1
|
Dixit T, Vaidya A, Ravindran S. Polymeric nanoparticles-based targeted delivery of drugs and bioactive compounds for arthritis management. Future Sci OA 2025; 11:2467591. [PMID: 39973324 PMCID: PMC11845113 DOI: 10.1080/20565623.2025.2467591] [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: 07/11/2024] [Accepted: 02/07/2025] [Indexed: 02/21/2025] Open
Abstract
This review explores the potential of polymeric nanoparticles (PNPs) as targeted drug delivery systems for arthritic treatment, overcoming the limitations of the present therapy. A thorough literature search was conducted on the databases PubMed, Scopus, and Web of Science to find published articles on the use of polymeric nanoparticles in the treatment of arthritis. This includes synthesis methods, mechanisms in drug delivery, and applications of PNPs. Polymeric nanoparticles showed excellent promise in the management of arthritis through enhanced stability of drugs, controlled and sustained drug release, and reduced systemic side effects. Some of the highlighted biocompatible and targeting capabilities of natural and synthetic polymers include chitosan, hyaluronic acid, and PLGA. Bioactive compounds such as curcumin and resveratrol delivered by PNPs enhanced therapeutic efficacy in preclinical arthritis models. Despite their promise, challenges such as rapid clearance and manufacturing scalability remain critical barriers. Polymeric nanoparticles offer a transformative approach to arthritis management by enabling targeted, sustained, and safe drug delivery. Translation into clinical applications would thus require developments in nanoparticle design, personalized medicine, and scalable production techniques.
Collapse
Affiliation(s)
- Tanu Dixit
- Symbiosis School of Biological Sciences, Symbiosis International (Deemed University), Pune, India
| | - Anuradha Vaidya
- Symbiosis School of Biological Sciences, Symbiosis International (Deemed University), Pune, India
| | - Selvan Ravindran
- Symbiosis School of Biological Sciences, Symbiosis International (Deemed University), Pune, India
| |
Collapse
|
2
|
Chen C, Wei Q, Zhu Z, Wu W, Gong T, Lv Y, Zhang A, Hu C, Zhan Q, Cao T, Cao P. A thermoresponsive magnetic nanocarrier with the DNA-mimetic base pairing-guided controlled release of methotrexate in the treatment of rheumatoid arthritis. J Control Release 2025; 384:113952. [PMID: 40513670 DOI: 10.1016/j.jconrel.2025.113952] [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: 01/18/2025] [Revised: 06/08/2025] [Accepted: 06/10/2025] [Indexed: 06/16/2025]
Abstract
Controlled drug delivery systems not only augment the therapeutic efficacy of drugs but also mitigate their adverse effects. Methotrexate (MTX) is a prominent first-line therapeutic agent in the management of rheumatoid arthritis (RA), yet it is characterized by its limited aqueous solubility, pronounced hemolytic activity, and propensity for off-target binding, which collectively contribute to its significant toxicity profile. To address these issues, a temperature-responsive controlled release system for MTX based on the analogous base pairing rule was designed for the RA treatment. Magnetic ferroferric oxide nanoparticles (Fe3O4 NPs) coated with polydopamine (PDA) were modified with thymidine-1-acetic acid (TAA) to prepare nanocarrier (FPT NPs). Furthermore, the thymine molecules within the FPT NPs possessed the strong ability to specifically bind with the pteridine structure present in MTX, thereby enabling the fabrication of a nanomedicine (designated as FPT-MTX NPs) through the base pairing rule. Under the influence of an applied magnetic field, the FPT-MTX NPs demonstrated a remarkable capacity for precisely targeting the joint tissue in a mouse model of RA. Analogous to DNA, where double-strand breaks occurred due to heating-induced disruption of base pairing, MTX was released from FPT-MTX NPs following near-infrared irradiation-induced light-to-heat conversion. This mechanism facilitated the achievement of satisfactory therapeutic outcomes in the RA treatment.
Collapse
Affiliation(s)
- Cheng Chen
- State Key Laboratory of Technologies for Chinese Medicine Pharmaceutical Process Control and Intelligent Manufacture, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210023, PR China
| | - Qingyun Wei
- Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou People's Hospital, Quzhou, Zhejiang 324000, PR China; Jiangsu Provincial Medicinal Innovation Center, Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210028, PR China
| | - Zhen Zhu
- College of Health and Rehabilitation & College of Acupuncture and Massage, Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210023, PR China
| | - Wenhui Wu
- State Key Laboratory of Technologies for Chinese Medicine Pharmaceutical Process Control and Intelligent Manufacture, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210023, PR China
| | - Tiantian Gong
- State Key Laboratory of Technologies for Chinese Medicine Pharmaceutical Process Control and Intelligent Manufacture, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210023, PR China
| | - Yangbo Lv
- Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou People's Hospital, Quzhou, Zhejiang 324000, PR China
| | - Aimei Zhang
- State Key Laboratory of Technologies for Chinese Medicine Pharmaceutical Process Control and Intelligent Manufacture, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210023, PR China
| | - Chunping Hu
- Jiangsu Provincial Medicinal Innovation Center, Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210028, PR China
| | - Qichen Zhan
- State Key Laboratory of Technologies for Chinese Medicine Pharmaceutical Process Control and Intelligent Manufacture, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210023, PR China; Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou People's Hospital, Quzhou, Zhejiang 324000, PR China.
| | - Tao Cao
- State Key Laboratory of Technologies for Chinese Medicine Pharmaceutical Process Control and Intelligent Manufacture, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210023, PR China.
| | - Peng Cao
- Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou People's Hospital, Quzhou, Zhejiang 324000, PR China; Jiangsu Provincial Medicinal Innovation Center, Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210028, PR China.
| |
Collapse
|
3
|
Liu Z, Yang J, Tan G, Shi Y, Tao D, Wang W, Li B, Jin F, He X. Methotrexate loaded extracellular vesicles attenuate periodontitis by suppressing ACSL1 and promoting anti-inflammatory macrophage. Mol Immunol 2025; 182:83-95. [PMID: 40245705 DOI: 10.1016/j.molimm.2025.04.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2025] [Revised: 03/31/2025] [Accepted: 04/14/2025] [Indexed: 04/19/2025]
Abstract
Macrophages are crucial immune cells in periodontal tissues, which play key roles in both the destruction and repair of associated with periodontitis. Targeted modulation of macrophage function has emerged as a potentially effective approach to influence periodontitis progression. This study investigates the effects of methotrexate-loaded extracellular vesicles (MTX-EVs) on inflammatory macrophage polarization both in vivo and in vitro. In a murine periodontitis model, MTX-EVs inhibited alveolar bone resorption, suppressed pro-inflammatory macrophage activation, and promoted anti-inflammatory macrophages. Mechanistically, MTX-EVs reduced acyl-CoA synthetase-1 (ACSL1) expression, which was elevated during inflammation. Inhibition of ACSL1 with triacsin-C in macrophages suppressed the inflammatory phenotype through the promotion of the oxidative phosphorylation (OXPHOS). In contrast, MTX-EVs counteracted the effects of ACSL1 overexpression on macrophage polarization and metabolism. Our findings suggest that targeting ACSL1 via MTX-EVs represents a therapeutic strategy for modulating macrophage polarization and improving periodontitis treatment outcomes.
