1
|
Vijayan S, Margesan T. Comprehensive investigation of network pharmacology, computational modeling, and pharmacokinetic assessment to evaluate the efficacy of flavonoids in rheumatoid arthritis. Mol Divers 2025; 29:2265-2282. [PMID: 39348084 DOI: 10.1007/s11030-024-10989-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Accepted: 09/05/2024] [Indexed: 10/01/2024]
Abstract
Rheumatoid arthritis is a chronic autoimmune disease characterized by inflammation and joint damage, imposing a significant burden on affected individuals worldwide. Flavonoids, a class of natural compounds abundant in various plant-based foods, have shown promising anti-inflammatory and immunomodulatory effects, suggesting their potential as therapeutic agents for RA. In this study, we conducted a comprehensive investigation of identified LCMS compounds utilizing network pharmacology, computational modeling, in silico approaches, and pharmacokinetic assessment to evaluate the efficacy of flavonoids in RA treatment. The study identified 5 flavonoid structures with common targets via LCMS and Integration of network pharmacology approaches enabled a comprehensive evaluation of the pharmacological profile of flavonoids in the context of RA treatment, guiding the selection of promising candidates for further experimental validation and clinical development. The top 10 targets were AKT1, PI3KR1, CDK2, EGFR, CDK6, NOS2, FLT3, ALOX5, CCNB1, and PTPRS via PPI network. The investigation emphasized several pathways, including the AGE-RAGE signaling pathway, resistance to EGFR tyrosine kinase inhibitors, the PI3K-AKT signaling network, and the Rap 1 signaling pathway. In silico studies estimated binding affinities that ranged from - 7.0 to - 10.0 kcal/mol. Schaftoside and Vitexin showed no toxicity in computational approach and found suitable for further investigations. Overall, our study underscores the potential of flavonoids as therapeutic agents for RA and highlights the utility of integrative approaches combining network pharmacology, computational modeling, in silico methods, and pharmacokinetic assessment in drug discovery and development processes.
Collapse
Affiliation(s)
- Sukanya Vijayan
- Department of Pharmacognosy, SRM College of Pharmacy, SRM Institute of Science and Technology, Kattankulathur, Chengalpattu, Tamil Nadu, 603203, India
| | - Thirumal Margesan
- Department of Pharmacognosy, SRM College of Pharmacy, SRM Institute of Science and Technology, Kattankulathur, Chengalpattu, Tamil Nadu, 603203, India.
| |
Collapse
|
2
|
El-Sayed JM, Khadrawy SM, Mohamed AE, Mohamed HM, Mohamed T, Aly MS. Therapeutic effects of a femtosecond laser on rheumatoid arthritis in rats: Attenuation of oxidative stress and inflammation. JOURNAL OF PHOTOCHEMISTRY AND PHOTOBIOLOGY. B, BIOLOGY 2025; 266:113150. [PMID: 40088516 DOI: 10.1016/j.jphotobiol.2025.113150] [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: 08/03/2024] [Revised: 02/16/2025] [Accepted: 03/10/2025] [Indexed: 03/17/2025]
Abstract
Rheumatoid arthritis (RA) is a chronic inflammatory autoimmune disorder characterized by joint damage and persistent pain. Despite advances in treatment, there is currently no definitive cure for RA, and the side effects of available medications often limit their long-term use. Therefore, this study investigated the therapeutic potential of femtosecond laser irradiation (FSL) in treating arthritis induced by complete Freund's adjuvant (CFA) in a rat model. Twenty-four adult male Wistar rats were divided into four groups. Groups 1 and 2 served, respectively, as the negative control and positive FSL irradiated group (at a wavelength of 830 nm, a power of 200 mW, an exposure time of 120 s, a beam area of 0.8 cm2 (0.5 cm radius), a power density of 0.25 W/cm2, and an energy dose of 30 J/cm2). Group 3 represented the arthritic group that received a single subcutaneous injection of 0.1 ml CFA. Group 4 was the arthritic rats irradiated with FSL (two sessions/week) for three weeks. Morphological changes including edema and swelling that increased the circumference of the right hind paw, as well as histological alterations marked by cellular infiltration, synovitis, and cartilage degeneration confirmed RA in the ankle joints. These changes correlated with elevated levels of rheumatoid factor (RF), TNF-α, and IL-6, and augmented oxidative stress associated with a declined antioxidant defense system. Exposure to FSL ameliorated the morphological and histopathological changes in the ankle joint, decreased RF, TNF-α, IL-6, and MDA, and increased GSH and GPx. In conclusion, femtosecond laser irradiation showed antioxidant and anti-inflammatory properties and exerted regenerative effects on the histological features of the ankle in a rheumatoid arthritis rat model.
Collapse
Affiliation(s)
- Jihad M El-Sayed
- Genetics and Molecular Biology Division, Zoology Department, Faculty of Science, Beni-Suef University, 62521 Beni-Suef, Egypt
| | - Sally M Khadrawy
- Department of Biology, College of Science, Imam Mohammad Ibn Saud Islamic University, Riyadh 11623, Saudi Arabia
| | - Aya E Mohamed
- Laser Institute for Research and Applications LIRA, Beni-Suef University, Beni-Suef 62511, Egypt
| | - Hanaa M Mohamed
- Genetics and Molecular Biology Division, Zoology Department, Faculty of Science, Beni-Suef University, 62521 Beni-Suef, Egypt
| | - Tarek Mohamed
- Laser Institute for Research and Applications LIRA, Beni-Suef University, Beni-Suef 62511, Egypt.
| | - Magdy Sayed Aly
- Genetics and Molecular Biology Division, Zoology Department, Faculty of Science, Beni-Suef University, 62521 Beni-Suef, Egypt
| |
Collapse
|
3
|
Zhang T, Ouyang Z, Zhang Y, Sun H, Kong L, Xu Q, Qu J, Sun Y. Marine Natural Products in Inflammation-Related Diseases: Opportunities and Challenges. Med Res Rev 2025. [PMID: 40202793 DOI: 10.1002/med.22109] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2025] [Revised: 03/14/2025] [Accepted: 03/18/2025] [Indexed: 04/11/2025]
Abstract
In recent decades, the potentiality of marine natural products (MNPs) in the medical field has been increasingly recognized. Natural compounds derived from marine microorganisms, algae, and invertebrates have shown significant promise for treating inflammation-related diseases. In this review, we cover the three primary sources of MNPs and their diverse and unique chemical structures and bioactivities. This review aims to summarize the progress of MNPs in combating inflammation-related diseases. Moreover, we cover the functions and mechanisms of MNPs in diseases, highlighting their functions in regulating inflammatory signaling pathways, cellular stress responses, and gut microbiota, among others. Meanwhile, we focus on key technologies and scientific methods to address the current limitations and challenges in MNPs.
