1
|
Kung C, Lin M, Chen B. Preparation of Collagen Peptide Nanoliposomes From Sturgeon Fish Cartilage and Explore Their Anti‐Osteoarthritis Effects in Rats. FOOD FRONTIERS 2025; 6:727-749. [DOI: 10.1002/fft2.539] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/14/2025] Open
Abstract
ABSTRACTThis study aims to extract pepsin soluble collagen (PSC) from sturgeon cartilage, hydrolyze to sturgeon cartilage collagen peptides (SCCP), and prepare SCCP nanoliposomes to explore the treatment effects of osteoarthritis (OA) in rats. PSC was extracted using 0.5 M acetic acid and pepsin (10%) and enzymatically hydrolyzed with 4.5% alcalase plus 4.5% flavourzyme to obtain SCCP. Amino acid analysis revealed the presence of glycine, proline, and hydroxyproline in high amounts, while SDS‐PAGE showed that the PSC belonged to type II collagen with molecular weight (MW) of SCCP being <2 kDa and MALDI‐TOF‐MS indicated the MW distribution to range from 302.594 to 683.050 Da with the peptide fragments <500 Da accounting for 89.71%. SCCP nanoliposomes composed of phosphatidylcholine, fatty acid sucrose ester, glycerol, and deionized water were prepared with size at 34.58 nm, polydispersity index at 0.19, zeta potential at ‐54.53 mV, and encapsulation efficiency at 88.14%. Tube feeding of SCCP/SCCP nanoliposomes into OA rats alleviated pain responses by joint damage through reduction in hind limb weight‐bearing difference, knee joint width difference, and levels of serum biomarkers including CTX‐II, TGF‐β1, PIICP, and COMP. Histopathologic images demonstrated the mitigation of joint damage symptoms in the tissue by reducing cartilage joint damage, inhibiting chondrocyte apoptosis, promoting chondrocyte regeneration, and reducing synovitis. Collectively, the high dose of SCCP nanoliposomes was the most effective in alleviating OA possessing a great potential to be developed into a health food or botanic drug for the treatment of joint‐related disease.
Collapse
Affiliation(s)
- Chia‐Chi Kung
- School of Medicine Fu Jen Catholic University Taipei
- Department of Anesthesia Fu Jen Catholic University Hospital Taipei
| | - Mei‐Zhen Lin
- Department of Food Science Fu Jen Catholic University Taipei
| | - Bing‐Huei Chen
- Department of Food Science Fu Jen Catholic University Taipei
- Department of Nutrition China Medical University Taichung
| |
Collapse
|
2
|
Wang W, Liu X, Nan H, Li H, Yan L. Specific gut microbiota and serum metabolite changes in patients with osteoarthritis. Front Cell Dev Biol 2025; 13:1543510. [PMID: 40027098 PMCID: PMC11868077 DOI: 10.3389/fcell.2025.1543510] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2024] [Accepted: 01/20/2025] [Indexed: 03/05/2025] Open
Abstract
Introduction Recent research indicated a strong link between the gut microbiota and osteoarthritis. However, the complex interplay between the gut microbiota, serum metabolites, and the progression of osteoarthritis in affected individuals remains largely unexplored. This study aimed to investigate the characteristics of the gut microbiota and serum metabolites in patients with osteoarthritis. Methods Participants with either healthy knees or osteoarthritis were enrolled and categorized into healthy control (HC) and osteoarthritis (OA) groups. Fecal and blood samples were collected for 16S rRNA gene sequencing, metabolomic analysis via liquid chromatography-mass spectrometry (LC-MS), and integrated evaluation. Results The results showed no significant variation in gut microbiota richness and diversity between the two groups. However, the abundance of Bacteroides plebeius and Faecalibacterium prausnitzii was reduced in the OA group, both of which are known for their potential as next-generation probiotics for human health. Metabolomic analysis indicated that serum metabolites, including pyrogallol and 3-hydroxybutyrate (3HB), were significantly lower in the OA group. These metabolites are known to positively impact osteoarthritis progression and other diseases and demonstrated good diagnostic performance for distinguishing osteoarthritis patients from healthy controls. Correlation analysis revealed a positive correlation between Bacteroides plebeius and Faecalibacterium prausnitzii and between pyrogallol and 3HB. Discussion This study highlighted specific gut microbiota and serum metabolite profiles in osteoarthritis patients, suggesting that the specific changes in bacteria and derived metabolites are closely tied to osteoarthritis progression. This underscores the potential of gut microbiota and serum metabolites as modifiable elements and therapeutic targets for osteoarthritis prevention.