Collapse
Affiliation(s)
- Zhi Liu
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi International Joint Research Center for Oral Diseases, Center for Tissue Engineering, School of Stomatology, The Fourth Military Medical University, Xi'an 710032, China; State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi Clinical Research Center for Oral Diseases, Department of Orthodontics, School of Stomatology, The Fourth Military Medical University, Xi'an 710032, China
| | - Jianhua Yang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi International Joint Research Center for Oral Diseases, Center for Tissue Engineering, School of Stomatology, The Fourth Military Medical University, Xi'an 710032, China
| | - Guodong Tan
- Air Force Medical Center, The Fourth Military Medical University, Beijing 100142, China
| | - Yuanyuan Shi
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi International Joint Research Center for Oral Diseases, Department of Periodontology, School of Stomatology, The Fourth Military Medical University, Xi'an 710032, China
| | - Dihao Tao
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi International Joint Research Center for Oral Diseases, Center for Tissue Engineering, School of Stomatology, The Fourth Military Medical University, Xi'an 710032, China
| | - Wenzhe Wang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi International Joint Research Center for Oral Diseases, Center for Tissue Engineering, School of Stomatology, The Fourth Military Medical University, Xi'an 710032, China
| | - Bei Li
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi International Joint Research Center for Oral Diseases, Center for Tissue Engineering, School of Stomatology, The Fourth Military Medical University, Xi'an 710032, China
| | - Fang Jin
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi Clinical Research Center for Oral Diseases, Department of Orthodontics, School of Stomatology, The Fourth Military Medical University, Xi'an 710032, China.
| | - Xiaoning He
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi International Joint Research Center for Oral Diseases, Center for Tissue Engineering, School of Stomatology, The Fourth Military Medical University, Xi'an 710032, China.
| |
Collapse
|
4
|
Cao J, Zhang H, Ni Y, Ning X. Neferine reduces synovial inflammation and cardiac complications in a collagen-induced arthritis mouse model via inhibiting NF-κB/NLRP3 inflammasome axis. Mol Immunol 2025; 182:117-125. [PMID: 40262197 DOI: 10.1016/j.molimm.2025.04.001] [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: 12/02/2024] [Revised: 02/21/2025] [Accepted: 04/03/2025] [Indexed: 04/24/2025]
Abstract
For the treatment of rheumatoid arthritis (RA), there are limited options for drugs with fewer side effects. Neferine possesses anti-inflammatory, anti-fibrotic, and cardioprotective properties, but its effectiveness in the treatment of RA remains unclear. Our study aimed to explore the impact of neferine administration on ankle joint inflammation and cardiac complications of collagen-induced arthritis (CIA) mice. The CIA model was introduced in male DBA/1 mice via subcutaneous injection of Type II collagen (CII) into the tail. We found that neferine alleviated ankle joint inflammation, cartilage erosion, and bone destruction, as well as reduced the levels of IL-6, TNF-α, IL-1β, and IL-18 in the serum of CIA mice. Furthermore, neferine reduced the expression of synovial damage markers (RANKL, MMP-3, and MMP-13) in the ankle joints of CIA mice. Mechanistically, neferine lowered the levels of NF-κB pathway-related molecules such as p-IκBα, p-p65, and nuclear p65 in the synovial tissue of CIA mice. Simultaneously, neferine reversed the upregulation of NLRP3, ASC, and cleaved-caspase-1 levels in the synovial tissue of CIA mice. Additionally, our results showed that neferine reduced the contents of myocardial injury markers (cTnI, CK-MB, and LDH), alleviated myocardial fibrosis, decreased expression of α-SMA and Collagen I, as well as mitigated the activation of fibrosis-related TGF-β/Smad signaling. In summary, our study demonstrates that neferine attenuates chondral and synovial inflammation in a CIA mouse model by inhibiting the activation of the NF-κB/NLRP3 inflammasome, and neferine has a protective effect on the hearts of CIA mice.
Collapse
Affiliation(s)
- Jingjing Cao
- Department of Rheumatology and Immunology, Hebei General Hospital, Shijiazhuang, Hebei 050051, China.
| | - Huaxing Zhang
- Division of Medical Service, Hebei General Hospital, Shijiazhuang, Hebei 050051, China
| | - Yanhui Ni
- Department of Cardiology, Hebei General Hospital, Shijiazhuang, Hebei 050051, China
| | - Xiaoran Ning
- Department of Rheumatology and Immunology, Hebei General Hospital, Shijiazhuang, Hebei 050051, China
| |
Collapse
|
5
|
Wang Y, Jan H, Zhong Z, Zhou L, Teng K, Chen Y, Xu J, Xie D, Chen D, Xu J, Qin L, Tuan RS, Li ZA. Multiscale metal-based nanocomposites for bone and joint disease therapies. Mater Today Bio 2025; 32:101773. [PMID: 40290898 PMCID: PMC12033929 DOI: 10.1016/j.mtbio.2025.101773] [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: 03/09/2025] [Revised: 04/15/2025] [Accepted: 04/16/2025] [Indexed: 04/30/2025] Open
Abstract
Bone and joint diseases are debilitating conditions that can result in significant functional impairment or even permanent disability. Multiscale metal-based nanocomposites, which integrate hierarchical structures ranging from the nanoscale to the macroscale, have emerged as a promising solution to this challenge. These materials combine the unique properties of metal-based nanoparticles (MNPs), such as enzyme-like activities, stimuli responsiveness, and photothermal conversion, with advanced manufacturing techniques, such as 3D printing and biohybrid systems. The integration of MNPs within polymer or ceramic matrices offers a degree of control over the mechanical strength, antimicrobial efficacy, and the manner of drug delivery, whilst concomitantly promoting the processes of osteogenesis and chondrogenesis. This review highlights breakthroughs in stimulus-responsive MNPs (e.g., photo-, magnetically-, or pH-activated systems) for on-demand therapy and their integration with biocomposite hybrids containing cells or extracellular vesicles to mimic the native tissue microenvironment. The applications of these composites are extensive, ranging from bone defects, infections, tumors, to degenerative joint diseases. The review emphasizes the enhanced load-bearing capacity, bioactivity, and tissue integration that can be achieved through hierarchical designs. Notwithstanding the potential of these applications, significant barriers to progress persist, including challenges related to long-term biocompatibility, regulatory hurdles, and scalable manufacturing. Finally, we propose future directions, including machine learning-guided design and patient-specific biomanufacturing to accelerate clinical translation. Multiscale metal-based nanocomposites, which bridge nanoscale innovations with macroscale functionality, are a revolutionary force in the field of biomedical engineering, providing personalized regenerative solutions for bone and joint diseases.
