here, we aimed to explore the potentially protective effect of TAN on sepsis-induced ALI, and the underlying mechanism of TAN in regulating NLRP3 inflammasome. MATERIAL AND METHODS The effect of TAN on sepsis-induced ALI and NLRP3 inflammasome-mediated pyroptosis of macrophage were examined in vivo and in vitro using a LPS-treated mice model and LPS-induced murine macrophages, respectively. The mechanism of TAN regulating the activation of NLRP3 inflammasome in sepsis-induced ALI was investigated with HE staining, Masson staining, immunofluorescent staining, ELISA, molecular docking, transmission electron microscope detection, qRT-PCR, and western blot. RESULTS TAN could evidently attenuate sepsis-induced ALI in mice, evidenced by reducing pulmonary edema, pulmonary congestion and lung interstitial fibrosis, and inhibiting macrophage infiltration in the lung tissue. Besides, TAN significantly suppressed inflammatory cytokine IL-1β and IL-18 expression in the serum or bronchoalveolar lavage fluid (BALF) samples of mice with LPS-induced ALI, and inhibited NLRP3 inflammasome-mediated pyroptosis of macrophages. Furthermore, we found TAN inhibited ROS production, preserved mitochondrial morphology, and alleviated excessive mitochondrial fission in LPS-induced ALI in mice. Through bioinformatic analysis and molecular docking, Polo-like kinase 1 (PLK1) was identified as a potential target of TAN for treating sepsis-induced ALI. Moreover, TAN significantly inhibited the reduction of PLK1 expression, AMP-activated protein kinase (AMPK) phosphorylation, and Dynamin related protein 1 (Drp1) phosphorylation (S637) in LPS-induced ALI in mice. In addition, Volasertib, a specific inhibitor of PLK1, abolished the protective effects of TAN against NLRP3 inflammasome-mediated pyroptosis of macrophage and lung injury in the cell and mice septic models. CONCLUSION TAN attenuates sepsis-induced ALI by inhibiting ROS-mediated NLRP3 inflammasome activation via regulating PLK1/AMPK/DRP1 signaling axis, and TAN is a potentially therapeutic candidate against ALI through inhibiting pyroptosis.
Collapse
Affiliation(s)
- Yuntao Liu
- State Key Laboratory of Traditional Chinese Medicine Syndrom,The second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, 510120, China
| | - Yuting Zhang
- School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou, 510006, China
| | - Guoxing You
- School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou, 510006, China
| | - Danwen Zheng
- State Key Laboratory of Traditional Chinese Medicine Syndrom,The second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, 510120, China
| | - Zhipeng He
- State Key Laboratory of Traditional Chinese Medicine Syndrom,The second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, 510120, China
| | - Wenjie Guo
- School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou, 510006, China
| | - Kim Antonina
- No. 1 Department of Internal Diseases, Samarkand State Medical University, Samarkand, Uzbekistan
| | - Ziyadullaev Shukhrat
- No. 1 Department of Internal Diseases, Samarkand State Medical University, Samarkand, Uzbekistan
| | - Banghan Ding
- State Key Laboratory of Traditional Chinese Medicine Syndrom,The second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, 510120, China.
| | - Jie Zan
- School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou, 510006, China.
| | - Zhongde Zhang
- State Key Laboratory of Traditional Chinese Medicine Syndrom,The second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, 510120, China.
| |
Collapse
|
23
|
Park WY, Byun EH. Capsicum annuum var. abbreviatum Induces a Macrophage M2-Like Phenotype Mediated by Nrf2/HO-1 in LPS-Stimulated Macrophages. J Med Food 2024; 27:88-94. [PMID: 38236694 DOI: 10.1089/jmf.2023.k.0031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2024] Open
Abstract
Capsicum annuum var. abbreviatum (CAAE), which is in the genus Capsicum L. (Solanaceae), was found to be richer in polyphenols and flavonoids than other prevalent peppers of Capsicum annuum var. angulosum and Capsicum annuum. L. Yet, it is still unclear how CAAE reduces inflammation. In this study, we used the lipopolysaccharide-stimulated RAW264.7 macrophage cell line and bone marrow-derived macrophages to assess its anti-inflammatory activities. Initially, we discovered that CAAE decreased the levels of nitric oxide and inducible nitric oxide synthase. In addition, CAAE decreased the intracellular reactive oxygen species levels and increased the nuclear factor-erythroid 2-related factor 2 and heme oxygenase-1 compared with the phenotype of M2 macrophages. CAAE inhibited the activation of mitogen-activated protein kinases (MAPKs), including extracellular signal-regulated kinases, c-Jun N-terminal kinases, and p38 MAPKs. CAAE also inhibited the translocation of nuclear factor kappa B into nuclear, hence preventing the production of proinflammatory cytokines. Therefore, we suggest that CAAE might have potential as a candidate therapeutic agent for inflammatory diseases.
Collapse
Affiliation(s)
- Woo Yong Park
- Department of Pharmacology, College of Korean Medicine, Kyung Hee University, Seoul, Korea
| | - Eui-Hong Byun
- Department of Food Science and Technology, Kongju National University, Yesan, Korea
| |
Collapse
|
24
|
Oh E, Jang H, Ok S, Eom J, Lee H, Kim SH, Kim JH, Jeong YM, Kim KJ, Yun SP, Kwon HJ, Lee IC, Park JY, Yang S. WGA-M001, a Mixture of Total Extracts of Tagetes erecta and Ocimum basilicum, Synergistically Alleviates Cartilage Destruction by Inhibiting ERK and NF-κB Signaling. Int J Mol Sci 2023; 24:17459. [PMID: 38139287 PMCID: PMC10743532 DOI: 10.3390/ijms242417459] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Revised: 12/11/2023] [Accepted: 12/12/2023] [Indexed: 12/24/2023] Open
Abstract
Tagetes erecta and Ocimum basilicum are medicinal plants that exhibit anti-inflammatory effects against various diseases. However, their individual and combined effects on osteoarthritis (OA) are unknown. Herein, we aimed to demonstrate the effects of T. erecta, O. basilicum, and their mixture, WGA-M001, on OA pathogenesis. The administration of total extracts of T. erecta and O. basilicum reduced cartilage degradation and inflammation without causing cytotoxicity. Although WGA-M001 contained lower concentrations of the individual extracts, it strongly inhibited the expression of pathogenic factors. In vivo OA studies also supported that WGA-M001 had protective effects against cartilage destruction at lower doses than those of T. erecta and O. basilicum. Moreover, its effects were stronger than those observed using Boswellia and Perna canaliculus. WGA-M001 effectively inhibited the interleukin (IL)-1β-induced nuclear factor kappa-light-chain-enhancer of the activated B cell (NF-κB) pathway and ERK phosphorylation. Furthermore, RNA-sequence analysis also showed that WGA-M001 decreased the expression of genes related to the IL-1β-induced NF-κB and ERK signaling pathways. Therefore, WGA-M001 is more effective than the single total extracts of T. erecta and O. basilicum in attenuating OA progression by regulating ERK and NF-κB signaling. Our results open new possibilities for WGA-M001 as a potential therapeutic agent for OA treatment.