Collapse
Affiliation(s)
- Tao Zhang
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Chemistry and Biomedicine Innovation Center (ChemBIC), Nanjing University, Nanjing, China
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou, China
| | - Zijun Ouyang
- School of Food and Drug, Shenzhen Polytechnic University, Shenzhen, China
| | - Yueran Zhang
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Chemistry and Biomedicine Innovation Center (ChemBIC), Nanjing University, Nanjing, China
| | - Haiyan Sun
- School of Food and Drug, Shenzhen Polytechnic University, Shenzhen, China
| | - Lingdong Kong
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Chemistry and Biomedicine Innovation Center (ChemBIC), Nanjing University, Nanjing, China
| | - Qiang Xu
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Chemistry and Biomedicine Innovation Center (ChemBIC), Nanjing University, Nanjing, China
| | - Jiao Qu
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Chemistry and Biomedicine Innovation Center (ChemBIC), Nanjing University, Nanjing, China
| | - Yang Sun
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Chemistry and Biomedicine Innovation Center (ChemBIC), Nanjing University, Nanjing, China
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou, China
| |
Collapse
|
4
|
Wang H, Yuan T, Wang J, Li D, Lee WYW, Li Z, Sun S. Quercetagetin alleviates inflammatory osteoclastogenesis and collagen antibody-induced arthritis via Nrf2 signaling and Pten/AKT/Nfatc1 axis. Arthritis Res Ther 2025; 27:54. [PMID: 40057805 PMCID: PMC11889843 DOI: 10.1186/s13075-025-03522-x] [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: 08/15/2024] [Accepted: 02/26/2025] [Indexed: 05/13/2025] Open
Abstract
PURPOSE Quercetagetin, a flavonoid derived from the natural herb Flos eriocauli, is used in traditional Chinese medicine for its fire-purging (anti-inflammation) and wind-expelling (pain-alleviating) properties. However, its potential effects concerning rheumatoid arthritis (RA) remain underexplored. This study was designed to elucidate the potential associations between Quercetagetin and RA, establishing the therapeutic potential of Quercetagetin and related mechanisms in RA treatment. METHODS Network pharmacology was conducted to decipher related targets and signaling pathways between Quercetagetin and RA. In vitro assays were then conducted to explore the effects of Quercetagetin on osteoclast cell behaviors and corresponding signaling pathways. In vivo study further validated the therapeutic effect of Quercetagetin in collagen antibody-induced arthritis (CAIA) mice. RESULTS The network pharmacological analysis indicated an intimate correlation of Quercetagetin with RA-related inflammatory osteolysis treatment. Pertaining to biological validations, 2 µM of Quercetagetin successfully inhibited LPS-driven osteoclast differentiation and function. qPCR assay and Western blot analyses denoted parallel changes in osteoclastic marker genes and proteins. Further mechanism study uncovered the effect of Quercetagetin in stimulating the Nrf2/Keap1 signaling pathway and moderating the Pten/AKT/Nfatc1 axis in osteoclasts. In vivo study revealed 40 mg/kg Quercetagetin every day could significantly relief joint destruction in CAIA mice. CONCLUSIONS Our study presents Quercetagetin 's therapeutic potential in treating RA, outlining its effects and potential mechanisms in suppressing LPS-induced osteoclast activity, and alleviating inflammatory bone destruction in CAIA model, thereby laying the groundwork for further translational research on Quercetagetin and Flos eriocauli in RA treatment.
Collapse
Affiliation(s)
- Haojue Wang
- Department of Joint Surgery, Cheeloo College of Medicine, Shandong Provincial Hospital, Shandong University, Jinan, Shandong, 250012, China
- Department of Joint Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250021, China
| | - Tao Yuan
- Department of Joint Surgery, Cheeloo College of Medicine, Shandong Provincial Hospital, Shandong University, Jinan, Shandong, 250012, China
- Department of Joint Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250021, China
| | | | - Dengju Li
- Department of Joint Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250021, China
- Orthopaedic Research Laboratory, Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, 250117, China
| | - Wayne Yuk-Wai Lee
- Department of Orthopaedics and Traumatology, The Chinese University of Hong Kong, Hong Kong, China.
- SH Ho Scoliosis Research Laboratory, Joint Scoliosis Research Centre of the Chinese University of Hong Kong and Nanjing University, The Chinese University of Hong Kong, Hong Kong, China.
- Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong, China.
| | - Ziqing Li
- Department of Joint Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250021, China.
- Orthopaedic Research Laboratory, Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, 250117, China.
| | - Shui Sun
- Department of Joint Surgery, Cheeloo College of Medicine, Shandong Provincial Hospital, Shandong University, Jinan, Shandong, 250012, China.
- Department of Joint Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250021, China.
- Orthopaedic Research Laboratory, Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, 250117, China.
| |
Collapse
|
5
|
Wang H, Yuan T, Yu X, Wang Y, Liu C, Li Z, Sun S. Norwogonin Attenuates Inflammatory Osteolysis and Collagen-Induced Arthritis via Modulating Redox Signalling and Calcium Oscillations. J Cell Mol Med 2025; 29:e70492. [PMID: 40099974 PMCID: PMC11915625 DOI: 10.1111/jcmm.70492] [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: 08/26/2024] [Revised: 02/06/2025] [Accepted: 03/05/2025] [Indexed: 03/20/2025] Open
Abstract
Norwogonin is a flavonoid extraction derived from Scutellaria baicalensis. However, its potential mechanisms in the context of rheumatoid arthritis (RA) are unclear. This study investigates the specific effects and associated targets of Norwogonin in RA-related inflammatory osteolysis. Network pharmacology was conducted to analyse the core targets and signalling pathways of Norwogonin in RA. In vitro experiments were carried out to explore the actual effects of Norwogonin on osteoclast behaviours and related signalling mechanisms. In vivo studies further validated the therapeutic effect of Norwogonin in collagen-induced arthritis (CIA) mice. The network pharmacological analysis identified 18 shared targets between Norwogonin and RA, indicating a connection with inflammatory response and oxidoreductase activity. For biological validations, the results of in vitro experiments revealed 160 μM of Norwogonin inhibited LPS-driven osteoclast differentiation and function. The qPCR assay and Western blot analysis also disclosed consistently diminished changes to osteoclastic marker genes and proteins due to Norwogonin treatment, including those for osteoclast differentiation (Traf6, Tnfrsf11a and Nfatc1), fusion (Atp6v0d2, Dcstamp and Ocstamp) and function (Mmp9, Ctsk and Acp5). Further mechanism study revealed Norwogonin suppressed LPS-driven ROS production and calcium (Ca2+) oscillations. Also, intraperitoneal injection of 30 mg/kg Norwogonin every other day successfully mitigated clinical arthritis progression and attenuated bone destruction in the CIA model. Our study scrutinises Norwogonin's therapeutic prospects in treating RA and illustrates its inhibitory effects and potential mechanism within LPS-induced osteoclastogenesis and CIA mice, providing a basis for further translational research on Norwogonin in the treatment of RA-related inflammatory osteolysis.