Collapse
Affiliation(s)
- Yuwen Wang
- Department of Biomedical Engineering, Faculty of Engineering, The Chinese University of Hong Kong, Shatin, Hong Kong Special Administrative Region of China
| | - Hasnain Jan
- Department of Biomedical Engineering, Faculty of Engineering, The Chinese University of Hong Kong, Shatin, Hong Kong Special Administrative Region of China
- Center for Neuromusculoskeletal Restorative Medicine, Hong Kong Science Park, NT, Hong Kong Special Administrative Region of China
- Department of Biomedical Engineering, City University of Hong Kong, Kowloon, Hong Kong Special Administrative Region of China
| | - Zheng Zhong
- Department of Biomedical Engineering, Faculty of Engineering, The Chinese University of Hong Kong, Shatin, Hong Kong Special Administrative Region of China
- Department of Orthopedic Surgery, Center for Orthopedic Surgery, and Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, The Third Affiliated Hospital of Southern Medical University, Guangzhou, 510630, China
| | - Liangbin Zhou
- Department of Biomedical Engineering, Faculty of Engineering, The Chinese University of Hong Kong, Shatin, Hong Kong Special Administrative Region of China
- Center for Neuromusculoskeletal Restorative Medicine, Hong Kong Science Park, NT, Hong Kong Special Administrative Region of China
| | - Kexin Teng
- Department of Biomedical Engineering, Faculty of Engineering, The Chinese University of Hong Kong, Shatin, Hong Kong Special Administrative Region of China
- Center for Neuromusculoskeletal Restorative Medicine, Hong Kong Science Park, NT, Hong Kong Special Administrative Region of China
| | - Ye Chen
- Department of Chemistry, Faculty of Science, The Chinese University of Hong Kong, Hong Kong Special Administrative Region of China
| | - Jiankun Xu
- Musculoskeletal Research Laboratory, Department of Orthopedics & Traumatology, Faculty of Medicine, and Innovative Orthopedic Biomaterial and Drug Translational Research Laboratory, Li Ka Shing Institute of Health Sciences, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong Special Administrative Region of China
| | - Denghui Xie
- Department of Orthopedic Surgery, Center for Orthopedic Surgery, and Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, The Third Affiliated Hospital of Southern Medical University, Guangzhou, 510630, China
| | - Dexin Chen
- Institute of Advanced Wear & Corrosion Resistant and Functional Materials, Jinan University, Guangzhou, 510632, China
| | - Jiake Xu
- Faculty of Pharmaceutical Sciences, Shenzhen University of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Ling Qin
- Musculoskeletal Research Laboratory, Department of Orthopedics & Traumatology, Faculty of Medicine, and Innovative Orthopedic Biomaterial and Drug Translational Research Laboratory, Li Ka Shing Institute of Health Sciences, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong Special Administrative Region of China
| | - Rocky S. Tuan
- Department of Biomedical Engineering, Faculty of Engineering, The Chinese University of Hong Kong, Shatin, Hong Kong Special Administrative Region of China
- Center for Neuromusculoskeletal Restorative Medicine, Hong Kong Science Park, NT, Hong Kong Special Administrative Region of China
- Institute for Tissue Engineering and Regenerative Medicine, and School of Biomedical Sciences, The Chinese University of Hong Kong, Shatin, Hong Kong Special Administrative Region of China
| | - Zhong Alan Li
- Department of Biomedical Engineering, Faculty of Engineering, The Chinese University of Hong Kong, Shatin, Hong Kong Special Administrative Region of China
- Center for Neuromusculoskeletal Restorative Medicine, Hong Kong Science Park, NT, Hong Kong Special Administrative Region of China
- Institute for Tissue Engineering and Regenerative Medicine, and School of Biomedical Sciences, The Chinese University of Hong Kong, Shatin, Hong Kong Special Administrative Region of China
- Shun Hing Institute of Advanced Engineering, The Chinese University of Hong Kong, NT, Hong Kong Special Administrative Region of China
| |
Collapse
|
6
|
Harimohan H, Huynh Q, Yasonova M, Moosavi L, Khamlong M, Garcia Pacheco I. Two Consecutive Days of Low-Dose Methotrexate Toxicity: A Diagnostic Challenge. Cureus 2025; 17:e86023. [PMID: 40520835 PMCID: PMC12167167 DOI: 10.7759/cureus.86023] [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] [Accepted: 06/09/2025] [Indexed: 06/18/2025] Open
Abstract
Methotrexate is an immunosuppressive medication commonly used to treat rheumatological disorders, primarily by inhibiting the folic acid cycle, with dose-dependent toxicity affecting multiple organ systems. A 54-year-old woman with a history of rheumatoid arthritis (RA), previously treated with methotrexate but later switched to leflunomide, etanercept, and prednisone, presented to the emergency department due to abnormal lab results. After running out of leflunomide and experiencing increased joint pain, she resumed methotrexate for two consecutive days without folic acid supplementation. Three days later, she developed oral ulcers, blisters, decreased oral intake, and fatigue. Lab results revealed pancytopenia, with markedly low white blood cells, hemoglobin, platelets, and absolute neutrophil count. Initially, Stevens-Johnson Syndrome (SJS) was considered due to mucosal symptoms, but lack of rash made methotrexate toxicity more likely. Rheumatology and hematology consultations led to the discontinuation of methotrexate, administration of filgrastim and leucovorin, and subsequent clinical improvement. This case highlights the diagnostic challenge in differentiating methotrexate toxicity from SJS, as both can present with mucosal lesions, though pancytopenia pointed toward toxicity. Despite methotrexate's known dose-dependent toxicity, this patient's reaction at a low dose suggests a rare idiosyncratic response, underscoring the importance of vigilance even with standard dosing and the necessity of folic acid supplementation to reduce adverse effects.
Collapse
Affiliation(s)
| | - Quynh Huynh
- Rheumatology, Kern Medical, Bakersfield, USA
| | - Mia Yasonova
- Internal Medicine, Kern Medical, Bakersfield, USA
| | | | | | | |
Collapse
|
7
|
León Fernández OS, Oru GT, Viebahn-Haensler R, López Cabreja G, Serrano Espinosa I, Corrales Vázquez ME. Medical Ozone Increases Methotrexate Effects in Rheumatoid Arthritis Through a Shared New Mechanism Which Involves Adenosine. Int J Mol Sci 2025; 26:5256. [PMID: 40508064 PMCID: PMC12155193 DOI: 10.3390/ijms26115256] [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: 04/08/2025] [Revised: 05/19/2025] [Accepted: 05/20/2025] [Indexed: 06/16/2025] Open
Abstract
Medical ozone is a redox regulator with beneficial effects in oxidative etiology diseases such as rheumatoid arthritis (RA). The aim of this study is to conduct a holistic review of different pharmacological trials involving ozone in model diseases, as well as the clinical responses of RA patients. The ROS (reactive oxygen species) involved in RA and their relationship with the main pathological pathways of this autoimmune disease are considered here. The integrative analysis of experimental results from animals with clinical findings reveals that both methotrexate (MTX) and medical ozone share common mechanisms via adenosinergic regulation. This finding enables us to propose a new pharmacological mechanism in the treatment of RA. We conclude that MTX + medical ozone combined therapy reduces ROS overproduction and the generation of proinflammatory cytokines and decreases anti-cyclic citrullinate peptide levels by a mutual mechanism involving adenosine A1 receptors.