Collapse
Affiliation(s)
- Wendong Wang
- Department of Engineering Mechanics, Dalian University of Technology, Dalian, China
| | - Xincheng Liu
- Department of Articular Orthopaedics, The Second People’s Hospital of Dalian, Dalian, China
| | - Hao Nan
- Department of Articular Orthopaedics, The First People’s Hospital of Changzhou, The Third Affiliated Hospital of Soochow University, Changzhou, China
| | - Huan Li
- Department of Articular Orthopaedics, The First People’s Hospital of Changzhou, The Third Affiliated Hospital of Soochow University, Changzhou, China
| | - Litao Yan
- Department of Articular Orthopaedics, The First People’s Hospital of Changzhou, The Third Affiliated Hospital of Soochow University, Changzhou, China
| |
Collapse
|
3
|
Zhao T, Wang X, Li Z, Qin D. Yiqi Yangxue formula inhibits cartilage degeneration in knee osteoarthritis by regulating LncRNA-UFC1/miR-34a/MMP-13 axis. JOURNAL OF ETHNOPHARMACOLOGY 2025; 337:118930. [PMID: 39393561 DOI: 10.1016/j.jep.2024.118930] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Revised: 10/08/2024] [Accepted: 10/09/2024] [Indexed: 10/13/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Knee osteoarthritis (KOA) is a prevalent and disabling clinical condition affecting joint structures worldwide. The Yiqi Yangxue formula (YQYXF) is frequently prescribed in clinical settings for the treatment of KOA. Existing research has primarily focused on alterations in drug metabolism, with limited investigation into the epigenetic effects of YQYXF, particularly in relation to non-coding RNA. AIM OF THE STUDY Exploring the effects of YQYXF on critical factors of long chain non-coding RNA UFC1/miR-34a/matrix metalloproteinase-13 (MMP-13) axis and their interrelationships. METHODS UHPLC-QE-MS technology was used to identify the YQYXF ingredients in rat serum. KEGG and GO analysis were performed on the targets of blood components acting on KOA using a database. Simultaneously, a protein interaction network was constructed using target proteins and metabolites to identify the core components and key pathways of YQYXF. The KOA rat model was established using an improved Hulth method. SPF SD rats were randomly divided into normal group, sham surgery group, model group, celecoxib capsules group (18 mg/kg), YQYXF low, medium and high dose groups (4.6 g/kg, 9.2 g/kg, 18.4 g/kg). Observe the synovial and cartilage tissues of rats using pathological methods. RT-PCR was used to detect the levels of UFC1, miR-34a, and MMP-13 in cartilage. Immunohistochemistry was used to detect the levels of MMP-13 and ADAMTS-5 in cartilage. ELISA method was used to detect the levels of MMP-13 and ADAMTS-5 in serum. In addition, we further validated the regulation of crucial factor expression levels of UFC1/miR-34a/MMP-13 axis in rat chondrocytes and degenerative chondrocytes of KOA patients by YQYXF, providing a basis for its treatment of KOA. RESULTS The compounds that YQYXF enters the bloodstream mainly contain flavonoids and phenylpropanoid compounds. The core components that act on OA include quercetin, fisetin, and demethylweldelolactone. The main target pathways are the IL-17 signaling pathway, lipid and atherosclerosis, cellular sensitivity, inflammatory mediator regulation of TRP channels, TNF signaling pathway, relaxin signaling pathway and C-type lectin receptor signaling pathway. YQYXF inhibited the expression of miR-34a and MMP-13 mRNA, and reduced the protein levels of MMP-13 and ADAMTS-5. In vitro studies have confirmed that 20% YQYXF serum promoted UFC1 and reduce miR-34a levels. In addition, miR-34a in sh-UFC1+10% YQYXF serum and sh-UFC1+20% YQYXF serum groups significantly decreased compared to the sh-UFC1 group. CONCLUSION The anti-KOA cartilage degeneration effect of YQYXF might be related to inhibiting cell apoptosis and promoting cell proliferation, which regulated the lncRNA-UFC1/miR-34a/MMP-13 axis.