Collapse
Affiliation(s)
- Eunjeong Oh
- Department of Biological Sciences, Sungkyunkwan University, Suwon 16419, Republic of Korea; (E.O.); (H.J.); (S.O.); (J.E.)
- Department of Biomedical Sciences, Ajou University Graduate School of Medicine, Suwon 16499, Republic of Korea
| | - Hahyeong Jang
- Department of Biological Sciences, Sungkyunkwan University, Suwon 16419, Republic of Korea; (E.O.); (H.J.); (S.O.); (J.E.)
| | - Subin Ok
- Department of Biological Sciences, Sungkyunkwan University, Suwon 16419, Republic of Korea; (E.O.); (H.J.); (S.O.); (J.E.)
| | - Jiwon Eom
- Department of Biological Sciences, Sungkyunkwan University, Suwon 16419, Republic of Korea; (E.O.); (H.J.); (S.O.); (J.E.)
| | - Hyunyong Lee
- Wooree Green Science, Ansan 15409, Republic of Korea; (H.L.); (S.H.K.); (J.H.K.); (Y.M.J.); (K.J.K.)
| | - Sung Hun Kim
- Wooree Green Science, Ansan 15409, Republic of Korea; (H.L.); (S.H.K.); (J.H.K.); (Y.M.J.); (K.J.K.)
| | - Jong Hwa Kim
- Wooree Green Science, Ansan 15409, Republic of Korea; (H.L.); (S.H.K.); (J.H.K.); (Y.M.J.); (K.J.K.)
| | - Yu Mi Jeong
- Wooree Green Science, Ansan 15409, Republic of Korea; (H.L.); (S.H.K.); (J.H.K.); (Y.M.J.); (K.J.K.)
| | - Kyeong Jin Kim
- Wooree Green Science, Ansan 15409, Republic of Korea; (H.L.); (S.H.K.); (J.H.K.); (Y.M.J.); (K.J.K.)
- Department of Horticulture, Chonnam National University, Gwangju 61186, Republic of Korea
| | - Seung Pil Yun
- Department of Pharmacology, Institute of Health Sciences, Gyeongsang National University College of Medicine, Jinju 52727, Republic of Korea;
| | - Hyung-Jun Kwon
- Center for Companion Animal New Drug Development, Jeonbuk Branch, Korea Institute of Toxicology, Jeongeup 53212, Republic of Korea; (H.-J.K.); (I.-C.L.); (J.-Y.P.)
| | - In-Chul Lee
- Center for Companion Animal New Drug Development, Jeonbuk Branch, Korea Institute of Toxicology, Jeongeup 53212, Republic of Korea; (H.-J.K.); (I.-C.L.); (J.-Y.P.)
| | - Ji-Young Park
- Center for Companion Animal New Drug Development, Jeonbuk Branch, Korea Institute of Toxicology, Jeongeup 53212, Republic of Korea; (H.-J.K.); (I.-C.L.); (J.-Y.P.)
| | - Siyoung Yang
- Department of Biological Sciences, Sungkyunkwan University, Suwon 16419, Republic of Korea; (E.O.); (H.J.); (S.O.); (J.E.)
| |
Collapse
|
25
|
Cao N, Wang D, Liu B, Wang Y, Han W, Tian J, Xiang L, Wang Z. Silencing of STUB1 relieves osteoarthritis via inducing NRF2-mediated M2 macrophage polarization. Mol Immunol 2023; 164:112-122. [PMID: 37992540 DOI: 10.1016/j.molimm.2023.11.010] [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: 08/01/2023] [Revised: 11/07/2023] [Accepted: 11/15/2023] [Indexed: 11/24/2023]
Abstract
OBJECTIVES Shifting macrophages towards an anti-inflammatory state is key in treating osteoarthritis (OA) by reducing inflammation and tissue damage. However, the underlying mechanisms guiding this shift remain largely undefined. STUB1, an E3 ubiquitin ligase, known for its regulatory role in macrophage polarization. This study aims to explore the function and underlying action mechanisms of STUB1 in OA. METHODS An in vivo OA model was established in rats. Hematoxylin-Eosin and safranin O-fast green staining were performed to reveal the hispathological injuries in knee-joint tissues. Immunohistochemistry and flow cytometry were performed to detect the distribution of M1 and M2 macrophages. The inflammatory response (TNF-α and IL-6 levels) was evaluated by ELISA. In vitro, the interaction between STUB1 and NFR2 was determined by CO-IP and pull-down assays. After treated with LPS (an in vitro model of OA), the viability and apoptosis of chondrocytes were measured by CCK-8 and flow cytometry, respectively. RESULTS Silencing STUB1 alleviated OA in rats, as indicated by reduced subchondral bone thickness, knee synovitis score, histopathological damages, and inflammatory response. STUB1 silencing also decreased M1 macrophages and increased M2 macrophages in both in vivo and in vitro settings. NRF2 was identified as a target of STUB1, with STUB1 mediating its ubiquitination. Silencing NRF2 reversed the effects of STUB1 silencing on inducing M2 macrophage polarization. Furthermore, silencing STUB1 upregulated NRF2 expression in LPS-treated chondrocytes, promoting cell viability and inhibiting apoptosis. CONCLUSION Silencing STUB1 induces M2 macrophage polarization by inhibiting NRF2 ubiquitination, thereby contributing to the mitigation of OA.
Collapse
Affiliation(s)
- Nan Cao
- Department of Orthopedics, General Hospital of Northern Theater Command, Shenyang 110016, Liaoning Province, China
| | - Danni Wang
- Department of Orthopedics, General Hospital of Northern Theater Command, Shenyang 110016, Liaoning Province, China
| | - Bin Liu
- Department of Orthopedics, General Hospital of Northern Theater Command, Shenyang 110016, Liaoning Province, China
| | - Yu Wang
- Department of Orthopedics, General Hospital of Northern Theater Command, Shenyang 110016, Liaoning Province, China
| | - Wenfeng Han
- Department of Orthopedics, General Hospital of Northern Theater Command, Shenyang 110016, Liaoning Province, China
| | - Jing Tian
- Department of Orthopedics, General Hospital of Northern Theater Command, Shenyang 110016, Liaoning Province, China
| | - Liangbi Xiang
- Department of Orthopedics, General Hospital of Northern Theater Command, Shenyang 110016, Liaoning Province, China.
| | - Zheng Wang
- Department of Orthopedics, General Hospital of Northern Theater Command, Shenyang 110016, Liaoning Province, China.