Collapse
Affiliation(s)
| | - Gabriel Takon Oru
- Pharmacy and Food Institute, University of Havana, Havana 13603, Cuba;
| | | | - Gilberto López Cabreja
- National Institute of Rheumatology, Ministry of Public Health, Havana 19210, Cuba; (G.L.C.); (I.S.E.)
| | - Irainis Serrano Espinosa
- National Institute of Rheumatology, Ministry of Public Health, Havana 19210, Cuba; (G.L.C.); (I.S.E.)
| | - María Elena Corrales Vázquez
- Clinical Laboratory of the Surgical and Clinical Teaching Hospital “10 de Octubre”, Faculty of Medical Sciences, Ministry of Public Health, Havana 19210, Cuba;
| |
Collapse
|
8
|
He X, Li R, He X, He F, Xiao Y, Chen X, Lai Y, Guo W, Liu P, Cai X, Yang X, Shu Y, Shao J, Chen Y, Chen D, Quan M, Wu J, Xian L, He J, Ren H, Chen Z, Zhong Z, Liu L, Pan H. Elucidation of the anti-arthritic mechanism of Guizhi-Shaoyao-Zhimu decoction by reshaping circadian rhythm. Pharmacol Res 2025; 216:107787. [PMID: 40412782 DOI: 10.1016/j.phrs.2025.107787] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/27/2025] [Revised: 04/24/2025] [Accepted: 05/17/2025] [Indexed: 05/27/2025]
Abstract
Rheumatoid Arthritis (RA) exhibits pronounced circadian oscillations, with variations in joint stiffness, pain, and swelling following rhythmic patterns. Our preliminary research suggests that inflammatory cytokines in RA serum display rhythmicity closely associated with circadian rhythms. Additionally, circadian clock genes show a tendency towards disordered expression in RA, suggesting potential new therapeutic avenues from a circadian perspective. Traditional Chinese Medicine's Guizhi-Shaoyao-Zhimu Decoction (GSZD) has demonstrated clinical efficacy in RA, yet its underlying mechanism remains elusive. Our study confirms the effectiveness of GSZD in alleviating arthritis symptoms, reducing bone destruction, and lowering the expression of related inflammatory cytokines. Notably, we found that GSZD modulates the core circadian gene NR1D1 in a dose-dependent and time-selective manner. The administration of GSZD at 10 AM demonstrated enhanced anti-arthritic effects, which were mediated by regulating NR1D1 and suppressing the NLRP3-IL-18/IL-1β inflammatory signaling pathway. Moreover, silencing of NR1D1 significantly upregulated the expression of NLRP3, IL-18, and IL-1β. In NR1D1-deficient macrophages, GSZD failed to inhibit the NLRP3 inflammasome and inflammatory cytokines such as IL-18, and IL-1β. Overall, GSZD exerts therapeutic benefits for RA patients via activating NR1D1 and the interplay between molecular-based circadian rhythms and immune balance develop new avenues for targeted RA treatments, offering innovative strategies for the timing and administration of therapies.
Collapse
Affiliation(s)
- Xizi He
- State Key Laboratory of Traditional Chinese Medicine Syndrome, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510006, China; The Second Clinical Medical School, Guangzhou University of Chinese Medicine, Guangzhou 510006, China
| | - Runze Li
- State Key Laboratory of Traditional Chinese Medicine Syndrome, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510006, China; Chinese Medicine Guangdong Laboratory/ Hengqin Laboratory, Hengqin, Guangdong 519031, China
| | - Xiaohong He
- State Key Laboratory of Traditional Chinese Medicine Syndrome, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510006, China
| | - Fan He
- State Key Laboratory of Traditional Chinese Medicine Syndrome, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510006, China
| | - Yao Xiao
- State Key Laboratory of Traditional Chinese Medicine Syndrome, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510006, China
| | - Xianghong Chen
- The Second Clinical Medical School, Guangzhou University of Chinese Medicine, Guangzhou 510006, China
| | - Yu Lai
- State Key Laboratory of Traditional Chinese Medicine Syndrome, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510006, China; The Second Clinical Medical School, Guangzhou University of Chinese Medicine, Guangzhou 510006, China
| | - Wanyi Guo
- State Key Laboratory of Traditional Chinese Medicine Syndrome, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510006, China
| | - Peiyu Liu
- State Key Laboratory of Traditional Chinese Medicine Syndrome, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510006, China
| | - Xiao Cai
- The Second Clinical Medical School, Guangzhou University of Chinese Medicine, Guangzhou 510006, China
| | - Xiaonan Yang
- State Key Laboratory of Traditional Chinese Medicine Syndrome, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510006, China
| | - Yongkang Shu
- State Key Laboratory of Traditional Chinese Medicine Syndrome, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510006, China
| | - Jie Shao
- State Key Laboratory of Traditional Chinese Medicine Syndrome, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510006, China
| | - Yulian Chen
- State Key Laboratory of Traditional Chinese Medicine Syndrome, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510006, China
| | - Danli Chen
- State Key Laboratory of Traditional Chinese Medicine Syndrome, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510006, China
| | - Minqi Quan
- State Key Laboratory of Traditional Chinese Medicine Syndrome, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510006, China
| | - Jiaqi Wu
- State Key Laboratory of Traditional Chinese Medicine Syndrome, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510006, China
| | - Leyao Xian
- State Key Laboratory of Traditional Chinese Medicine Syndrome, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510006, China
| | - Jiayan He
- State Key Laboratory of Traditional Chinese Medicine Syndrome, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510006, China
| | - Hong Ren
- State Key Laboratory of Traditional Chinese Medicine Syndrome, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510006, China
| | - Zhengmin Chen
- State Key Laboratory of Traditional Chinese Medicine Syndrome, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510006, China
| | - Zishao Zhong
- State Key Laboratory of Traditional Chinese Medicine Syndrome, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510006, China
| | - Liang Liu
- State Key Laboratory of Traditional Chinese Medicine Syndrome, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510006, China; Chinese Medicine Guangdong Laboratory/ Hengqin Laboratory, Hengqin, Guangdong 519031, China.
| | - Hudan Pan
- State Key Laboratory of Traditional Chinese Medicine Syndrome, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510006, China; Chinese Medicine Guangdong Laboratory/ Hengqin Laboratory, Hengqin, Guangdong 519031, China.