Collapse
Affiliation(s)
- Haojue Wang
- Department of Joint SurgeryShandong Provincial Hospital, Cheeloo College of Medicine, Shandong UniversityJinanShandongChina
- Department of Joint SurgeryShandong Provincial Hospital Affiliated to Shandong First Medical UniversityJinanShandongChina
| | - Tao Yuan
- Department of Joint SurgeryShandong Provincial Hospital, Cheeloo College of Medicine, Shandong UniversityJinanShandongChina
- Department of Joint SurgeryShandong Provincial Hospital Affiliated to Shandong First Medical UniversityJinanShandongChina
| | - Xiao Yu
- Department of Obstetrics and GynecologyJian Gong HospitalBeijingChina
| | - Yi Wang
- Department of Joint SurgeryShandong Provincial Hospital Affiliated to Shandong First Medical UniversityJinanShandongChina
- Orthopaedic Research LaboratoryMedical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical SciencesJinanShandongChina
| | - Changxing Liu
- Department of Joint SurgeryShandong Provincial Hospital, Cheeloo College of Medicine, Shandong UniversityJinanShandongChina
| | - Ziqing Li
- Department of Joint SurgeryShandong Provincial Hospital Affiliated to Shandong First Medical UniversityJinanShandongChina
- Orthopaedic Research LaboratoryMedical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical SciencesJinanShandongChina
| | - Shui Sun
- Department of Joint SurgeryShandong Provincial Hospital, Cheeloo College of Medicine, Shandong UniversityJinanShandongChina
- Department of Joint SurgeryShandong Provincial Hospital Affiliated to Shandong First Medical UniversityJinanShandongChina
- Orthopaedic Research LaboratoryMedical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical SciencesJinanShandongChina
| |
Collapse
|
6
|
Song H, Yu J, Yang Y, Zhou L, Liu X, Yu J, Huang Q, Wang S, Zhang X, Liu Y, Zhang D, Meng J, Han T, Li W, Niu X. Exploring molecular mechanism of Panlongqi Tablet (PLQT) against RA: Integrated network pharmacology, molecular docking and experiment validation. Int Immunopharmacol 2025; 144:113639. [PMID: 39616851 DOI: 10.1016/j.intimp.2024.113639] [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: 09/10/2024] [Revised: 11/11/2024] [Accepted: 11/12/2024] [Indexed: 12/15/2024]
Abstract
BACKGROUND AND PURPOSE Panlongqi Tablet (PLQT), a proprietary Chinese medicine composed of 29 herbs, has been included in the Chinese Medical Insurance List and has shown promising therapeutic effects on patients with rheumatoid arthritis (RA) in clinical practice. However, the molecular mechanisms of PLQT against RA have not been fully elucidated. This study aimed to further decipher the active ingredients and molecular mechanism of PLQT anti-RA. METHODS A Complete Freund's adjuvant (CFA)-induced rat model was established to evaluate the pharmacodynamic effects of PLQT against RA, the assessment included arthritis index, paw thickness, ankle diameter, morphological and histopathological analysis. Network analysis was used to elucidate the active ingredients and underlying mechanisms of PLQT in the treatment of RA, molecular docking was conducted to assess the binding of active ingredients to key targets. In vitro and in vivo experimental verification were employed to reveal the mechanism of PLQT against RA. RESULTS Experimentally, PLQT improved CFA-induced arthritis without evident side effects. Network analysis revealed that the active ingredients in PLQT were mainly flavonoids, biscoumarin derivatives, alklaloid and lignans. Integrated with molecular docking studies, the molecular mechanisms of PLQT against RA were enriched in inflammatory response, immune regulation, angiogenesis, osteoclast differentiation and autophagy. In vitro experiments confirmed that PLQT exerted anti-inflammatory and immune regulatory effects by targeting the inflammatory response of M1 macrophages and the biological functions of T lymphocytes. In addition, In vivo experiments verified that PLQT could further inhibit synovial angiogenesis to prevent RA. CONCLUSION This study integrated network pharmacology analysis, molecular docking and experimental validation to elucidate the active components of PLQT and its mechanisms in intervening the pathological progression of RA, providing a more comprehensive theoretical basis for the clinical application of PLQT in the treatment of RA.
Collapse
Affiliation(s)
- Huixin Song
- School of Pharmacy, Xi'an Jiaotong University, Xi'an, PR China
| | - Jinjin Yu
- School of Pharmacy, Xi'an Jiaotong University, Xi'an, PR China
| | - Yajie Yang
- School of Pharmacy, Xi'an Jiaotong University, Xi'an, PR China
| | - Lili Zhou
- School of Pharmacy, Xi'an Jiaotong University, Xi'an, PR China
| | - Xinyao Liu
- School of Pharmacy, Xi'an Jiaotong University, Xi'an, PR China
| | - Jiabao Yu
- School of Pharmacy, Xi'an Jiaotong University, Xi'an, PR China
| | - Qiuxia Huang
- School of Pharmacy, Xi'an Jiaotong University, Xi'an, PR China
| | - Siqi Wang
- School of Pharmacy, Xi'an Jiaotong University, Xi'an, PR China
| | - Xinya Zhang
- School of Pharmacy, Xi'an Jiaotong University, Xi'an, PR China
| | - Yang Liu
- School of Pharmacy, Xi'an Jiaotong University, Xi'an, PR China
| | - Dezhu Zhang
- Shaanxi Panlong Pharmaceutical Group Limited by Share LTD, Xi'an, PR China
| | - Jianguo Meng
- Shaanxi Panlong Pharmaceutical Group Limited by Share LTD, Xi'an, PR China
| | - Tengfei Han
- Shaanxi Panlong Pharmaceutical Group Limited by Share LTD, Xi'an, PR China
| | - Weifeng Li
- School of Pharmacy, Xi'an Jiaotong University, Xi'an, PR China.
| | - Xiaofeng Niu
- School of Pharmacy, Xi'an Jiaotong University, Xi'an, PR China.
| |
Collapse
|
7
|
Chaudhary S, Sharma S, Fuloria S, Sharma PK. Anti-Inflammatory and Anti-Arthritis Activity of Quercetin: A Comprehensive Review. Curr Rheumatol Rev 2025; 21:144-159. [PMID: 38808723 DOI: 10.2174/0115733971280645240415101912] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2023] [Revised: 01/11/2024] [Accepted: 01/24/2024] [Indexed: 05/30/2024]
Abstract
This comprehensive exploration delves into the multifaceted attributes of quercetin, a flavonoid with extensive health-promoting potential. The review navigates through its fundamental properties, encompassing its chemical structure, classification as a flavonoid, and its natural prevalence in various sources. Addressing solubility, stability, and bioavailability challenges, the investigation delves into innovative isolation techniques, including solvent extraction, solid-phase extraction, natural deep eutectic solvents, supercritical fluid extraction, microwave-assisted extraction, column chromatography, and high-performance thin-layer chromatography. Transitioning into pharmacological implications, the study unveils quercetin's roles in anti-inflammatory pathways, antioxidant effects, and immune modulation, reflecting its versatile significance in health management. The review highlights its impact on wound healing processes and its potential to mitigate arthritis, elucidating its holistic contributions. Culminating in an exploration of recent studies, the analysis underscores quercetin's remarkable anti-inflammatory and anti-arthritis activities, reflecting its substantial potential across various ailments. The review concludes by projecting future trajectories, emphasizing prospects for an advanced understanding of quercetin's mechanisms, sustainable extraction techniques, clinical integration, and exploration of synergistic combinations. Collectively, this review investigation underscores quercetin's dynamic role at the intersection of natural compounds and medicinal applications, offering profound implications for well- being and health enhancement.