| |
Collapse
|
26
|
Che J, Yang X, Jin Z, Xu C. Nrf2: A promising therapeutic target in bone-related diseases. Biomed Pharmacother 2023; 168:115748. [PMID: 37865995 DOI: 10.1016/j.biopha.2023.115748] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Revised: 10/15/2023] [Accepted: 10/17/2023] [Indexed: 10/24/2023] Open
Abstract
Nuclear factor erythroid-2-related factor 2 (Nrf2) plays an important role in maintaining cellular homeostasis, as it suppresses cell damage caused by external stimuli by regulating the transcription of intracellular defense-related genes. Accumulating evidence has highlighted the crucial role of reduction-oxidation (REDOX) imbalance in the development of bone-related diseases. Nrf2, a transcription factor linked to nuclear factor-erythrocyte 2, plays a pivotal role in the regulation of oxidative stress and induction of antioxidant defenses. Therefore, further investigation of the mechanism and function of Nrf2 in bone-related diseases is essential. Considerable evidence suggests that increased nuclear transcription of Nrf2 in response to external stimuli promotes the expression of intracellular antioxidant-related genes, which in turn leads to the inhibition of bone remodeling imbalance, improved fracture recovery, reduced occurrence of osteoarthritis, and greater tumor resistance. Certain natural extracts can selectively target Nrf2, potentially offering therapeutic benefits for osteogenic arthropathy. In this article, the biological characteristics of Nrf2 are reviewed, the intricate interplay between Nrf2-regulated REDOX imbalance and bone-related diseases is explored, and the potential preventive and protective effects of natural products targeting Nrf2 in these diseases are elucidated. A comprehensive understanding of the role of Nrf2 in the development of bone-related diseases provides valuable insights into clinical interventions and can facilitate the discovery of novel Nrf2-targeting drugs.
Collapse
Affiliation(s)
- Jingmin Che
- Shaanxi Provincial Key Laboratory of Infection and Immune Diseases, Shaanxi Provincial People's Hospital, Xi'an, Shaanxi, China; Shaanxi Engineering Research Center of Cell Immunology, Shaanxi Provincial People's Hospital, Xi'an, Shaanxi, China.
| | - Xiaoli Yang
- Shaanxi Provincial Key Laboratory of Infection and Immune Diseases, Shaanxi Provincial People's Hospital, Xi'an, Shaanxi, China; Shaanxi Engineering Research Center of Cell Immunology, Shaanxi Provincial People's Hospital, Xi'an, Shaanxi, China
| | - Zhankui Jin
- Department of Orthopedics, Shaanxi Provincial People's Hospital, Xi'an, Shaanxi, China.
| | - Cuixiang Xu
- Shaanxi Provincial Key Laboratory of Infection and Immune Diseases, Shaanxi Provincial People's Hospital, Xi'an, Shaanxi, China; Shaanxi Engineering Research Center of Cell Immunology, Shaanxi Provincial People's Hospital, Xi'an, Shaanxi, China
| |
Collapse
|
27
|
Huang W, Zhong Y, Gao B, Zheng B, Liu Y. Nrf2-mediated therapeutic effects of dietary flavones in different diseases. Front Pharmacol 2023; 14:1240433. [PMID: 37767395 PMCID: PMC10520786 DOI: 10.3389/fphar.2023.1240433] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Accepted: 08/30/2023] [Indexed: 09/29/2023] Open
Abstract
Oxidative stress (OS) is a pathological status that occurs when the body's balance between oxidants and antioxidant defense systems is broken, which can promote the development of many diseases. Nrf2, a redox-sensitive transcription encoded by NFE2L2, is the master regulator of phase II antioxidant enzymes and cytoprotective genes. In this context, Nrf2/ARE signaling can be a compelling target against OS-induced diseases. Recently, natural Nrf2/ARE regulators like dietary flavones have shown therapeutic potential in various acute and chronic diseases such as diabetes, neurodegenerative diseases, ischemia-reperfusion injury, and cancer. In this review, we aim to summarize nrf2-mediated protective effects of flavones in different conditions. Firstly, we retrospected the mechanisms of how flavones regulate the Nrf2/ARE pathway and introduced the mediator role Nrf2 plays in inflammation and apoptosis. Then we review the evidence that flavones modulated Nrf2/ARE pathway to prevent diseases in experimental models. Based on these literature, we found that flavones could regulate Nrf2 expression by mechanisms below: 1) dissociating the binding between Nrf2 and Keap1 via PKC-mediated Nrf2 phosphorylation and P62-mediated Keap1 autophagic degradation; 2) regulating Nrf2 nuclear translocation by various kinases like AMPK, MAPKs, Fyn; 3) decreasing Nrf2 ubiquitination and degradation via activating sirt1 and PI3K/AKT-mediated GSK3 inhibition; and 4) epigenetic alternation of Nrf2 such as demethylation at the promoter region and histone acetylation. In conclusion, flavones targeting Nrf2 can be promising therapeutic agents for various OS-related disorders. However, there is a lack of investigations on human subjects, and new drug delivery systems to improve flavones' treatment efficiency still need to be developed.
Collapse
Affiliation(s)
- Wenkai Huang
- Liaoning Provincial Key Laboratory of Oral Disease, School and Hospital of Stomatology, China Medical University, Shenyang, China
| | - Yuan Zhong
- Liaoning Provincial Key Laboratory of Oral Disease, School and Hospital of Stomatology, China Medical University, Shenyang, China
| | - Botao Gao
- Liaoning Provincial Key Laboratory of Oral Disease, School and Hospital of Stomatology, China Medical University, Shenyang, China
| | - Bowen Zheng
- Liaoning Provincial Key Laboratory of Oral Disease, Department of Orthodontics, School and Hospital of Stomatology, China Medical University, Shenyang, China
| | - Yi Liu
- Liaoning Provincial Key Laboratory of Oral Disease, Department of Orthodontics, School and Hospital of Stomatology, China Medical University, Shenyang, China
| |
Collapse
|
28
|
Wu J, Yu H, Jin Y, Wang J, Zhou L, Cheng T, Zhang Z, Lin B, Miao J, Lin Z. Ajugol's upregulation of TFEB-mediated autophagy alleviates endoplasmic reticulum stress in chondrocytes and retards osteoarthritis progression in a mouse model. Chin Med 2023; 18:113. [PMID: 37679844 PMCID: PMC10483732 DOI: 10.1186/s13020-023-00824-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Accepted: 08/24/2023] [Indexed: 09/09/2023] Open
Abstract
BACKGROUND Osteoarthritis (OA), a degenerative disease with a high global prevalence, is characterized by the degradation of the extracellular matrix (ECM) and the apoptosis of chondrocytes. Ajugol, a extract derived from the herb Rehmannia glutinosa, has not yet been investigated for its potential in modulating the development of OA. METHODS We employed techniques such as western blotting, immunofluorescence, immunohistochemistry, X-ray imaging, HE staining, and SO staining to provide biological evidence supporting the role of Ajugol as a potential therapeutic agent for modulating OA. Furthermore, in an in vivo experiment, intra-peritoneal injection of 50 mg/kg Ajugol effectively mitigated the progression of OA following destabilization of the medial meniscus (DMM) surgery. RESULTS Our findings revealed that treatment with 50 μM Ajugol activated TFEB-mediated autophagy, alleviating ER stress-induced chondrocyte apoptosis and ECM degradation caused by TBHP. Furthermore, in an in vivo experiment, intra-peritoneal injection of 50 mg/kg Ajugol effectively mitigated the progression of OA following destabilization of the medial meniscus (DMM) surgery. CONCLUSION These results provide compelling biological evidence supporting the role of Ajugol as a potential therapeutic agent for modulating OA by activating autophagy and attenuating ER stress-induced cell death and ECM degradation. The promising in vivo results further suggest the potential of Ajugol as a treatment strategy for OA progression.