Collapse
MESH Headings
- Animals
- Matrix Metalloproteinase 13/metabolism
- Matrix Metalloproteinase 13/genetics
- MicroRNAs/metabolism
- MicroRNAs/genetics
- Drugs, Chinese Herbal/pharmacology
- Rats, Sprague-Dawley
- RNA, Long Noncoding/genetics
- RNA, Long Noncoding/metabolism
- Osteoarthritis, Knee/drug therapy
- Osteoarthritis, Knee/pathology
- Osteoarthritis, Knee/metabolism
- Rats
- Male
- Chondrocytes/drug effects
- Chondrocytes/metabolism
- Cartilage, Articular/drug effects
- Cartilage, Articular/pathology
- Cartilage, Articular/metabolism
- Disease Models, Animal
Collapse
Affiliation(s)
- Ting Zhao
- The Second Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, 310000, China; School of Basic Medical Sciences, Yunnan University of Chinese Medicine, Kunming, 650500, China
| | - Xinchang Wang
- The Second Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, 310000, China.
| | - Zhaofu Li
- School of Basic Medical Sciences, Yunnan University of Chinese Medicine, Kunming, 650500, China.
| | - Dongdong Qin
- The Second Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, 310000, China; School of Basic Medical Sciences, Yunnan University of Chinese Medicine, Kunming, 650500, China.
| |
Collapse
|
4
|
Hridayanka KSN, Duttaroy AK, Basak S. Bioactive Compounds and Their Chondroprotective Effects for Osteoarthritis Amelioration: A Focus on Nanotherapeutic Strategies, Epigenetic Modifications, and Gut Microbiota. Nutrients 2024; 16:3587. [PMID: 39519419 PMCID: PMC11547880 DOI: 10.3390/nu16213587] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2024] [Revised: 10/20/2024] [Accepted: 10/21/2024] [Indexed: 11/16/2024] Open
Abstract
In degenerative joint disease like osteoarthritis (OA), bioactive compounds like resveratrol, epigallocatechin gallate, curcumin, and other polyphenols often target various signalling pathways, including NFκB, TGFβ, and Wnt/β-catenin by executing epigenetic-modifying activities. Epigenetic modulation can target genes of disease pathophysiology via histone modification, promoter DNA methylation, and non-coding RNA expression, some of which are directly involved in OA but have been less explored. OA patients often seek options that can improve the quality of their life in addition to existing treatment with nonsteroidal anti-inflammatory drugs (NSAIDs). Although bioactive and natural compounds exhibit therapeutic potential against OA, several disadvantages loom, like insolubility and poor bioavailability. Nanoformulated bioactive compounds promise a better way to alleviate OA since they also control systemic events, including metabolic, immunological, and inflammatory responses, by modulating host gut microbiota that can regulate OA pathogenesis. Recent data suggest gut dysbiosis in OA. However, limited evidence is available on the role of bioactive compounds as epigenetic and gut modulators in ameliorating OA. Moreover, it is not known whether the effects of polyphenolic bioactive compounds on gut microbial response are mediated by epigenetic modulatory activities in OA. This narrative review highlights the nanotherapeutic strategies utilizing bioactive compounds, reporting their effects on chondrocyte growth, metabolism, and epigenetic modifications in osteoarthritis amelioration.