Collapse
Affiliation(s)
- Shikha Chaudhary
- Department of Pharmacy, School of Medical and Allied Sciences, Galgotias University, Uttar Pradesh, India
| | - Shaweta Sharma
- Department of Pharmacy, School of Medical and Allied Sciences, Galgotias University, Uttar Pradesh, India
| | - Shivkanya Fuloria
- Faculty of Pharmacy, AIMST University, Semeling Campus, Bedong, Kedah Aman, Malaysia
| | - Pramod Kumar Sharma
- Department of Pharmacy, School of Medical and Allied Sciences, Galgotias University, Uttar Pradesh, India
| |
Collapse
|
8
|
Feng Y, Pan X, Li Z, Li Y, Sun Y, Yang S, He C, Dang Y, Huang L, Xiang B. Innovative Lipid Nanoparticles Co-Delivering Hydroxychloroquine and siRNA for Enhanced Rheumatoid Arthritis Therapy. Pharmaceutics 2025; 17:45. [PMID: 39861693 PMCID: PMC11769357 DOI: 10.3390/pharmaceutics17010045] [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: 12/10/2024] [Revised: 12/25/2024] [Accepted: 12/26/2024] [Indexed: 01/27/2025] Open
Abstract
Background: Rheumatoid arthritis (RA) is a debilitating autoimmune disorder characterized by chronic inflammation and joint damage. Despite advancements in treatment, complete remission remains elusive. Methods: In this study, we introduce a novel lipid nanoparticle formulation co-delivering hydroxychloroquine (HCQ) and siRNA targeting TNF-α (siTNF-α) using microfluidic technology, marking the first use of such a combination for RA therapy. Results: In LPS-stimulated RAW 264.7 cells, the nanoparticles effectively reduced inflammatory markers. When administered via an intra-articular injection in a rat model, they significantly decreased joint inflammation and demonstrated good biological safety. Conclusions: This pioneering approach highlights the potential of lipid nanoparticles as a dual-delivery platform for enhanced RA treatment through targeted intra-articular administration.
Collapse
Affiliation(s)
- Yanru Feng
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Hebei Medical University, Shijiazhuang 050017, China; (Y.F.); (X.P.); (Z.L.); (Y.L.); (Y.S.); (S.Y.); (C.H.); (Y.D.)
| | - Xintong Pan
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Hebei Medical University, Shijiazhuang 050017, China; (Y.F.); (X.P.); (Z.L.); (Y.L.); (Y.S.); (S.Y.); (C.H.); (Y.D.)
| | - Ziqian Li
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Hebei Medical University, Shijiazhuang 050017, China; (Y.F.); (X.P.); (Z.L.); (Y.L.); (Y.S.); (S.Y.); (C.H.); (Y.D.)
| | - Yue Li
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Hebei Medical University, Shijiazhuang 050017, China; (Y.F.); (X.P.); (Z.L.); (Y.L.); (Y.S.); (S.Y.); (C.H.); (Y.D.)
| | - Ya’nan Sun
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Hebei Medical University, Shijiazhuang 050017, China; (Y.F.); (X.P.); (Z.L.); (Y.L.); (Y.S.); (S.Y.); (C.H.); (Y.D.)
| | - Shaokun Yang
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Hebei Medical University, Shijiazhuang 050017, China; (Y.F.); (X.P.); (Z.L.); (Y.L.); (Y.S.); (S.Y.); (C.H.); (Y.D.)
| | - Chaoxing He
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Hebei Medical University, Shijiazhuang 050017, China; (Y.F.); (X.P.); (Z.L.); (Y.L.); (Y.S.); (S.Y.); (C.H.); (Y.D.)
| | - Yunjie Dang
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Hebei Medical University, Shijiazhuang 050017, China; (Y.F.); (X.P.); (Z.L.); (Y.L.); (Y.S.); (S.Y.); (C.H.); (Y.D.)
| | - Lu Huang
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Hebei Medical University, Shijiazhuang 050017, China; (Y.F.); (X.P.); (Z.L.); (Y.L.); (Y.S.); (S.Y.); (C.H.); (Y.D.)
| | - Bai Xiang
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Hebei Medical University, Shijiazhuang 050017, China; (Y.F.); (X.P.); (Z.L.); (Y.L.); (Y.S.); (S.Y.); (C.H.); (Y.D.)
- Hebei Key Laboratory of Innovative Drug Research and Evaluation, Hebei Medical University, Shijiazhuang 050017, China
| |
Collapse
|
9
|
Wu J, Lv T, Liu Y, Liu Y, Han Y, Liu X, Peng X, Tang F, Cai J. The role of quercetin in NLRP3-associated inflammation. Inflammopharmacology 2024; 32:3585-3610. [PMID: 39306817 DOI: 10.1007/s10787-024-01566-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2024] [Accepted: 08/30/2024] [Indexed: 11/10/2024]
Abstract
Quercetin is a natural flavonoid that is widely found in fruits and vegetables. As an important flavonoid, it exhibits a wide range of biological activities, including antioxidant, anti-inflammatory, antiviral, immunomodulatory, and analgesic activities. Quercetin exerts powerful antioxidant activity by regulating glutathione, enzyme activity, and the production of reactive oxygen species (ROS). Quercetin exerts powerful anti-inflammatory effects by acting on the Nod-like receptor protein 3 (NLRP3) inflammasome. In diabetes, quercetin has been shown to improve insulin sensitivity and reduce high blood sugar level, while, in neurological diseases, it potentially prevents neuronal degeneration and cognitive decline by regulating neuroinflammation. In addition, in liver diseases, quercetin may improve liver inflammation and fibrosis by regulating the NLRP3 activity. In addition, quercetin may improve inflammation in other diseases based on the NLRP3 inflammasome. With this background, in this review, we have discussed the progress in the study on the mechanism of quercetin toward improving inflammation via NLRP3 inflammasome in the past decade. In addition, from the perspective of quercetin glycoside derivatives, the anti-inflammatory mechanism of hyperoside, rutin, and isoquercetin based on NLRP3 inflammasome has been discussed. Moreover, we have discussed the pharmacokinetics of quercetin and its nanoformulation application, with the aim to provide new ideas for further research on the anti-inflammatory effect of quercetin and its glycoside derivatives based on NLRP3 inflammasome, as well as in drug development and application.
Collapse
Affiliation(s)
- Jiaqi Wu
- Department of Pathophysiology, School of Basic Medicine, Health Science Center, Yangtze University, Jingzhou, 434023, China
- Laboratory of Oncology, Center for Molecular Medicine, School of Basic Medicine, Health Science Center, Yangtze University, Jingzhou, 434023, China
| | - Tongtong Lv
- Department of Pathophysiology, School of Basic Medicine, Health Science Center, Yangtze University, Jingzhou, 434023, China
- Laboratory of Oncology, Center for Molecular Medicine, School of Basic Medicine, Health Science Center, Yangtze University, Jingzhou, 434023, China
| | - Yu Liu
- Department of Oncology, Gong'an County People's Hospital, Jingzhou, 434000, China
| | - Yifan Liu
- Laboratory of Oncology, Center for Molecular Medicine, School of Basic Medicine, Health Science Center, Yangtze University, Jingzhou, 434023, China
- Department of Oncology, Jingzhou Hospital Affiliated to Yangtze University, Jingzhou, 434023, China
| | - Yukun Han
- Laboratory of Oncology, Center for Molecular Medicine, School of Basic Medicine, Health Science Center, Yangtze University, Jingzhou, 434023, China
- Department of Medical Imaging, School of Medicine, and Positron Emission Computed Tomography (PET) Center of the First Affiliated Hospital, Yangtze University, Jingzhou, 434023, China
| | - Xin Liu
- Department of Pathophysiology, School of Basic Medicine, Health Science Center, Yangtze University, Jingzhou, 434023, China
- Laboratory of Oncology, Center for Molecular Medicine, School of Basic Medicine, Health Science Center, Yangtze University, Jingzhou, 434023, China
| | - Xiaochun Peng
- Department of Pathophysiology, School of Basic Medicine, Health Science Center, Yangtze University, Jingzhou, 434023, China.