Collapse
Affiliation(s)
- Jingtao Wu
- Department of Orthopaedics, Wenzhou Key Laboratory of Perinatal Medicine, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325000, Zhejiang Province, China
- Key Laboratory of Orthopaedics of Zhejiang Province, Wenzhou, 325000, Zhejiang Province, China
- The Second School of Medicine, Wenzhou Medical University, Wenzhou, 325000, Zhejiang Province, China
| | - Heng Yu
- Department of Orthopaedics, Wenzhou Key Laboratory of Perinatal Medicine, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325000, Zhejiang Province, China
- Key Laboratory of Orthopaedics of Zhejiang Province, Wenzhou, 325000, Zhejiang Province, China
- The Second School of Medicine, Wenzhou Medical University, Wenzhou, 325000, Zhejiang Province, China
| | - Yangcan Jin
- Department of Orthopaedics, Wenzhou Key Laboratory of Perinatal Medicine, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325000, Zhejiang Province, China
- Key Laboratory of Orthopaedics of Zhejiang Province, Wenzhou, 325000, Zhejiang Province, China
- The Second School of Medicine, Wenzhou Medical University, Wenzhou, 325000, Zhejiang Province, China
| | - Jingquan Wang
- Department of Orthopaedics, Wenzhou Key Laboratory of Perinatal Medicine, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325000, Zhejiang Province, China
- Key Laboratory of Orthopaedics of Zhejiang Province, Wenzhou, 325000, Zhejiang Province, China
- The Second School of Medicine, Wenzhou Medical University, Wenzhou, 325000, Zhejiang Province, China
| | - Liwen Zhou
- The First School of Medicine, Wenzhou Medical University, Wenzhou, 325000, Zhejiang Province, China
| | - Teng Cheng
- Department of Orthopaedics, Wenzhou Key Laboratory of Perinatal Medicine, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325000, Zhejiang Province, China
- Key Laboratory of Orthopaedics of Zhejiang Province, Wenzhou, 325000, Zhejiang Province, China
- The Second School of Medicine, Wenzhou Medical University, Wenzhou, 325000, Zhejiang Province, China
| | - Zhao Zhang
- Department of Orthopaedics, Wenzhou Key Laboratory of Perinatal Medicine, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325000, Zhejiang Province, China
- Key Laboratory of Orthopaedics of Zhejiang Province, Wenzhou, 325000, Zhejiang Province, China
- The Second School of Medicine, Wenzhou Medical University, Wenzhou, 325000, Zhejiang Province, China
| | - Binghao Lin
- Department of Orthopaedics, Wenzhou Key Laboratory of Perinatal Medicine, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325000, Zhejiang Province, China
- Key Laboratory of Orthopaedics of Zhejiang Province, Wenzhou, 325000, Zhejiang Province, China
- The Second School of Medicine, Wenzhou Medical University, Wenzhou, 325000, Zhejiang Province, China
| | - Jiansen Miao
- Department of Orthopaedics, Wenzhou Key Laboratory of Perinatal Medicine, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325000, Zhejiang Province, China
- Key Laboratory of Orthopaedics of Zhejiang Province, Wenzhou, 325000, Zhejiang Province, China
- The Second School of Medicine, Wenzhou Medical University, Wenzhou, 325000, Zhejiang Province, China
| | - Zhongke Lin
- Department of Orthopaedics, Wenzhou Key Laboratory of Perinatal Medicine, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325000, Zhejiang Province, China.
- Key Laboratory of Orthopaedics of Zhejiang Province, Wenzhou, 325000, Zhejiang Province, China.
- The Second School of Medicine, Wenzhou Medical University, Wenzhou, 325000, Zhejiang Province, China.
| |
Collapse
|
29
|
Jiang H, Yu J, Yan Z, Lin Z, Lin M, Mao Y, Hong Z, Lin J, Xue X, Pan X. Pharmacological activation of the Nrf2 pathway by Taxifolin remodels articular cartilage microenvironment for the therapy of Osteoarthritis. Int Immunopharmacol 2023; 122:110587. [PMID: 37399606 DOI: 10.1016/j.intimp.2023.110587] [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: 05/25/2023] [Revised: 06/17/2023] [Accepted: 06/29/2023] [Indexed: 07/05/2023]
Abstract
BACKGROUND Osteoarthritis (OA) is a widely prevalent degenerative disease marked by extracellular matrix (ECM) degradation, inflammation, and apoptosis. Taxifolin (TAX) is a natural antioxidant possessing various pharmacological benefits, such as combating inflammation, oxidative stress, apoptosis, and serves as a potential chemopreventive agent by regulating genes through an antioxidant response element (ARE)-dependent mechanism. Currently, no studies have investigated the therapeutic impact and precise mechanism of TAX on OA. PURPOSE The aim of this study is to examine the potential role and mechanism of TAX in reshaping the cartilage microenvironment, thereby offering a stronger theoretical foundation for pharmacologically activating the Nrf2 pathway to manage OA. STUDY DESIGN AND METHODS The pharmacological effects of TAX were examined in chondrocytes through in vitro studies and in a destabilization of the medial meniscus (DMM) rat model for in vivo analysis. RESULTS TAX suppresses IL-1β triggered secretion of inflammatory agents, chondrocyte apoptosis, and ECM degradation, contributing to the remodeling of the cartilage microenvironment. In vivo experiment results demonstrated that TAX counteracted cartilage degeneration induced by DMM in rats. Mechanistic investigations revealed that TAX hinders OA development by reducing NF-κB activation and ROS production through the activation of the Nrf2/HO-1 axis. CONCLUSION TAX reshapes the articular cartilage microenvironment by suppressing inflammation, mitigating apoptosis, and decreasing ECM degradation through the activation of the Nrf2 pathway. As a result, pharmacological activation of the Nrf2 pathway by TAX holds potential clinical significance in remodeling the joint microenvironment for OA treatment.
Collapse
Affiliation(s)
- Hongyi Jiang
- Department of Orthopedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, China; Key Laboratory of Orthopedics of Zhejiang Province, Wenzhou, Zhejiang Province, China; The Second Clinical School of Medicine, Wenzhou Medical University, Wenzhou, Zhejiang Province, China
| | - Jiachen Yu
- Department of Orthopedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, China; Key Laboratory of Orthopedics of Zhejiang Province, Wenzhou, Zhejiang Province, China; The Second Clinical School of Medicine, Wenzhou Medical University, Wenzhou, Zhejiang Province, China
| | - Zijian Yan
- Department of Orthopedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, China; Key Laboratory of Orthopedics of Zhejiang Province, Wenzhou, Zhejiang Province, China; The Second Clinical School of Medicine, Wenzhou Medical University, Wenzhou, Zhejiang Province, China
| | - Zhongnan Lin
- Department of Orthopedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, China; Key Laboratory of Orthopedics of Zhejiang Province, Wenzhou, Zhejiang Province, China; The Second Clinical School of Medicine, Wenzhou Medical University, Wenzhou, Zhejiang Province, China
| | - Minghao Lin
- Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, Zhejiang, China
| | - Yiwen Mao
- Department of Orthopedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, China; Key Laboratory of Orthopedics of Zhejiang Province, Wenzhou, Zhejiang Province, China; The Second Clinical School of Medicine, Wenzhou Medical University, Wenzhou, Zhejiang Province, China
| | - Zechao Hong
- Department of Orthopedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, China; Key Laboratory of Orthopedics of Zhejiang Province, Wenzhou, Zhejiang Province, China; The Second Clinical School of Medicine, Wenzhou Medical University, Wenzhou, Zhejiang Province, China
| | - Jian Lin
- Department of Orthopedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, China; Key Laboratory of Orthopedics of Zhejiang Province, Wenzhou, Zhejiang Province, China; The Second Clinical School of Medicine, Wenzhou Medical University, Wenzhou, Zhejiang Province, China.