| |
Collapse
|
9
|
Yang J, Zhang H, Wang W, Yin Q, He X, Tao D, Wang H, Liu W, Wang Y, Dong Z, Chen X, Li B. CD80 Antibody and MTX Co-Engineered Extracellular Vesicles Targets CD80 + Macrophages to Suppress Inflammation and Alleviate Chronic Inflammatory Diseases. Int J Nanomedicine 2025; 20:6379-6398. [PMID: 40416732 PMCID: PMC12103861 DOI: 10.2147/ijn.s517357] [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: 01/26/2025] [Accepted: 05/13/2025] [Indexed: 05/27/2025] Open
Abstract
Introduction Aberrant interaction between innate immune and adaptive immune cells can disrupt tissue homeostasis, consequently triggering chronic inflammatory diseases such as rheumatoid arthritis (RA) and periodontitis (PD). Pro-inflammatory macrophages serve as critical mediators in the early immune response, constituting a major population of CD80+ cells, while anti-inflammatory macrophages modulating inflammatory processes through the secretion of transforming growth factor-beta (TGF-β). This cytokine facilitates the differentiation of peripheral regulatory T cells (Tregs) and contributes to the establishment of immune tolerance. However, there are no definitive therapies to reshape the tissue homeostasis between innate immune and adaptive immune cells. Methods (1) anti-CD80-MTX-EVs was obtained by gradient centrifugation, which were characterized by TEM and DLS, and the associated membrane proteins were identified by Western Blot. (2) The mouse bone marrow-derived macrophages were co-cultured separately with EVs, anti-CD80-EVs, and anti-CD80-MTX-EVs in vitro, and the expression of CD80 on the macrophages surface as well as the proportion of Treg cell generation were detected. (3) EVs, anti-CD80-EVs and anti-CD80-MTX-EVs were injected into mice models of arthritis and periodontitis for treatment, the therapeutic effect was evaluated by the expressions of related cytokines, staining of HE, the proportion of CD80+ macrophages and the phenotypic differentiation of T cells in the tissues. Results We successfully constructed engineered EVs (anti-CD80-MTX-EVs) targeting inflammatory macrophages for intracellular MTX delivering, which inducing the anti-inflammatory transformation while upregulating the expression of TGF-β of macrophages. Furthermore, our findings demonstrate that anti-CD80-MTX-EVs effectively reduce CD80+ macrophage levels, promote Treg cell generation, and inhibit Th1 cell production in vivo. Conclusion In this study, the anti-CD80-MTX-Evs demonstrated significant therapeutic effects in both rheumatoid arthritis and periodontitis models through a triple mechanism: reducing CD80+ macrophage population, enhancing Treg cell differentiation, and suppressing Th1 cell development. Overall, this study presents an innovative strategy for resolving inflammation within chronic inflammatory diseases.
Collapse
Affiliation(s)
- Jianhua Yang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi International Joint Research Center for Oral Diseases, Center for Tissue Engineering, School of Stomatology, The Fourth Military Medical University, Xi’an, People’s Republic of China
| | - Handan Zhang
- Department of Chemical Engineering, Shaanxi Key Laboratory of Energy Chemical Process Intensification, Institute of Polymer Science in Chemical Engineering, School of Chemical Engineering and Technology, Xi’an Jiaotong University, Xi’an, 710049, People’s Republic of China
| | - Wenzhe Wang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi International Joint Research Center for Oral Diseases, Center for Tissue Engineering, School of Stomatology, The Fourth Military Medical University, Xi’an, People’s Republic of China
| | - Qiqi Yin
- Department of Chemical Engineering, Shaanxi Key Laboratory of Energy Chemical Process Intensification, Institute of Polymer Science in Chemical Engineering, School of Chemical Engineering and Technology, Xi’an Jiaotong University, Xi’an, 710049, People’s Republic of China
| | - Xiaoning He
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi International Joint Research Center for Oral Diseases, Center for Tissue Engineering, School of Stomatology, The Fourth Military Medical University, Xi’an, People’s Republic of China
| | - Dihao Tao
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi International Joint Research Center for Oral Diseases, Center for Tissue Engineering, School of Stomatology, The Fourth Military Medical University, Xi’an, People’s Republic of China
| | - Hanzhe Wang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi International Joint Research Center for Oral Diseases, Center for Tissue Engineering, School of Stomatology, The Fourth Military Medical University, Xi’an, People’s Republic of China
| | - Wenhao Liu
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi International Joint Research Center for Oral Diseases, Center for Tissue Engineering, School of Stomatology, The Fourth Military Medical University, Xi’an, People’s Republic of China
| | - Yiming Wang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi International Joint Research Center for Oral Diseases, Center for Tissue Engineering, School of Stomatology, The Fourth Military Medical University, Xi’an, People’s Republic of China
| | - Zhiwei Dong
- Department of Oral and Maxillofacial Surgery, College of Stomatology, Xi’an Jiaotong University, Xi’an, People’s Republic of China
| | - Xin Chen
- Department of Chemical Engineering, Shaanxi Key Laboratory of Energy Chemical Process Intensification, Institute of Polymer Science in Chemical Engineering, School of Chemical Engineering and Technology, Xi’an Jiaotong University, Xi’an, 710049, People’s Republic of China
| | - Bei Li
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi International Joint Research Center for Oral Diseases, Center for Tissue Engineering, School of Stomatology, The Fourth Military Medical University, Xi’an, People’s Republic of China
| |
Collapse
|
10
|
Eldadamony SM, El-Haggar SM, Ali AMA, Mostafa TM. Clinical study evaluating the efficacy and safety of Cilostazol as an adjuvant therapy to methotrexate on patients with rheumatoid arthritis. Inflammopharmacology 2025:10.1007/s10787-025-01782-2. [PMID: 40380042 DOI: 10.1007/s10787-025-01782-2] [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: 01/06/2025] [Accepted: 01/23/2025] [Indexed: 05/19/2025]
Abstract
OBJECTIVE This study aims to assess the safety as well as effectiveness of Cilostazol as a complementary therapy to methotrexate among individuals with active rheumatoid arthritis. METHOD This study was a randomly allocated, double-blind, placebo-controlled parallel design involving 70 patients who were diagnosed with active rheumatoid arthritis. Participants were randomly assigned to two sets: the control group (n = 35) which received methotrexate "MTX" (7.5 mg IM weekly) plus placebo tablets twice daily and the Cilostazol group (n = 35), which received the same MTX" dose plus Cilostazol 50 mg twice daily for 3 months. Patients were assessed to determine the serum levels of C-reactive protein (CRP) nuclear factor kappa B (NF-κB), hemoxygenase-1 (HO-1), and cyclic adenosine monophosphate (cAMP). Disease Activity Score (DAS28-CRP), Multidimensional Health Assessment Score (MDHAQ), and morning stiffness duration were also assessed. RESULTS The Cilostazol group produced a significant improvement in cAMP level as compared to the control group (P = 0.001). cAMP level showed a significant inverse correlation with DAS28-CRP (r = -0.336; P = 0.004). However, Cilostazol group produced non-significant improvements in the serum levels of the other biological markers (CRP, NF-κB, and HO-1), DAS28-CRP score, MDHAQ score, and morning stiffness duration as compared to the control group (P > 0.05). The implication of Cilostazol for patients with rheumatoid arthritis was tolerable and safe. CONCLUSION The beneficial effect of Cilostazol on cAMP and the negative correlation between cAMP and DAS28-CRP could support its impact on the disease activity. Further research seems necessary to elucidate the mechanisms underlying the link between cAMP and disease activity. TRIAL REGISTRATION Clinical Trials.gov identifier: NCT05594680, The date of registration is: 30/10/2022.