- Laboratory of Oncology, Center for Molecular Medicine, School of Basic Medicine, Health Science Center, Yangtze University, Jingzhou, 434023, China.
| | - Fengru Tang
- Radiation Physiology Laboratory, Singapore Nuclear Research and Safety Initiative, National University of Singapore, 1 CREATE Way #04-01, CREATE Tower, Singapore, 138602, Singapore.
| | - Jun Cai
- Department of Oncology, First Affiliated Hospital of Yangtze University, Jingzhou, 434023, China.
| |
Collapse
|
10
|
Anjiki K, Hayashi S, Ikuta K, Suda Y, Kamenaga T, Tsubosaka M, Kuroda Y, Nkano N, Maeda T, Tsumiyama K, Matsumoto T, Kuroda R, Matsubara T. JAK inhibitors inhibit angiogenesis by reducing VEGF production from rheumatoid arthritis-derived fibroblast-like synoviocytes. Clin Rheumatol 2024; 43:3525-3536. [PMID: 39302595 DOI: 10.1007/s10067-024-07142-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Revised: 09/03/2024] [Accepted: 09/09/2024] [Indexed: 09/22/2024]
Abstract
INTRODUCTION/OBJECTIVES JAK/STAT signaling inhibition exerts therapeutic effects on angiogenesis in rheumatoid arthritis (RA). However, whether the inhibitory effect differs among JAK inhibitors because of differing selectivity is unknown. Therefore, we compared the inhibitory effects of tofacitinib, baricitinib, peficitinib, upadacitinib, and filgotinib on angiogenesis. METHOD RA-derived fibroblast-like synoviocytes (RA-FLS) were seeded on type I collagen gel, and human umbilical vein endothelial cells (HUVECs) were directly added. The control and aforementioned JAK inhibitors were added to the medium, followed by stimulation with interleukin (IL)-6 and soluble IL-6 receptor (sIL-6R). Each JAK inhibitor's concentration was determined based on estimated blood concentrations. The vascular endothelial growth factor (VEGF) concentration was evaluated with an enzyme-linked immunosorbent assay using the medium from the first exchange. A migration assay was performed, and HUVEC migration was evaluated using CD31 fluorescence immunostaining. RESULTS Hematoxylin-eosin staining showed that compared with the non-JAKi treatment group, the JAKi treatment group markedly degenerated in the sub-lining and deep lining, with decreased lymphocyte infiltration and neovascularization [Rooney's score subscale, non-JAKi vs JAKi (median, 6.5 vs 2.5, p = 0.005)]. In vitro, IL-6 and sIL-6R administration increased VEGF production from RA-FLS and promoted neovascularization in HUVECs, and JAK-inhibitor administration, which decreased VEGF production from RA-FLS and suppressed HUVEC migration, inhibited neovascularization in RA-FLS and HUVEC co-cultures. CONCLUSIONS The JAK inhibitors suppressed IL-6-induced angiogenesis via decreased VEGF production and HUVEC migration in RA-FLS and HUVEC co-cultures. No significant differences were observed among the JAK inhibitors, whose anti-angiogenic effect may be an important mechanism for RA treatment. Key Points • JAK inhibitors inhibit angiogenesis in RA by reducing VEGF production from RA-derived fibroblast-like synoviocytes. • Our study provides new insights into RA treatment by elucidating the anti-angiogenic effect of JAK inhibitors.
Collapse
Affiliation(s)
- Kensuke Anjiki
- Department of Orthopedic Surgery, Kobe University Graduate School of Medicine, 7-5-1, Kusunoki-Cho, Chuo-Ku, Kobe, Hyogo, 650-0017, Japan
| | - Shinya Hayashi
- Department of Orthopedic Surgery, Kobe University Graduate School of Medicine, 7-5-1, Kusunoki-Cho, Chuo-Ku, Kobe, Hyogo, 650-0017, Japan.
| | - Kenmei Ikuta
- Department of Orthopedic Surgery, Kobe University Graduate School of Medicine, 7-5-1, Kusunoki-Cho, Chuo-Ku, Kobe, Hyogo, 650-0017, Japan
| | - Yoshihito Suda
- Department of Orthopedic Surgery, Kobe University Graduate School of Medicine, 7-5-1, Kusunoki-Cho, Chuo-Ku, Kobe, Hyogo, 650-0017, Japan
| | - Tomoyui Kamenaga
- Department of Orthopedic Surgery, Kobe University Graduate School of Medicine, 7-5-1, Kusunoki-Cho, Chuo-Ku, Kobe, Hyogo, 650-0017, Japan
| | - Masanori Tsubosaka
- Department of Orthopedic Surgery, Kobe University Graduate School of Medicine, 7-5-1, Kusunoki-Cho, Chuo-Ku, Kobe, Hyogo, 650-0017, Japan
| | - Yuichi Kuroda
- Department of Orthopedic Surgery, Kobe University Graduate School of Medicine, 7-5-1, Kusunoki-Cho, Chuo-Ku, Kobe, Hyogo, 650-0017, Japan
| | - Naoki Nkano
- Department of Orthopedic Surgery, Kobe University Graduate School of Medicine, 7-5-1, Kusunoki-Cho, Chuo-Ku, Kobe, Hyogo, 650-0017, Japan
| | - Toshihisa Maeda
- Department of Orthopedic Surgery, Matsubara Mayflower Hospital, 944-25, Fujita, Katō, Hyogo, Japan
| | - Ken Tsumiyama
- Department of Orthopedic Surgery, Matsubara Mayflower Hospital, 944-25, Fujita, Katō, Hyogo, Japan
| | - Tomoyuki Matsumoto
- Department of Orthopedic Surgery, Kobe University Graduate School of Medicine, 7-5-1, Kusunoki-Cho, Chuo-Ku, Kobe, Hyogo, 650-0017, Japan
| | - Ryosuke Kuroda
- Department of Orthopedic Surgery, Kobe University Graduate School of Medicine, 7-5-1, Kusunoki-Cho, Chuo-Ku, Kobe, Hyogo, 650-0017, Japan
| | - Tsukasa Matsubara
- Department of Orthopedic Surgery, Matsubara Mayflower Hospital, 944-25, Fujita, Katō, Hyogo, Japan
| |
Collapse
|
11
|
Li Y, Liu W, Wang Y, Liu T, Feng Y. Nanotechnology-Mediated Immunomodulation Strategy for Inflammation Resolution. Adv Healthc Mater 2024; 13:e2401384. [PMID: 39039994 DOI: 10.1002/adhm.202401384] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Revised: 07/02/2024] [Indexed: 07/24/2024]
Abstract
Inflammation serves as a common characteristic across a wide range of diseases and plays a vital role in maintaining homeostasis. Inflammation can lead to tissue damage and the onset of inflammatory diseases. Although significant progress is made in anti-inflammation in recent years, the current clinical approaches mainly rely on the systemic administration of corticosteroids and antibiotics, which only provide short-term relief. Recently, immunomodulatory approaches have emerged as promising strategies for facilitating the resolution of inflammation. Especially, the advanced nanosystems with unique biocompatibility and multifunctionality have provided an ideal platform for immunomodulation. In this review, the pathophysiology of inflammation and current therapeutic strategies are summarized. It is mainly focused on the nanomedicines that modulate the inflammatory signaling pathways, inflammatory cells, oxidative stress, and inflammation targeting. Finally, the challenges and opportunities of nanomaterials in addressing inflammation are also discussed. The nanotechnology-mediated immunomodulation will open a new treatment strategy for inflammation therapy.