| | - Xinghe Xue
- Department of Orthopedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, China; Key Laboratory of Orthopedics of Zhejiang Province, Wenzhou, Zhejiang Province, China; The Second Clinical School of Medicine, Wenzhou Medical University, Wenzhou, Zhejiang Province, China.
| | - Xiaoyun Pan
- Department of Orthopedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, China; Key Laboratory of Orthopedics of Zhejiang Province, Wenzhou, Zhejiang Province, China; The Second Clinical School of Medicine, Wenzhou Medical University, Wenzhou, Zhejiang Province, China.
| |
Collapse
|
30
|
Restivo I, Basilicata MG, Giardina IC, Massaro A, Pepe G, Salviati E, Pecoraro C, Carbone D, Cascioferro S, Parrino B, Diana P, Ostacolo C, Campiglia P, Attanzio A, D’Anneo A, Pojero F, Allegra M, Tesoriere L. A Combination of Polymethoxyflavones from Citrus sinensis and Prenylflavonoids from Humulus lupulus Counteracts IL-1β-Induced Differentiated Caco-2 Cells Dysfunction via a Modulation of NF-κB/Nrf2 Activation. Antioxidants (Basel) 2023; 12:1621. [PMID: 37627616 PMCID: PMC10451557 DOI: 10.3390/antiox12081621] [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: 07/19/2023] [Revised: 08/10/2023] [Accepted: 08/15/2023] [Indexed: 08/27/2023] Open
Abstract
We here investigated the anti-inflammatory activity of a polymethoxylated flavone-containing fraction (PMFF) from Citrus sinensis and of a prenylflavonoid-containing one (PFF) from Humulus lupulus, either alone or in combination (MIX). To this end, an in vitro model of inflammatory bowel disease (IBD), consisting of differentiated, interleukin (IL)-1β-stimulated Caco-2 cells, was employed. We demonstrated that non-cytotoxic concentrations of either PMFF or PFF or MIX reduced nitric oxide (NO) production while PFF and MIX, but not PMFF, also inhibited prostaglandin E2 release. Coherently, MIX suppressed both inducible NO synthase and cyclooxygenase-2 over-expression besides NF-κB activation. Moreover, MIX increased nuclear factor erythroid 2-related factor 2 (Nrf2) activation, heme oxygenase-1 expression, restoring GSH and reactive oxygen and nitrogen species (RONs) levels. Remarkably, these effects with MIX were stronger than those produced by PMFF or PFF alone. Noteworthy, nobiletin (NOB) and xanthohumol (XTM), two of the most represented phytochemicals in PMFF and PFF, respectively, synergistically inhibited RONs production. Overall, our results demonstrate that MIX enhances the anti-inflammatory and anti-oxidative effects of the individual fractions in a model of IBD, via a mechanism involving modulation of NF-κB and Nrf2 signalling. Synergistic interactions between NOB and XTM emerge as a relevant aspect underlying this evidence.
Collapse
Affiliation(s)
- Ignazio Restivo
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies, University of Palermo, Via Archirafi 28, 90123 Palermo, Italy; (I.R.); (I.C.G.); (A.M.); (A.A.); (A.D.); (F.P.); (L.T.)
| | | | - Ilenia Concetta Giardina
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies, University of Palermo, Via Archirafi 28, 90123 Palermo, Italy; (I.R.); (I.C.G.); (A.M.); (A.A.); (A.D.); (F.P.); (L.T.)
| | - Alessandro Massaro
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies, University of Palermo, Via Archirafi 28, 90123 Palermo, Italy; (I.R.); (I.C.G.); (A.M.); (A.A.); (A.D.); (F.P.); (L.T.)
| | - Giacomo Pepe
- Department of Pharmacy, University of Salerno, 84084 Fisciano, Italy; (M.G.B.); (E.S.); (C.O.); (P.C.)
| | - Emanuela Salviati
- Department of Pharmacy, University of Salerno, 84084 Fisciano, Italy; (M.G.B.); (E.S.); (C.O.); (P.C.)
| | - Camilla Pecoraro
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies, University of Palermo, Via Archirafi 32, 90123 Palermo, Italy; (C.P.); (D.C.); (S.C.); (B.P.); (P.D.)
| | - Daniela Carbone
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies, University of Palermo, Via Archirafi 32, 90123 Palermo, Italy; (C.P.); (D.C.); (S.C.); (B.P.); (P.D.)
| | - Stella Cascioferro
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies, University of Palermo, Via Archirafi 32, 90123 Palermo, Italy; (C.P.); (D.C.); (S.C.); (B.P.); (P.D.)
| | - Barbara Parrino
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies, University of Palermo, Via Archirafi 32, 90123 Palermo, Italy; (C.P.); (D.C.); (S.C.); (B.P.); (P.D.)
| | - Patrizia Diana
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies, University of Palermo, Via Archirafi 32, 90123 Palermo, Italy; (C.P.); (D.C.); (S.C.); (B.P.); (P.D.)
| | - Carmine Ostacolo
- Department of Pharmacy, University of Salerno, 84084 Fisciano, Italy; (M.G.B.); (E.S.); (C.O.); (P.C.)
| | - Pietro Campiglia
- Department of Pharmacy, University of Salerno, 84084 Fisciano, Italy; (M.G.B.); (E.S.); (C.O.); (P.C.)
| | - Alessandro Attanzio
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies, University of Palermo, Via Archirafi 28, 90123 Palermo, Italy; (I.R.); (I.C.G.); (A.M.); (A.A.); (A.D.); (F.P.); (L.T.)
| | - Antonella D’Anneo
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies, University of Palermo, Via Archirafi 28, 90123 Palermo, Italy; (I.R.); (I.C.G.); (A.M.); (A.A.); (A.D.); (F.P.); (L.T.)
| | - Fanny Pojero
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies, University of Palermo, Via Archirafi 28, 90123 Palermo, Italy; (I.R.); (I.C.G.); (A.M.); (A.A.); (A.D.); (F.P.); (L.T.)
| | - Mario Allegra
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies, University of Palermo, Via Archirafi 28, 90123 Palermo, Italy; (I.R.); (I.C.G.); (A.M.); (A.A.); (A.D.); (F.P.); (L.T.)