Collapse
Affiliation(s)
- Kota Sri Naga Hridayanka
- Molecular Biology Division, National Institute of Nutrition, Indian Council of Medical Research, Hyderabad 500007, India;
| | - Asim K. Duttaroy
- Department of Nutrition, Institute of Basic Medical Sciences, Faculty of Medicine, University of Oslo, 0317 Oslo, Norway;
| | - Sanjay Basak
- Molecular Biology Division, National Institute of Nutrition, Indian Council of Medical Research, Hyderabad 500007, India;
| |
Collapse
|
5
|
Zhou J, Li X, Han Z, Qian Y, Bai L, Han Q, Gao M, Xue Y, Geng D, Yang X, Hao Y. Acetyl-11-keto-β-boswellic acid restrains the progression of synovitis in osteoarthritis via the Nrf2/HO-1 pathway. Acta Biochim Biophys Sin (Shanghai) 2024; 56:1644-1658. [PMID: 38982914 PMCID: PMC11659770 DOI: 10.3724/abbs.2024102] [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: 01/15/2024] [Accepted: 04/01/2024] [Indexed: 07/11/2024] Open
Abstract
Synovial inflammation plays a key role in osteoarthritis (OA) pathogenesis. Fibroblast-like synoviocytes (FLSs) represent a distinct cell subpopulation within the synovium, and their unique phenotypic alterations are considered significant contributors to inflammation and fibrotic responses. The underlying mechanism by which acetyl-11-keto-β-boswellic acid (AKBA) modulates FLS activation remains unclear. This study aims to assess the beneficial effects of AKBA through both in vitro and in vivo investigations. Network pharmacology evaluation is used to identify potential targets of AKBA in OA. We evaluate the effects of AKBA on FLSs activation in vitro and the regulatory role of AKBA on the Nrf2/HO-1 signaling pathway. ML385 (an Nrf2 inhibitor) is used to verify the binding of AKBA to its target in FLSs. We validate the in vivo efficacy of AKBA in alleviating OA using anterior cruciate ligament transection and destabilization of the medial meniscus (ACLT+DMM) in a rat model. Network pharmacological analysis reveals the potential effect of AKBA on OA. AKBA effectively attenuates lipopolysaccharide (LPS)-induced abnormal migration and invasion and the production of inflammatory mediators, matrix metalloproteinases (MMPs), and reactive oxygen species (ROS) in FLSs, contributing to the restoration of the synovial microenvironment. After treatment with ML385, the effect of AKBA on FLSs is reversed. In vivo studies demonstrate that AKBA mitigates synovial inflammation and fibrotic responses induced by ACLT+DMM in rats via activation of the Nrf2/HO-1 axis. AKBA exhibits theoretical potential for alleviating OA progression through the Nrf2/HO-1 pathway and represents a viable therapeutic candidate for this patient population.
Collapse
Affiliation(s)
- Jing Zhou
- Orthopedics and Sports Medicine Centerthe Affiliated Suzhou Hospital of Nanjing Medical UniversitySuzhou215006China
- Gusu SchoolNanjing Medical UniversitySuzhou215006China
| | - Xueyan Li
- Gusu SchoolNanjing Medical UniversitySuzhou215006China
- Department of Anesthesiathe Affiliated Suzhou Hospital of Nanjing Medical UniversitySuzhou215006China
| | - Zeyu Han
- Department of Foot and Ankle SurgeryBeijing Tongren HospitalCapital Medical UniversityBeijing100730China
| | - Yinhua Qian
- Orthopedics and Sports Medicine Centerthe Affiliated Suzhou Hospital of Nanjing Medical UniversitySuzhou215006China
- Gusu SchoolNanjing Medical UniversitySuzhou215006China
| | - Lang Bai
- Orthopedics and Sports Medicine Centerthe Affiliated Suzhou Hospital of Nanjing Medical UniversitySuzhou215006China
- Gusu SchoolNanjing Medical UniversitySuzhou215006China
| | - Qibin Han
- Orthopedics and Sports Medicine Centerthe Affiliated Suzhou Hospital of Nanjing Medical UniversitySuzhou215006China
- Gusu SchoolNanjing Medical UniversitySuzhou215006China
| | - Maofeng Gao
- Department of Orthopaedicsthe First Affiliated Hospital of Soochow UniversitySuzhou215006China
| | - Yi Xue
- Department of OrthopaedicsChangshu Hospital Affiliated to Nanjing University of Traditional Chinese MedicineSuzhou215500China
| | - Dechun Geng
- Department of Orthopaedicsthe First Affiliated Hospital of Soochow UniversitySuzhou215006China
| | - Xing Yang
- Orthopedics and Sports Medicine Centerthe Affiliated Suzhou Hospital of Nanjing Medical UniversitySuzhou215006China
- Gusu SchoolNanjing Medical UniversitySuzhou215006China
| | - Yuefeng Hao
- Orthopedics and Sports Medicine Centerthe Affiliated Suzhou Hospital of Nanjing Medical UniversitySuzhou215006China
- Gusu SchoolNanjing Medical UniversitySuzhou215006China
| |
Collapse
|
6
|