Collapse
Affiliation(s)
- Samar M Eldadamony
- Clinical Pharmacy Department, Faculty of Pharmacy, Tanta University, Tanta, Egypt.
| | - Sahar M El-Haggar
- Clinical Pharmacy Department, Faculty of Pharmacy, Tanta University, Tanta, Egypt
| | - Abdel Moaty A Ali
- Department of Rheumatology, Rehabilitation and Physical Medicine, Faculty of Medicine, Mansoura University, Mansoura, Egypt
| | - Tarek M Mostafa
- Clinical Pharmacy Department, Faculty of Pharmacy, Tanta University, Tanta, Egypt
| |
Collapse
|
11
|
Laudani C, Occhipinti G, Greco A, Giacoppo D, Spagnolo M, Capodanno D. A pairwise and network meta-analysis of anti-inflammatory strategies after myocardial infarction: the TITIAN study. EUROPEAN HEART JOURNAL. CARDIOVASCULAR PHARMACOTHERAPY 2025; 11:218-229. [PMID: 39756386 PMCID: PMC12046582 DOI: 10.1093/ehjcvp/pvae100] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/16/2024] [Revised: 10/18/2024] [Accepted: 01/02/2025] [Indexed: 01/07/2025]
Abstract
BACKGROUND AND AIMS Multiple anti-inflammatory drugs have been tested for secondary prevention after myocardial infarction (MI), giving mixed results and questioning the efficacy of anti-inflammatory therapy. No head-to-head comparisons between anti-inflammatory drugs have been performed. This study aimed to compare the efficacy and safety of anti-inflammatory drugs for secondary prevention after MI and the relative merits of specific drugs and administration strategies. METHODS AND RESULTS Randomized trials of anti-inflammatory therapy for secondary prevention after MI were identified. Primary efficacy and safety endpoints were trial-defined major adverse cardiovascular events (MACEs) and serious adverse events. Secondary endpoints included all-cause death, individual MACE components, serious infection, cancer, and gastrointestinal adverse events. Pairwise meta-analyses were conducted with interaction analyses for drug type and timing of administration, in addition to network meta-analyses. Multiple sensitivity and meta-regression analyses were conducted to explore potential heterogeneity sources. Twenty-eight studies, involving 44 406 patients with a mean follow-up of 11 months, were included. Anti-inflammatory therapy reduced the incidence of MACEs [incidence rate ratio (IRR): 0.92; 95% confidence interval (CI): 0.86-0.98] compared to control, without increasing serious adverse events. However, it was associated with a higher incidence of gastrointestinal adverse events (IRR: 1.21; 95% CI: 1.07-1.36). No significant interaction was observed between the effects of anti-inflammatory therapy on MACE and the timing of administration. CONCLUSION In secondary prevention for MI, anti-inflammatory therapy significantly reduces MACE without increasing serious adverse events, but it is associated with an increased risk of gastrointestinal adverse events.
Collapse
Affiliation(s)
- Claudio Laudani
- Division of Cardiology, Azienda Ospedaliero-Universitaria Policlinico “Rodolico—San Marco”, University of Catania, Via S. Sofia 76, 95125 Catania, Italy
| | - Giovanni Occhipinti
- Division of Cardiology, Azienda Ospedaliero-Universitaria Policlinico “Rodolico—San Marco”, University of Catania, Via S. Sofia 76, 95125 Catania, Italy
- Institut Clinic Cardiovascular, Hospital Clínic de Barcelona, University of Barcelona, Carrer de Villarroel, 170, L'Eixample, 08036 Barcelona, Spain
| | - Antonio Greco
- Division of Cardiology, Azienda Ospedaliero-Universitaria Policlinico “Rodolico—San Marco”, University of Catania, Via S. Sofia 76, 95125 Catania, Italy
| | - Daniele Giacoppo
- Division of Cardiology, Azienda Ospedaliero-Universitaria Policlinico “Rodolico—San Marco”, University of Catania, Via S. Sofia 76, 95125 Catania, Italy
| | - Marco Spagnolo
- Division of Cardiology, Azienda Ospedaliero-Universitaria Policlinico “Rodolico—San Marco”, University of Catania, Via S. Sofia 76, 95125 Catania, Italy
| | - Davide Capodanno
- Division of Cardiology, Azienda Ospedaliero-Universitaria Policlinico “Rodolico—San Marco”, University of Catania, Via S. Sofia 76, 95125 Catania, Italy
| |
Collapse
|
12
|
Iqbal U, Malik A, Sial NT, Mehmood MH, Uttra AM, Tulain UR, Erum A, Fayyaz-Ur-Rehman M, Welson NN, Mahmoud MH, Alexiou A, Papadakis M, El-Saber Bathia G. Eucalyptol attenuates indomethacin-induced gastric ulcers in rats by modulating the ICAM-1, eNOS and COX/LOX pathways: Insights from in silico, in vitro and in vivo approaches. Food Chem Toxicol 2025; 199:115319. [PMID: 39965739 DOI: 10.1016/j.fct.2025.115319] [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: 11/14/2024] [Revised: 01/07/2025] [Accepted: 02/10/2025] [Indexed: 02/20/2025]
Abstract
In order to evaluate anti-inflammatory role of eucalyptol (100, 200, and 400 mg/kg orally), inflammation was induced in rats using 0.1 ml of histamine and 0.1 ml of formaldehyde. Furthermore, in vivo gastroprotective potential of eucalyptol (100, 200 and 400 mg/kg) was determined via the intraperitoneal injection of 25 mg/kg indomethacin as an ulcerative agent and omeprazole (30 mg/kg) orally as a standard. Estimation of biochemical (PGE2, ICAM-1, COX-I, COX-II, eNOS and 5-LOX) and oxidative stress (SOD, CAT, GSH, and MDA) markers were carried out in gastric tissues using ELISA. The morphological and histopathological features of the gastric tissues were studied. In vitro, eucalyptol stabilized red blood cell membranes and inhibited protein denaturation, with the maximum effect observed at a concentration of 6400 μg/mL. Eucalyptol significantly reduced rat paw edema in histamine- and formaldehyde-induced inflammation models. It increased gastric PGE2, COX-I and eNOS levels, and decreased COX-II, 5-LOX and ICAM-1. Eucalyptol reduced ulcer indices and improved histopathological changes. Eucalyptol also increased antioxidants levels with decreased MDA levels in isolated rat stomach tissues. Therefore, eucalyptol shows gastroprotective effects against histamine- and formaldehyde induced inflammation and indomethacin-induced gastric ulcers through the modulation of the COX/LOX, ICAM-1, eNOS pathways and oxidative stress biomarkers.
Collapse
Affiliation(s)
- Urooj Iqbal
- Department of Pharmacology, College of Pharmacy, University of Sargodha, Pakistan; Primary and Secondary Health Care Department, Lahore, Punjab, Pakistan.
| | - Abdul Malik
- Department of Pharmacology, College of Pharmacy, University of Sargodha, Pakistan.
| | - Nabeela Tabassum Sial
- Department of Pharmacology, College of Pharmacy, University of Sargodha, Pakistan; Institute of Pharmacy, Lahore College for Women University, Lahore, Pakistan.
| | - Malik Hassan Mehmood
- Department of Pharmaceutical Sciences, Government College University Lahore, Pakistan.
| | - Ambreen Malik Uttra
- Department of Pharmacology, College of Pharmacy, University of Sargodha, Pakistan.
| | - Ume Ruqia Tulain
- Department of Pharmaceutics, College of Pharmacy, University of Sargodha, Pakistan.
| | - Alia Erum
- Department of Pharmaceutics, College of Pharmacy, University of Sargodha, Pakistan.
| | | | - Nermeen N Welson
- Department of Forensic Medicine and Clinical Toxicology, Faculty of Medicine, Beni-Suef University, 62511, Beni Suef, Egypt.