| | - Luisa Tesoriere
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies, University of Palermo, Via Archirafi 28, 90123 Palermo, Italy; (I.R.); (I.C.G.); (A.M.); (A.A.); (A.D.); (F.P.); (L.T.)
| |
Collapse
|
31
|
Wang J, Xiao S, Cai Q, Miao J, Li J. Antioxidant Capacity and Protective Effects on H 2O 2-Induced Oxidative Damage in PC12 Cells of the Active Fraction of Brassica rapa L. Foods 2023; 12:2075. [PMID: 37238893 PMCID: PMC10217163 DOI: 10.3390/foods12102075] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2023] [Revised: 05/13/2023] [Accepted: 05/19/2023] [Indexed: 05/28/2023] Open
Abstract
Brassica rapa L. (BR), a traditional biennial herb belonging to the Brassica species of Brassicaceae, has been widely used for functions of anti-inflammatory, antitumor, antioxidation, antiaging, and regulation of immunity. In this study, antioxidant activity and protective effects on H2O2-induced oxidative damage in PC12 cells of the active fractions of BR were investigated in vitro. Among all active fractions, the ethyl acetate fraction of ethanol extract from BR (BREE-Ea) showed the strongest antioxidant activity. Additionally, it was noted that BREE-Ea and n-butyl alcohol fraction of ethanol extract from BR (BREE-Ba) both have protective effects in oxidatively damaged PC12 cells, while BREE-Ea displayed the best protective effect in all determined experimental doses. Furthermore, flow cytometry (DCFH-DA staining) analysis indicated that BREE-Ea could reduce the H2O2-induced apoptosis in PC12 cells by reducing the production of intracellular reactive oxygen species (ROS) and increasing enzymatic activities of superoxide dismutase (SOD) and glutathione peroxidase (GSH-Px). Moreover, BREE-Ea could decrease the malondialdehyde (MDA) content and reduce the release of extracellular lactic dehydrogenase (LDH) from H2O2-induced PC12 cells. All these results demonstrate that BREE-Ea has a good antioxidant capacity and protective effect on PC12 cells against apoptosis induced by H2O2 and that it can be used as a good edible antioxidant to improve the body's endogenous antioxidant defense.
Collapse
Affiliation(s)
- Jin Wang
- College of Life Science and Technology, Xinjiang University, Urumqi 830000, China; (J.W.); (S.X.); (Q.C.)
| | - Shuang Xiao
- College of Life Science and Technology, Xinjiang University, Urumqi 830000, China; (J.W.); (S.X.); (Q.C.)
| | - Qi Cai
- College of Life Science and Technology, Xinjiang University, Urumqi 830000, China; (J.W.); (S.X.); (Q.C.)
| | - Jing Miao
- Pharmaceutical Institute, Xinjiang University, Urumqi 830000, China
- Xinjiang Key Laboratory of Biological Resources and Genetic Engineering, Xinjiang University, Urumqi 830000, China
| | - Jinyao Li
- College of Life Science and Technology, Xinjiang University, Urumqi 830000, China; (J.W.); (S.X.); (Q.C.)
- Pharmaceutical Institute, Xinjiang University, Urumqi 830000, China
- Xinjiang Key Laboratory of Biological Resources and Genetic Engineering, Xinjiang University, Urumqi 830000, China
| |
Collapse
|
32
|
Xie W, Qi S, Dou L, Wang L, Wang X, Bi R, Li N, Zhang Y. Achyranthoside D attenuates chondrocyte loss and inflammation in osteoarthritis via targeted regulation of Wnt3a. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2023; 111:154663. [PMID: 36657317 DOI: 10.1016/j.phymed.2023.154663] [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: 10/17/2022] [Revised: 12/28/2022] [Accepted: 01/09/2023] [Indexed: 06/17/2023]
Abstract
BACKGROUND Achyranthes bidentata Blume (A. bidentata) is a common Chinese herb used to treat osteoarthritis (OA). Achyranthoside D (Ach-D) is a glucuronide saponin isolated from A. bidentata. PURPOSE To assess the mechanisms of action of Ach-D and its effects on OA. METHODS The effects of Ach-D were evaluated in rats underwent anterior cruciate ligament transection (ACLT) with medial meniscectomy (MMx) and in interleukin (IL)-1β-induced chondrocytes. Histological changes in rat cartilage tissues were detected using Safranin O-Fast green and haematoxylin-eosin staining. Immunohistochemical staining, qRT-PCR, ELISA, immunoblotting, and immunofluorescence were conducted to examine cartilage degeneration-related and inflammation-related factor expression. CCK-8, LDH assay, and EdU staining were performed to detect chondrocyte death. RESULTS Ach-D dose-dependently reduced the Osteoarthritis Research Society International (OARSI) scores, alleviated cartilage injury, and decreased the serum concentrations of CTX-II and COMP in ACLT-MMx models. Ach-D increased the expression levels of collagen II and aggrecan and decreased the levels of cartilage degeneration-related proteins, ADAMTS-5, MMP13, and MMP3, in rat cartilage tissues. Additionally, nod-like receptor protein 3 (NLRP3)-related inflammation was reduced by Ach-D, as shown by the significantly inhibited expression levels of NLRP3, ASC, GSDMD, IL-6, TNF-α, IL-1β, and IL-18 in rat cartilage tissues. In primary rat chondrocytes, Ach-D protected against IL-1β-induced viability loss and LDH release. Wnt3a is the target protein of Ach-D. Mechanistically, Ach-D alleviated OA by inhibiting Wnt signalling. CONCLUSION ACH-D may reduce inflammation and cartilage degeneration by inhibiting the Wnt signalling pathway, thereby reducing OA.
Collapse
Affiliation(s)
- Wenpeng Xie
- Department of Orthopedics, Shandong University of Traditional Chinese Medicine Affiliated Hospital, Jinan, 250000, Shandong, PR China
| | - Shangfeng Qi
- Department of Orthopedics, Shandong University of Traditional Chinese Medicine Affiliated Hospital, Jinan, 250000, Shandong, PR China
| | - Luming Dou
- Bone traumatology department, Yantai Penglai Traditional Chinese Medicine Hospital, Yantai, 265600, Shandong, PR China
| | - Lei Wang
- Department of Orthopedics, Shandong University of Traditional Chinese Medicine Second Affiliated Hospital, Jinan, 250000, Shandong, PR China
| | - Xiangpeng Wang
- Department of Orthopedics, Shandong University of Traditional Chinese Medicine Affiliated Hospital, Jinan, 250000, Shandong, PR China
| | - Rongxiu Bi
- Department of Orthopedics, Shandong University of Traditional Chinese Medicine Affiliated Hospital, Jinan, 250000, Shandong, PR China
| | - Nianhu Li
- Department of Orthopedics, Shandong University of Traditional Chinese Medicine Affiliated Hospital, Jinan, 250000, Shandong, PR China.
| | - Yongkui Zhang
- Department of Orthopedics, Shandong University of Traditional Chinese Medicine Affiliated Hospital, Jinan, 250000, Shandong, PR China.