Fan SH, Chang Y, Xiong XY, Xiang M, Yuan WL, Yang XQ, Wei WH, Chen L, Cheng MN, Zhu FH, He SJ, Zuo JP, Lin ZM. Reversible SAHH inhibitor ameliorates MIA-induced osteoarthritis of rats through suppressing MEK/ERK pathway. Biomed Pharmacother 2024; 170:115975. [PMID: 38070246 DOI: 10.1016/j.biopha.2023.115975] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2023] [Revised: 11/23/2023] [Accepted: 11/29/2023] [Indexed: 01/10/2024] Open
Abstract
Osteoarthritis (OA) is characterized by gradual articular cartilage degradation, accompanied by persistent low-grade joint inflammation, correlating with radiographic and pain-related progression. The latent therapeutic potential of DZ2002, a reversible inhibitor of S-adenosyl-L-homocysteine hydrolase (SAHH), holds promise for OA intervention. This study endeavored to examine the therapeutic efficacy of DZ2002 within the milieu of OA. The cytotoxicity of DZ2002 was evaluated using the MTT assay on bone marrow-derived macrophages. The inhibitory impact of DZ2002 during the process of osteoclastogenesis was assessed using TRAP staining, analysis of bone resorption pits, and F-actin ring formation. Mechanistic insights were derived from qPCR and Western blot analyses. Through the intra-articular injection of monosodium iodoacetate (MIA), an experimental rat model of OA was successfully instituted. This was subsequently accompanied by a series of assessments including Von Frey filament testing, analysis of weight-bearing behaviors, and micro-CT imaging, all aimed at assessing the effectiveness of DZ2002. The findings emphasized the effectiveness of DZ2002 in mitigating osteoclastogenesis induced by M-CSF/RANKL, evident through a reduction in TRAP-positive OCs and bone resorption. Moreover, DZ2002 modulated bone resorption-associated gene and protein expression (CTSK, CTR, Integrin β3) via the MEK/ERK pathway. Encouragingly, DZ2002 also alleviates MIA-induced pain, cartilage degradation, and bone loss. In conclusion, DZ2002 emerges as a potential therapeutic contender for OA, as evidenced by its capacity to hinder in vitro M-CSF/RANKL-induced osteoclastogenesis and mitigate in vivo osteoarthritis progression. This newfound perspective provides substantial support for considering DZ2002 as a compelling agent for osteoarthritis intervention.
Collapse
Affiliation(s)
- Shu-Hui Fan
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, 138 Xianlin Avenue, Nanjing, Jiangsu Province 210029, China; Laboratory of Immunopharmacology, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Yuan Chang
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, 138 Xianlin Avenue, Nanjing, Jiangsu Province 210029, China; Laboratory of Immunopharmacology, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Xiao-Yu Xiong
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, 138 Xianlin Avenue, Nanjing, Jiangsu Province 210029, China; Laboratory of Immunopharmacology, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Mai Xiang
- Laboratory of Immunopharmacology, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China; School of Pharmacy, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Wen-Long Yuan
- Laboratory of Immunopharmacology, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China; School of Pharmacy, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Xiao-Qian Yang
- Laboratory of Immunopharmacology, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Wen-Hui Wei
- Laboratory of Immunopharmacology, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China; School of Pharmacy, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Li Chen
- Laboratory of Immunopharmacology, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Meng-Nan Cheng
- Laboratory of Immunopharmacology, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Feng-Hua Zhu
- Laboratory of Immunopharmacology, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Shi-Jun He
- Innovation Research Institute of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China.
| | - Jian-Ping Zuo
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, 138 Xianlin Avenue, Nanjing, Jiangsu Province 210029, China; Laboratory of Immunopharmacology, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China; School of Pharmacy, University of Chinese Academy of Sciences, Beijing 100049, China; Experiment Center for Science and Technology, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China.
| | - Ze-Min Lin
- Laboratory of Immunopharmacology, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China; School of Pharmacy, University of Chinese Academy of Sciences, Beijing 100049, China.
| |
Collapse
|