| | - Mohamed H Mahmoud
- Department of Biochemistry, College of Science, King Saud University, Kingdom of Saudi Arabia.
| | - Athanasios Alexiou
- University Centre for Research & Development, Chandigarh University, Chandigarh-Ludhiana Highway, Mohali, Punjab, India; Department of Research & Development, Funogen, Athens, 11741, Greece.
| | - Marios Papadakis
- University Hospital Witten-Herdecke, Heusnerstrasse 40, University of Witten-Herdecke, 42283, Wuppertal, Germany.
| | - Gaber El-Saber Bathia
- Department of Pharmacology and Therapeutics, Faculty of Veterinary Medicine, Damanhour University, AlBeheira, Damanhour, 22511, Egypt.
| |
Collapse
|
13
|
Wang C, Huang W, Chen Q, Yang C, Zhu H, Chen X, He Q, Yu X. Exploring the mechanism of Cynanchum paniculatum (Bunge) Kitag's therapeutic strategy for rheumatoid arthritis: integrating network pharmacology, molecular docking and in vivo experiments. J Biomol Struct Dyn 2025:1-15. [PMID: 40269643 DOI: 10.1080/07391102.2025.2494840] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2023] [Accepted: 04/04/2024] [Indexed: 04/25/2025]
Abstract
Rheumatoid arthritis (RA) is a chronic inflammatory disorder characterized by joint swelling, cartilage degradation, and joint deformity. The traditional Chinese herb Cynanchum paniculatum (Bunge) Kitag has been utilized in the management of RA, but the underlying mechanisms are unknown. This study utilized network pharmacology analysis to identify 26 active compounds associated with RA treatment and elucidate their interactions with 23 critical targets linked to RA. Subsequently, molecular docking studies revealed eight compounds with the capacity to bind to multiple key targets, with butyl isobutyl phthalate and geranyl acetone emerging as the most promising candidates based on their drug-likeness properties. To validate these findings, a rat model of adjuvant-induced arthritis was employed. Oral administration of geranyl acetone led to a significant reduction in paw swelling and pro-inflammatory markers, including TNF-α, IL-6, IL-1β, and MPO. Furthermore, it resulted in histological improvements in ankle tissues, all without adverse effects on weight or immune organs. Mechanistically, geranyl acetone was found to impede the progression of RA by modulating the TLR4/MyD88/NF-κB signaling pathway. In conclusion, C. paniculatum demonstrates substantial therapeutic potential for RA due to its multi-target and multi-pathway activities. Moreover, geranyl acetone, when used as a standalone agent, exhibits significant promise in alleviating RA symptoms, offering a compelling avenue for further research and potential clinical applications.
Collapse
Affiliation(s)
- Chen Wang
- Engineering Research Center of Active Substance and Biotechnology, Ministry of Education, College of Life Science, Chongqing Normal University, Chongqing, China
| | - Wangxiang Huang
- Engineering Research Center of Active Substance and Biotechnology, Ministry of Education, College of Life Science, Chongqing Normal University, Chongqing, China
| | - Qianzi Chen
- Engineering Research Center of Active Substance and Biotechnology, Ministry of Education, College of Life Science, Chongqing Normal University, Chongqing, China
| | - Chenying Yang
- Engineering Research Center of Active Substance and Biotechnology, Ministry of Education, College of Life Science, Chongqing Normal University, Chongqing, China
| | - Haiting Zhu
- Engineering Research Center of Active Substance and Biotechnology, Ministry of Education, College of Life Science, Chongqing Normal University, Chongqing, China
| | - Xiya Chen
- Engineering Research Center of Active Substance and Biotechnology, Ministry of Education, College of Life Science, Chongqing Normal University, Chongqing, China
| | - Qiyi He
- Engineering Research Center of Active Substance and Biotechnology, Ministry of Education, College of Life Science, Chongqing Normal University, Chongqing, China
| | - Xiaodong Yu
- Engineering Research Center of Active Substance and Biotechnology, Ministry of Education, College of Life Science, Chongqing Normal University, Chongqing, China
| |
Collapse
|
14
|
Zhang G, Wang R, Chen G, Liu S, Jie H, Chen W, Li Q. Hydroxychloroquine increases the exposure of methotrexate in plasma and red blood cells: a pharmacokinetic interaction study in rats in vivo. Front Pharmacol 2025; 16:1561001. [PMID: 40342994 PMCID: PMC12059348 DOI: 10.3389/fphar.2025.1561001] [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: 01/15/2025] [Accepted: 04/14/2025] [Indexed: 05/11/2025] Open
Abstract
Introduction Hydroxychloroquine (HCQ) has been demonstrated to be potential to enhance the therapeutic efficacy of methotrexate (MTX) for rheumatoid arthritis (RA) patients. However, the pharmacokinetics (PK) alterations and underlying mechanisms differentiating MTX-HCQ combination therapy from MTX monotherapy remain uncharted. Methods Thirty-three Sprague-Dawley rats were divided into single-dose and multiple-dose groups, with each group further randomized into an MTX monotherapy group an Hydroxychloroquine monotherapy group (HTG), and an MTX-HCQ combination therapy group Blood samples were collected at various time points before and after dosing to determine drug concentrations in plasma and red blood cells (RBC). The area under the concentration-time curve (AUC) for each compound was calculated, and pharmacokinetics models were established to analyze parameter variations across groups. Results In the single-dose group, the CTG exhibited a significant increase in the RBC MTX Cmax compared to the MTG (P = 0.023), whereas the AUC of RBC MTX showed an increasing trend (P = 0.056). In the multiple-dose group, the CTG demonstrated significant increases in plasma MTX Cmax and AUC (P = 0.023, P = 0.028, respectively) as well as RBC MTX Cmax and AUC (P = 0.010, P = 0.003, respectively). The RBC MTX polyglutamates (MTXPG2 and MTXPG3) also showed an increasing trend in Cmax and AUC for the CTG. Additionally, the CTG displayed a significant reduction in clearance rate (CLe) (P = 0.001). No significant differences were observed in the Cmax or AUC of HCQ or desethylhydroxychloroquine (DHCQ) in plasma or RBC across dosing groups. Conclusion These findings provide insights into the enhanced efficacy, faster onset, and prolonged effect of MTX-HCQ combination therapy compared to MTX monotherapy. The observed increases in MTX Cmax and AUC suggest the need for careful monitoring of MTX-related adverse effects, particularly in patients with renal insufficiency, during combination treatment with HCQ.