| |
Collapse
|
33
|
Teng L, Shen Y, Qu Y, Yang L, Yang Y, Jian X, Fan S, Zhang L, Fu Q. Cyasterone inhibits IL-1β-mediated apoptosis and inflammation via the NF-κB and MAPK signaling pathways in rat chondrocytes and ameliorates osteoarthritisin vivo. Chin J Nat Med 2023; 21:99-112. [PMID: 36871986 DOI: 10.1016/s1875-5364(23)60388-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2022] [Indexed: 03/07/2023]
Abstract
Osteoarthritis is a prevalent global joint disease, which is characterized by inflammatory reaction and cartilage degradation. Cyasterone, a sterone derived from the roots of Cyathula officinalis Kuan, exerts protective effect against several inflammation-related diseases. However, its effect on osteoarthritis remains unclear. The current study was designed to investigate the potential anti-osteoarthritis activity of cyasterone. Primary chondrocytes isolated from rats induced by interleukin (IL)-1β and a rat model stimulated by monosodium iodoacetate (MIA) were used for in vitro and in vivo experiments, respectively. The results of in vitro experiments showed that cyasterone apparently counteracted chondrocyte apoptosis, increased the expression of collagen II and aggrecan, and restrained the production of the inflammatory factors inducible nitric oxide synthase (iNOS), cyclooxygenase-2 (COX-2), a disintegrin and metalloproteinase with thrombospondin motifs-5 (ADAMTS-5), metalloproteinase-3 (MMP-3), and metalloproteinase-13 (MMP-13) induced by IL-1β in chondrocytes. Furthermore, cyasterone ameliorated the inflammation and degenerative progression of osteoarthritis potentially by regulating the nuclear factor kappa B (NF-κB) and mitogen-activated protein kinase (MAPK) pathways. For in vivo experiments, cyasterone significantly alleviated the inflammatory response and cartilage destruction of rats induced by monosodium iodoacetate, where dexamethasone was used as the positive control. Overall, this study laid a theoretical foundation for developing cyasterone as an effective agent for the alleviation of osteoarthritis.
Collapse
Affiliation(s)
- Li Teng
- Sichuan Industrial Institute of Antibiotics, School of Pharmacy, Chengdu University, Chengdu 610106, China; School of Food and Biological Engineering, Chengdu University, Chengdu 610106, China
| | - Yue Shen
- Sichuan Industrial Institute of Antibiotics, School of Pharmacy, Chengdu University, Chengdu 610106, China
| | - Yuhan Qu
- Sichuan Industrial Institute of Antibiotics, School of Pharmacy, Chengdu University, Chengdu 610106, China; School of Food and Biological Engineering, Chengdu University, Chengdu 610106, China
| | - Longfei Yang
- Sichuan Industrial Institute of Antibiotics, School of Pharmacy, Chengdu University, Chengdu 610106, China
| | - Yuting Yang
- Sichuan Industrial Institute of Antibiotics, School of Pharmacy, Chengdu University, Chengdu 610106, China
| | - Xi Jian
- Sichuan Industrial Institute of Antibiotics, School of Pharmacy, Chengdu University, Chengdu 610106, China
| | - Shengli Fan
- Sichuan Industrial Institute of Antibiotics, School of Pharmacy, Chengdu University, Chengdu 610106, China
| | - Lele Zhang
- School of Basic Medical Sciences, Chengdu University, Chengdu 610106, China.
| | - Qiang Fu
- Sichuan Industrial Institute of Antibiotics, School of Pharmacy, Chengdu University, Chengdu 610106, China.
| |
Collapse
|
34
|
Gambari L, Cellamare A, Grassi F, Grigolo B, Panciera A, Ruffilli A, Faldini C, Desando G. Overview of Anti-Inflammatory and Anti-Nociceptive Effects of Polyphenols to Halt Osteoarthritis: From Preclinical Studies to New Clinical Insights. Int J Mol Sci 2022; 23:ijms232415861. [PMID: 36555503 PMCID: PMC9779856 DOI: 10.3390/ijms232415861] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Revised: 12/10/2022] [Accepted: 12/11/2022] [Indexed: 12/15/2022] Open
Abstract
Knee osteoarthritis (OA) is one of the most multifactorial joint disorders in adults. It is characterized by degenerative and inflammatory processes that are responsible for joint destruction, pain and stiffness. Despite therapeutic advances, the search for alternative strategies to target inflammation and pain is still very challenging. In this regard, there is a growing body of evidence for the role of several bioactive dietary molecules (BDMs) in targeting inflammation and pain, with promising clinical results. BDMs may be valuable non-pharmaceutical solutions to treat and prevent the evolution of early OA to more severe phenotypes, overcoming the side effects of anti-inflammatory drugs. Among BDMs, polyphenols (PPs) are widely studied due to their abundance in several plants, together with their benefits in halting inflammation and pain. Despite their biological relevance, there are still many questionable aspects (biosafety, bioavailability, etc.) that hinder their clinical application. This review highlights the mechanisms of action and biological targets modulated by PPs, summarizes the data on their anti-inflammatory and anti-nociceptive effects in different preclinical in vitro and in vivo models of OA and underlines the gaps in the knowledge. Furthermore, this work reports the preliminary promising results of clinical studies on OA patients treated with PPs and discusses new perspectives to accelerate the translation of PPs treatment into the clinics.