Collapse
Affiliation(s)
- Ying Li
- School of Chemical Engineering and Technology, Tianjin University, Yaguan Road 135, Tianjin, 300350, P. R. China
| | - Wen Liu
- School of Chemical Engineering and Technology, Tianjin University, Yaguan Road 135, Tianjin, 300350, P. R. China
| | - Yuanchao Wang
- School of Chemical Engineering and Technology, Tianjin University, Yaguan Road 135, Tianjin, 300350, P. R. China
| | - Taotao Liu
- Department of Gastroenterology and Hepatology, Characteristic Medical Center of the Chinese People's Armed Police Force, Tianjin Key Laboratory of Hepatopancreatic Fibrosis and Molecular Diagnosis & Treatment, Tianjin, 300162, China
| | - Yakai Feng
- School of Chemical Engineering and Technology, Tianjin University, Yaguan Road 135, Tianjin, 300350, P. R. China
- Frontiers Science Center for Synthetic Biology, Tianjin University, Weijin Road 92, Tianjin, 300072, P. R. China
- Key Laboratory of Systems Bioengineering (Ministry of Education), Tianjin University, Weijin Road 92, Tianjin, 300072, P. R. China
| |
Collapse
|
12
|
Chen S, Zhao W, Du J, Chen S, Li J, Shen B, Zhou Y, Chen S. The expression of RBPJ and its potential role in rheumatoid arthritis. BMC Genomics 2024; 25:899. [PMID: 39350019 PMCID: PMC11441141 DOI: 10.1186/s12864-024-10804-2] [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: 04/24/2024] [Accepted: 09/16/2024] [Indexed: 10/04/2024] Open
Abstract
BACKGROUND Recombination signal-binding protein for immunoglobulin kappa J region (RBPJ) is a transcriptional regulator that plays an important role in maintaining immune homeostasis. This study aimed to estimate the expression of RBPJ in rheumatoid arthritis (RA) patients and investigate its relationship with RA. METHODS A total of 83 newly diagnosed RA patients and 70 healthy controls were included. mRNA was extracted from peripheral blood mononuclear cells (PBMCs), and the expression of RBPJ was detected using quantitative real-time PCR (qRT‒PCR). An RA dataset (GSE89408) was obtained from the Gene Expression Omnibus (GEO) database, and RA synovial tissues were divided into two groups. The differentially expressed genes (DEGs) were selected with the "DESeq2" R package. RESULTS RBPJ expression was lower in RA patients than in health controls and was negatively correlated with the DAS28 score, C-reactive protein (CRP) level and erythrocyte sedimentation rate (ESR). RA synovial tissues from GSE89408 were classified into RBPJ-low (≤ 25%) and RBPJ-high (≥ 75%) groups according to RBPJ expression, and 562 DEGs were identified. Gene Ontology (GO) enrichment analyses revealed that the DEGs significantly affected the regulation of T cell activation and lymphocyte/mononuclear cell differentiation. Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analysis revealed that the most enriched pathways of DEGs were the T cell receptor signaling pathway, Th1/2 and Th17 cell differentiation, the PI3K - Akt signaling pathway and cytokine‒cytokine receptor interaction. CytoHubba Plugin revealed that most of the top 10 genes were involved in osteoclast differentiation, the T cell receptor signaling pathway and cytokine‒cytokine receptor interaction. CONCLUSIONS RBPJ expression was significantly lower in RA patients and negatively correlated with disease activity. GEO dataset analysis demonstrated that RBPJ may be involved in osteoclast differentiation, T cell activation and differentiation, and the T cell receptor signaling pathway. Our research may contribute to understanding the potential mechanisms by which RBPJ regulates T cell differentiation and cytokine‒cytokine receptor interaction in RA patients.
Collapse
Affiliation(s)
- Shuaishuai Chen
- Department of Clinical Laboratory, Taizhou Hospital of Zhejiang Province affiliated to Wenzhou Medical University, 150 Ximen Street of Linhai City, Linhai, 317000, China
- Key Laboratory of System Medicine and Precision Diagnosis and Treatment of Taizhou, Luqiao, China
| | - Weibo Zhao
- Department of Orthopedics, Taizhou Hospital of Zhejiang Province affiliated to Wenzhou Medical University, Linhai, China
| | - Juping Du
- Department of Clinical Laboratory, Taizhou Hospital of Zhejiang Province affiliated to Wenzhou Medical University, 150 Ximen Street of Linhai City, Linhai, 317000, China
- Key Laboratory of System Medicine and Precision Diagnosis and Treatment of Taizhou, Luqiao, China
| | - Suyun Chen
- Department of Clinical Laboratory, Taizhou Hospital of Zhejiang Province affiliated to Wenzhou Medical University, 150 Ximen Street of Linhai City, Linhai, 317000, China
- Key Laboratory of System Medicine and Precision Diagnosis and Treatment of Taizhou, Luqiao, China
| | - Jun Li
- Department of Clinical Laboratory, Taizhou Hospital of Zhejiang Province affiliated to Wenzhou Medical University, 150 Ximen Street of Linhai City, Linhai, 317000, China
- Key Laboratory of System Medicine and Precision Diagnosis and Treatment of Taizhou, Luqiao, China
| | - Bo Shen
- Department of Clinical Laboratory, Taizhou Hospital of Zhejiang Province affiliated to Wenzhou Medical University, 150 Ximen Street of Linhai City, Linhai, 317000, China
- Key Laboratory of System Medicine and Precision Diagnosis and Treatment of Taizhou, Luqiao, China
| | - Yuanlin Zhou
- Department of Neurology, Taizhou Hospital of Zhejiang Province affiliated to Wenzhou Medical University, 150 Ximen Street of Linhai City, Linhai, 317000, China.
| | - Shiyong Chen
- Department of Clinical Laboratory, Taizhou Hospital of Zhejiang Province affiliated to Wenzhou Medical University, 150 Ximen Street of Linhai City, Linhai, 317000, China.
- Key Laboratory of System Medicine and Precision Diagnosis and Treatment of Taizhou, Luqiao, China.
| |
Collapse
|
13
|
Li Y, Wang X, Gao Y, Zhang Z, Liu T, Zhang Z, Wang Y, Chang F, Yang M. Hyaluronic acid-coated polypeptide nanogel enhances specific distribution and therapy of tacrolimus in rheumatoid arthritis. J Nanobiotechnology 2024; 22:547. [PMID: 39238027 PMCID: PMC11378632 DOI: 10.1186/s12951-024-02784-y] [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] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Accepted: 08/18/2024] [Indexed: 09/07/2024] Open
Abstract
Rheumatoid arthritis (RA) involves chronic inflammation, oxidative stress, and complex immune cell interactions, leading to joint destruction. Traditional treatments are often limited by off-target effects and systemic toxicity. This study introduces a novel therapeutic approach using hyaluronic acid (HA)-conjugated, redox-responsive polyamino acid nanogels (HA-NG) to deliver tacrolimus (TAC) specifically to inflamed joints. The nanogels' disulfide bonds enable controlled TAC release in response to high intracellular glutathione (GSH) levels in activated macrophages, prevalent in RA-affected tissues. In vitro results demonstrated that HA-NG/TAC significantly reduced TAC toxicity to normal macrophages and showed high biocompatibility. In vivo, HA-NG/TAC accumulated more in inflamed joints compared to non-targeted NG/TAC, enhancing therapeutic efficacy and minimizing side effects. Therapeutic evaluation in collagen-induced arthritis (CIA) mice revealed HA-NG/TAC substantially reduced paw swelling, arthritis scores, synovial inflammation, and bone erosion while suppressing pro-inflammatory cytokine levels. These findings suggest that HA-NG/TAC represents a promising targeted drug delivery system for RA, offering potential for more effective and safer clinical applications.