Collapse
Affiliation(s)
- Laura Gambari
- Laboratorio RAMSES, IRCCS Istituto Ortopedico Rizzoli, via di Barbiano 1/10, 40136 Bologna, Italy
| | - Antonella Cellamare
- Laboratorio RAMSES, IRCCS Istituto Ortopedico Rizzoli, via di Barbiano 1/10, 40136 Bologna, Italy
| | - Francesco Grassi
- Laboratorio RAMSES, IRCCS Istituto Ortopedico Rizzoli, via di Barbiano 1/10, 40136 Bologna, Italy
| | - Brunella Grigolo
- Laboratorio RAMSES, IRCCS Istituto Ortopedico Rizzoli, via di Barbiano 1/10, 40136 Bologna, Italy
| | - Alessandro Panciera
- 1st Orthopedic and Traumatology Clinic, IRCCS Istituto Ortopedico Rizzoli, via G.C. Pupilli 1, 40136 Bologna, Italy
| | - Alberto Ruffilli
- 1st Orthopedic and Traumatology Clinic, IRCCS Istituto Ortopedico Rizzoli, via G.C. Pupilli 1, 40136 Bologna, Italy
| | - Cesare Faldini
- 1st Orthopedic and Traumatology Clinic, IRCCS Istituto Ortopedico Rizzoli, via G.C. Pupilli 1, 40136 Bologna, Italy
| | - Giovanna Desando
- Laboratorio RAMSES, IRCCS Istituto Ortopedico Rizzoli, via di Barbiano 1/10, 40136 Bologna, Italy
- Correspondence: ; Tel.: +39-0516366803
| |
Collapse
|
35
|
Xu X, Li N, Wu Y, Yan K, Mi Y, Yi N, Tan X, Kuang G, Lu M. Zhuifeng tougu capsules inhibit the TLR4/MyD88/NF-κB signaling pathway and alleviate knee osteoarthritis: In vitro and in vivo experiments. Front Pharmacol 2022; 13:951860. [PMID: 36188596 PMCID: PMC9521277 DOI: 10.3389/fphar.2022.951860] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Accepted: 08/22/2022] [Indexed: 01/17/2023] Open
Abstract
Background: Knee osteoarthritis (KOA), a chronic degenerative disease, is mainly characterized by destruction of articular cartilage and inflammatory reactions. At present, there is a lack of economical and effective clinical treatment. Zhuifeng Tougu (ZFTG) capsules have been clinically approved for treatment of OA as they relieve joint pain and inflammatory manifestations. However, the mechanism of ZFTG in KOA remains unknown. Purpose: This study aimed to investigate the effect of ZFTG on the TLR4/MyD88/NF-κB signaling pathway and its therapeutic effect on rabbits with KOA. Study design: In vivo, we established a rabbit KOA model using the modified Videman method. In vitro, we treated chondrocytes with IL-1β to induce a pro-inflammatory phenotype and then intervened with different concentrations of ZFTG. Levels of IL-1β, IL-6, TNF-α, and IFN-γ were assessed with histological observations and ELISA data. The effect of ZFTG on the viability of chondrocytes was detected using a Cell Counting Kit-8 and flow cytometry. The protein and mRNA expressions of TLR2, TLR4, MyD88, and NF-κB were detected using Western blot and RT-qPCR and immunofluorescence observation of NF-κB p65 protein expression, respectively, to investigate the mechanism of ZFTG in inhibiting inflammatory injury of rabbit articular chondrocytes and alleviating cartilage degeneration. Results: The TLR4/MyD88/NF-κB signaling pathway in rabbits with KOA was inhibited, and the levels of IL-1β, IL-6, TNF-α, and IFN-γ in blood and cell were significantly downregulated, consistent with histological results. Both the protein and mRNA expressions of TLR2, TLR4, MyD88, NF-κB, and NF-κB p65 proteins in that nucleus decreased in the ZFTG groups. Moreover, ZFTG promotes the survival of chondrocytes and inhibits the apoptosis of inflammatory chondrocytes. Conclusion: ZFTG alleviates the degeneration of rabbit knee joint cartilage, inhibits the apoptosis of inflammatory chondrocytes, and promotes the survival of chondrocytes. The underlying mechanism may be inhibition of the TLR4/MyD88/NF-kB signaling pathway and secretion of inflammatory factors.
Collapse
Affiliation(s)
- Xiaotong Xu
- Department of Orthopedic Surgery, The First Hospital of Hunan University of Chinese Medicine, Changsha, Hunan, China
- Graduate School, Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Naping Li
- Department of Orthopedic Surgery, The First Hospital of Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Yongrong Wu
- School of Traditional Chinese Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Ke Yan
- Department of Orthopedic Surgery, The First Hospital of Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Yilin Mi
- Graduate School, Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Nanxing Yi
- Graduate School, Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Xuyi Tan
- Department of Orthopedic Surgery, Affiliated Hospital of Hunan Academy of Chinese Medical Science, Changsha, Hunan, China
| | - Gaoyan Kuang
- Department of Orthopedic Surgery, The First Hospital of Hunan University of Chinese Medicine, Changsha, Hunan, China
- Postdoctoral Research Workstation, Hinye Pharmaceutical Co., Ltd., Changsha, Hunan, China
| | - Min Lu
- Department of Orthopedic Surgery, The First Hospital of Hunan University of Chinese Medicine, Changsha, Hunan, China
| |
Collapse
|
36
|
Wu X, Zhao K, Fang X, Lu F, Cheng P, Song X, Zhang W, Yao C, Zhu J, Chen H. Saikosaponin D Inhibited IL-1β Induced ATDC 5 Chondrocytes Apoptosis In Vitro and Delayed Articular Cartilage Degeneration in OA Model Mice In Vivo. Front Pharmacol 2022; 13:845959. [PMID: 35370642 PMCID: PMC8975252 DOI: 10.3389/fphar.2022.845959] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Accepted: 02/24/2022] [Indexed: 11/13/2022] Open
Abstract
Osteoarthritis (OA) is the most common joint disease in the elderly, characterized by cartilage degradation and proliferation of subchondral bone. The pathogenesis of OA involves a variety of inflammatory mediators, including nitric oxide (NO), prostaglandin E2 (PGE2), tumor necrosis factor (TNF)-α, and interleukin (IL)-1β. From the molecular mechanism, the nuclear factor-erythroid 2-related factor (Nrf2)/heme oxygenase-1 (HO-1) pathway and the expression of ROS regulated the production of the above inflammatory mediators. Saikosaponin D (SSD), which is an active ingredient isolated from Bupleurum, has various biological functions. In this study, IL-1β was used as a pro-inflammatory factor to create an in vitro OA model. According to the results of high-density culture, qPCR, ROS measurement, Western blot, and immunofluorescence, SSD activated the Nrf2/HO-1/ROS axis, inhibited the production of inflammatory mediators, and protected against ECM destruction. The DMM mouse model was used as a model of OA in mice. From the results of safranin O/fast green staining, hematoxylin–eosin staining, tartrate-resistant acid phosphatase (TRAP) staining, and OARSI scores, SSD protected against the mice knee articular cartilage degeneration and reduced the number of osteoclasts in the subchondral bone. Experimental results found that SSD suppressed IL-1β–induced differentiated ATDC 5 chondrocytes apoptosis via the Nrf2/HO-1/ROS axis in vitro. SSD delayed the progression of OA in DMMs model mice in vivo. Therefore, SSD has the potential to become a drug for clinical treatment of OA.
Collapse
Affiliation(s)
- Xinhui Wu
- Department of Orthopedics, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Linhai, China
- Wenzhou Medical University, Wenzhou, China
| | - Kangxian Zhao
- Department of Orthopedics, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Linhai, China
- Wenzhou Medical University, Wenzhou, China
| | - Xiaoxin Fang
- Zhejiang University School of Medicine, Hangzhou, China
- Taizhou Hospital of Zhejiang Province, Zhejiang University, Linhai, China
| | - Feng Lu
- Zhejiang University School of Medicine, Hangzhou, China
- Taizhou Hospital of Zhejiang Province, Zhejiang University, Linhai, China
| | - Pu Cheng
- Department of Orthopedics, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Linhai, China
- Wenzhou Medical University, Wenzhou, China
| | - Xiaoting Song
- Department of Orthopedics, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Linhai, China
- Wenzhou Medical University, Wenzhou, China
| | - Weikang Zhang
- Department of Orthopedics, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Linhai, China
- Wenzhou Medical University, Wenzhou, China
| | - Can Yao
- Department of Orthopedics, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Linhai, China
| | - Jiling Zhu
- Wenzhou Medical University, Wenzhou, China
| | - Haixiao Chen
- Department of Orthopedics, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Linhai, China
- Wenzhou Medical University, Wenzhou, China
- Taizhou Hospital of Zhejiang Province, Zhejiang University, Linhai, China
- *Correspondence: Haixiao Chen,
| |
Collapse
|