Collapse
Affiliation(s)
- Yuhuan Li
- Department of Orthopedics, China-Japan Union Hospital of Jilin University, Changchun, 130033, PR China
| | - Xin Wang
- Department of Orthopedics, The Second Hospital of Jilin University, Changchun, 130041, PR China
| | - Yu Gao
- Department of Orthopedics, China-Japan Union Hospital of Jilin University, Changchun, 130033, PR China
- Department of Orthopedics, The Second Hospital of Jilin University, Changchun, 130041, PR China
| | - Ziyi Zhang
- Department of Orthopedics, China-Japan Union Hospital of Jilin University, Changchun, 130033, PR China
| | - Te Liu
- Scientific Research Center, China-Japan Union Hospital of Jilin University, Changchun, Jilin, China
- Yibin Jilin University Research Institute, Jilin University, Yibin, Sichuan, China
| | - Zhuo Zhang
- Department of Orthopedics, China-Japan Union Hospital of Jilin University, Changchun, 130033, PR China
| | - Yinan Wang
- Department of Biobank, Division of Clinical Research, The First Hospital of Jilin University, Changchun, China
| | - Fei Chang
- Department of Orthopedics, The Second Hospital of Jilin University, Changchun, 130041, PR China.
| | - Modi Yang
- Department of Orthopedics, China-Japan Union Hospital of Jilin University, Changchun, 130033, PR China.
| |
Collapse
|
14
|
Kang W, Xu Q, Dong H, Wang W, Huang G, Zhang J. Eriodictyol attenuates osteoarthritis progression through inhibiting inflammation via the PI3K/AKT/NF-κB signaling pathway. Sci Rep 2024; 14:18853. [PMID: 39143134 PMCID: PMC11324885 DOI: 10.1038/s41598-024-69028-9] [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: 03/18/2024] [Accepted: 07/30/2024] [Indexed: 08/16/2024] Open
Abstract
Eriodictyol, a flavonoid distributed in citrus fruits, has been known to exhibit anti-inflammatory activity. In this study, destabilized medial meniscus (DMM)-induced OA model was used to investigate the protective role of eriodictyol on OA. Meanwhile, we used an IL-1β-stimulated human osteoarthritis chondrocytes model to investigate the anti-inflammatory mechanism of eriodictyol on OA. The production of nitric oxide was detected by Griess reaction. The productions of MMP1, MMP3, and PGE2 were detected by ELISA. The expression of LXRα, ABCA1, PI3K, AKT, and NF-κB were measured by western blot analysis. The results demonstrated that eriodictyol could alleviate DMM-induced OA in mice. In vitro, eriodictyol inhibited IL-1β-induced NO, PGE2, MMP1, and MMP3 production in human osteoarthritis chondrocytes. Eriodictyol also suppressed the phosphorylation of PI3K, AKT, NF-κB p65, and IκBα induced by IL-1β. Meanwhile, eriodictyol significantly increased the expression of LXRα and ABCA1. Furthermore, eriodictyol disrupted lipid rafts formation through reducing the cholesterol content. And cholesterol replenishment experiment showed that adding water-soluble cholesterol could reverse the anti-inflammatory effect of eriodictyol. In conclusion, the results indicated eriodictyol inhibited IL-1β-induced inflammation in human osteoarthritis chondrocytes through suppressing lipid rafts formation, which subsequently inhibiting PI3K/AKT/NF-κB signaling pathway.
Collapse
Affiliation(s)
- Wenbo Kang
- Department of Orthopedics, China-Japan Union Hospital of Jilin University, 126 Xiantai Street, Changchun, 130033, People's Republic of China
| | - Qinli Xu
- Department of Orthopedics, China-Japan Union Hospital of Jilin University, 126 Xiantai Street, Changchun, 130033, People's Republic of China
| | - Hang Dong
- Department of Orthopedics, China-Japan Union Hospital of Jilin University, 126 Xiantai Street, Changchun, 130033, People's Republic of China
| | - Wenjun Wang
- Department of Orthopedics, China-Japan Union Hospital of Jilin University, 126 Xiantai Street, Changchun, 130033, People's Republic of China
| | - Guanning Huang
- Department of Orthopedics, China-Japan Union Hospital of Jilin University, 126 Xiantai Street, Changchun, 130033, People's Republic of China
| | - Jingzhe Zhang
- Department of Orthopedics, China-Japan Union Hospital of Jilin University, 126 Xiantai Street, Changchun, 130033, People's Republic of China.
| |
Collapse
|
15
|
Verma VK, Mutneja E, Malik S, Sahu AK, Prajapati V, Bhardwaj P, Ray R, Nag TC, Bhatia J, Arya DS. Abatacept: A Promising Repurposed Solution for Myocardial Infarction-Induced Inflammation in Rat Models. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2024; 2024:3534104. [PMID: 38957586 PMCID: PMC11219209 DOI: 10.1155/2024/3534104] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/30/2023] [Revised: 02/12/2024] [Accepted: 02/28/2024] [Indexed: 07/04/2024]
Abstract
Myocardial infarction (MI) is irreversible damage to the myocardial tissue caused by prolonged ischemia/hypoxia, subsequently leading to loss of contractile function and myocardial damage. However, after a perilous period, ischemia-reperfusion (IR) itself causes the generation of oxygen free radicals, disturbance in cation homeostasis, depletion of cellular energy stores, and activation of innate and adaptive immune responses. The present study employed Abatacept (ABT), which is an anti-inflammatory drug, originally used as an antirheumatic response agent. To investigate the cardioprotective potential of ABT, primarily, the dose was optimized in a chemically induced model of myocardial necrosis. Thereafter, ABT optimized the dose of 5 mg/kg s.c. OD was investigated for its cardioprotective potential in a surgical model of myocardial IR injury, where animals (n = 30) were randomized into five groups: Sham, IR-C, Telmi10 + IR (Telmisartan, 10 mg/kg oral OD), ABT5 + IR, ABT perse. ABT and telmisartan were administered for 21 days. On the 21st day, animals were subjected to LAD coronary artery occlusion for 60 min, followed by reperfusion for 45 min. Further, the cardioprotective potential was assessed through hemodynamic parameters, oxidant-antioxidant biochemical enzymatic parameters, cardiac injury, inflammatory markers, histopathological analysis, TUNEL assay, and immunohistochemical evaluation, followed by immunoblotting to explore signaling pathways. The statistics were performed by one-way analysis of variance, followed by the Tukey comparison post hoc tests. Noteworthy, 21 days of ABT pretreatment amended the hemodynamic and ventricular functions in the rat models of MI. The cardioprotective potential of ABT is accompanied by inhibiting MAP kinase signaling and modulating Nrf-2/HO-1 proteins downstream signaling cascade. Overall, the present work bolsters the previously known anti-inflammatory role of ABT in MI and contributes a mechanistic insight and application of clinically approved drugs in averting the activation of inflammatory response.
Collapse
Affiliation(s)
- Vipin Kumar Verma
- Cardiovascular Research Laboratory, Department of Pharmacology, All India Institute of Medical Sciences, New Delhi-110029, India
| | - Ekta Mutneja
- Cardiovascular Research Laboratory, Department of Pharmacology, All India Institute of Medical Sciences, New Delhi-110029, India
| | - Salma Malik
- Cardiovascular Research Laboratory, Department of Pharmacology, All India Institute of Medical Sciences, New Delhi-110029, India
| | - Anil Kumar Sahu
- Cardiovascular Research Laboratory, Department of Pharmacology, All India Institute of Medical Sciences, New Delhi-110029, India
| | - Vaishali Prajapati
- Cardiovascular Research Laboratory, Department of Pharmacology, All India Institute of Medical Sciences, New Delhi-110029, India
| | - Priya Bhardwaj
- Cardiovascular Research Laboratory, Department of Pharmacology, All India Institute of Medical Sciences, New Delhi-110029, India
| | - Ruma Ray
- Cardiac Pathology Laboratory, Department of Pathology, All India Institute of Medical Sciences, New Delhi-110029, India
| | - Tapas Chandra Nag
- Department of Anatomy, All India Institute of Medical Sciences, New Delhi-110029, India
| | - Jagriti Bhatia
- Cardiovascular Research Laboratory, Department of Pharmacology, All India Institute of Medical Sciences, New Delhi-110029, India
| | - Dharamvir Singh Arya
- Cardiovascular Research Laboratory, Department of Pharmacology, All India Institute of Medical Sciences, New Delhi-110029, India
| |
Collapse
|
16
|
Hung SY, Chen JL, Tu YK, Tsai HY, Lu PH, Jou IM, Mbuyisa L, Lin MW. Isoliquiritigenin inhibits apoptosis and ameliorates oxidative stress in rheumatoid arthritis chondrocytes through the Nrf2/HO-1-mediated pathway. Biomed Pharmacother 2024; 170:116006. [PMID: 38091640 DOI: 10.1016/j.biopha.2023.116006] [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: 09/14/2023] [Revised: 12/03/2023] [Accepted: 12/06/2023] [Indexed: 01/10/2024] Open
Abstract
Rheumatoid arthritis (RA) is a chronic inflammatory condition known for its irreversible destructive impact on the joints. Chondrocytes play a pivotal role in the production and maintenance of the cartilage matrix. However, the presence of inflammatory cytokines can hinder chondrocyte proliferation and promote apoptosis. Isoliquiritigenin (ISL), a flavonoid, potentially exerts protective effects against various inflammatory diseases. However, its specific role in regulating the nuclear factor E2-associated factor 2 (Nrf2)/heme oxygenase-1 (HO-1) pathway in chondrocytes in RA remains unclear. To investigate this, this study used human chondrocytes and Sprague-Dawley rats to construct in vitro and in vivo RA models, respectively. The study findings reveal that cytokines markedly induced oxidative stress, the activation of matrix metalloproteinases, and apoptosis both in vitro and in vivo. Notably, ISL treatment significantly mitigated these effects. Moreover, Nrf2 or HO-1 inhibitors reversed the protective effects of ISL, attenuated the expression of Nrf2/HO-1 and peroxisome proliferator-activated receptor gamma-coactivator-1α, and promoted chondrocyte apoptosis. This finding indicates that ISL primarily targets the Nrf2/HO-1 pathway in RA chondrocytes. Moreover, ISL treatment led to improved behavior scores, reduced paw thickness, and mitigated joint damage as well as ameliorated oxidative stress in skeletal muscles in an RA rat model. In conclusion, this study highlights the pivotal role of the Nrf2/HO-1 pathway in the protective effects of ISL and demonstrates the potential of ISL as a treatment option for RA.
Collapse
Affiliation(s)
- Shih-Ya Hung
- Graduate Institute of Acupuncture Science, China Medical University, Taichung 40402, Taiwan; Division of Surgery, Department of Medical Research, China Medical University Hospital, Taichung 40447, Taiwan
| | - Jen-Lung Chen
- Department of Surgery, E-Da Hospital, Kaohsiung 82445, Taiwan
| | - Yuan-Kun Tu
- Department of Orthopaedic Surgery, E-Da Hospital, I-Shou University, Kaohsiung, Taiwan; School of Medicine, College of Medicine, I-Shou University, Kaohsiung, Taiwan
| | - Hsin-Yi Tsai
- Department of Medical Research, E-Da Hospital/ E-Da Cancer Hospital, Kaohsiung 82445, Taiwan
| | - Pin-Hsuan Lu
- Department of Medical Research, E-Da Hospital/ E-Da Cancer Hospital, Kaohsiung 82445, Taiwan
| | - I-Ming Jou
- Department of Orthopaedic Surgery, E-Da Hospital, I-Shou University, Kaohsiung, Taiwan
| | - Lulekiwe Mbuyisa
- School of Medicine for International Students, College of Medicine, I-Shou University, Kaohsiung 82445, Taiwan
| | - Ming-Wei Lin
- Department of Medical Research, E-Da Hospital/ E-Da Cancer Hospital, Kaohsiung 82445, Taiwan; Department of Nursing, College of Medicine, I-Shou University, Kaohsiung 82445, Taiwan; Regenerative Medicine and Cell Therapy Research Center, Kaohsiung Medical University, Kaohsiung 80708, Taiwan.
| |
Collapse
|
17
|
Shubina VS, Kozina VI, Shatalin YV. A Comparative Study of the Inhibitory Effect of Some Flavonoids and a Conjugate of Taxifolin with Glyoxylic Acid on the Oxidative Burst of Neutrophils. Int J Mol Sci 2023; 24:15068. [PMID: 37894747 PMCID: PMC10606308 DOI: 10.3390/ijms242015068] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Revised: 09/30/2023] [Accepted: 10/09/2023] [Indexed: 10/29/2023] Open
Abstract
During the storage, processing, and digestion of flavonoid-rich foods and beverages, a condensation of flavonoids with toxic carbonyl compounds occurs. The effect of the resulting products on cells remains largely unknown. The aim of the present study was to evaluate the effects of quercetin, taxifolin, catechin, eriodictyol, hesperetin, naringenin, and a condensation product of taxifolin with glyoxylic acid on the oxidative burst of neutrophils. It was found that the flavonoids and the condensation product inhibited the total production of ROS. Flavonoids decreased both the intra and extracellular ROS production. The condensation product had no effect on intracellular ROS production but effectively inhibited the extracellular production of ROS. Thus, the condensation of flavonoids with toxic carbonyl compounds may lead to the formation of compounds exhibiting potent inhibitory effects on the oxidative burst of neutrophils. The data also suggest that, during these reactions, the influence of a fraction of flavonoids and their polyphenolic derivatives on cellular functions may change. On the whole, the results of the study provide a better understanding of the effects of polyphenols on human health. In addition, these results reveal the structure-activity relationship of these polyphenols and may be useful in a search for new therapeutic agents against diseases associated with oxidative stress.
Collapse
Affiliation(s)
- Victoria S. Shubina
- Institute of Theoretical and Experimental Biophysics, Russian Academy of Sciences, Institutskaya 3, 142290 Pushchino, Russia;
| | | | - Yuri V. Shatalin
- Institute of Theoretical and Experimental Biophysics, Russian Academy of Sciences, Institutskaya 3, 142290 Pushchino, Russia;
| |
Collapse
|