Collapse
Affiliation(s)
- Guijie Zhang
- Department of Pharmacy, The Third Affiliated Hospital of Southern Medical University, Guangzhou, Guangdong, China
| | - Rui Wang
- Department of Pharmacy, The Third Affiliated Hospital of Southern Medical University, Guangzhou, Guangdong, China
| | - Geping Chen
- Department of Pharmacy, The Third Affiliated Hospital of Southern Medical University, Guangzhou, Guangdong, China
| | - Simin Liu
- School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, Guangdong, China
| | - Hongyu Jie
- Department of Rheumatology and Immunology, The Third Affiliated Hospital of Southern Medical University, Guangzhou, Guangdong, China
| | - Wenying Chen
- Department of Pharmacy, The Third Affiliated Hospital of Southern Medical University, Guangzhou, Guangdong, China
| | - Qiang Li
- Department of Pharmacy, The Third Affiliated Hospital of Southern Medical University, Guangzhou, Guangdong, China
| |
Collapse
|
15
|
Jiang YP, Wen JJ, Ma X, Yuan CR, Zhou F, Zheng MJ, Tang X, Yu XK, Lai WD, Zhou YH, Yu WH, You WT, Jin Y, Park KD, Khanna R, Wen CP, Yu J. CRMP2 phosphorylation regulates polarization and spinal infiltration of CD4+ T lymphocytes, inhibits spinal glial activation, and arthritic pain. Pain 2025:00006396-990000000-00879. [PMID: 40258133 DOI: 10.1097/j.pain.0000000000003599] [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: 07/19/2024] [Accepted: 02/17/2025] [Indexed: 04/23/2025]
Abstract
ABSTRACT Chronic pain, a hallmark symptom of rheumatoid arthritis (RA), is strongly linked to central sensitization driven by spinal glial cell activation. Despite its clinical significance, the precise mechanisms remain unclear. Recent findings highlight the crucial role of interactions between circulating monocytes and central nervous system glial cells in chronic pain associated with autoimmune conditions. Our study focuses on CD4+ T-cell infiltration into the spinal dorsal horn (SDH) after collagen-induced arthritis (CIA) immunization. Immunohistochemistry results indicate that CD4+ T cells are critical in initiating arthritic pain. Intrathecal injection of CD4+ T cells in naïve mice induced glial activation and pain-like behaviors, while neutralizing antibodies suppressed these effects. Elevated phosphorylation of collapsin response mediator protein 2 (CRMP2) in CIA-derived CD4+ T lymphocytes was closely associated with pathological spinal infiltration. To modulate CRMP2 phosphorylation, we used naringenin (NAR), a known CRMP2 regulator, and (S)-Lacosamide ((S)-LCM), a specific inhibitor of phosphorylated CRMP2. Both compounds reduced CD4+ T-cell infiltration into the SDH and attenuated central sensitization in CIA rats. CRMP2 conditional knockout (cKO) in CD4+ T cells significantly alleviated arthritic pain. In addition, in vitro blood brain barrier models and Transwell assays showed impaired CD4+ T-cell migration and transendothelial invasion upon cKO or treatment with NAR and (S)-LCM. These interventions also decreased the proportion of polarized CD4+ T cells in CIA-induced mice. Our research highlights the role of CRMP2 phosphorylation in CD4+ T-cell behavior, spinal infiltration, and pain modulation, suggesting potential novel therapeutic strategies for RA-associated chronic pain.
Collapse
Affiliation(s)
- Yue-Peng Jiang
- College of Basic Medical Science, The Second Affiliated Hospital, Zhejiang Chinese Medical University, Key Laboratory of Neuropharmacology and Translational Medicine of Zhejiang Province, Innovative Research Center for Basic Medicine on Autoimmune Diseases of Ministry of Education, Hangzhou, China
| | - Jun-Jun Wen
- College of Basic Medical Science, The Second Affiliated Hospital, Zhejiang Chinese Medical University, Key Laboratory of Neuropharmacology and Translational Medicine of Zhejiang Province, Innovative Research Center for Basic Medicine on Autoimmune Diseases of Ministry of Education, Hangzhou, China
| | - Xiao Ma
- College of Basic Medical Science, The Second Affiliated Hospital, Zhejiang Chinese Medical University, Key Laboratory of Neuropharmacology and Translational Medicine of Zhejiang Province, Innovative Research Center for Basic Medicine on Autoimmune Diseases of Ministry of Education, Hangzhou, China
| | - Cun-Rui Yuan
- College of Basic Medical Science, The Second Affiliated Hospital, Zhejiang Chinese Medical University, Key Laboratory of Neuropharmacology and Translational Medicine of Zhejiang Province, Innovative Research Center for Basic Medicine on Autoimmune Diseases of Ministry of Education, Hangzhou, China
| | - Feng Zhou
- College of Basic Medical Science, The Second Affiliated Hospital, Zhejiang Chinese Medical University, Key Laboratory of Neuropharmacology and Translational Medicine of Zhejiang Province, Innovative Research Center for Basic Medicine on Autoimmune Diseases of Ministry of Education, Hangzhou, China
- Department of Neurosurgery, Hangzhou First People's Hospital Affiliated to Westlake University School of Medicine, Hangzhou, China
| | - Meng-Jia Zheng
- College of Basic Medical Science, The Second Affiliated Hospital, Zhejiang Chinese Medical University, Key Laboratory of Neuropharmacology and Translational Medicine of Zhejiang Province, Innovative Research Center for Basic Medicine on Autoimmune Diseases of Ministry of Education, Hangzhou, China
| | - Xin Tang
- College of Basic Medical Science, The Second Affiliated Hospital, Zhejiang Chinese Medical University, Key Laboratory of Neuropharmacology and Translational Medicine of Zhejiang Province, Innovative Research Center for Basic Medicine on Autoimmune Diseases of Ministry of Education, Hangzhou, China
| | - Xi-Kang Yu
- College of Basic Medical Science, The Second Affiliated Hospital, Zhejiang Chinese Medical University, Key Laboratory of Neuropharmacology and Translational Medicine of Zhejiang Province, Innovative Research Center for Basic Medicine on Autoimmune Diseases of Ministry of Education, Hangzhou, China
| | - Wei-Dong Lai
- College of Basic Medical Science, The Second Affiliated Hospital, Zhejiang Chinese Medical University, Key Laboratory of Neuropharmacology and Translational Medicine of Zhejiang Province, Innovative Research Center for Basic Medicine on Autoimmune Diseases of Ministry of Education, Hangzhou, China
| | - Yi-Han Zhou
- College of Basic Medical Science, The Second Affiliated Hospital, Zhejiang Chinese Medical University, Key Laboratory of Neuropharmacology and Translational Medicine of Zhejiang Province, Innovative Research Center for Basic Medicine on Autoimmune Diseases of Ministry of Education, Hangzhou, China
| | - Wen-Hua Yu
- Department of Neurosurgery, Hangzhou First People's Hospital Affiliated to Westlake University School of Medicine, Hangzhou, China
| | - Wen-Ting You
- The Affiliated Wenling Hospital of Wenzhou Medical University, Wenling, PR China
| | - Yan Jin
- College of Basic Medical Science, The Second Affiliated Hospital, Zhejiang Chinese Medical University, Key Laboratory of Neuropharmacology and Translational Medicine of Zhejiang Province, Innovative Research Center for Basic Medicine on Autoimmune Diseases of Ministry of Education, Hangzhou, China
| | - Ki Duk Park
- Korea Institute of Science and Technology, Seoul, South Korea
| | - Rajesh Khanna
- Department of Pharmacology & Therapeutics; Pain Research and Integrated Neuroscience Center (PRINC), University of Florida College of Medicine, Gainesville, FL, United States
| | - Cheng-Ping Wen
- College of Basic Medical Science, The Second Affiliated Hospital, Zhejiang Chinese Medical University, Key Laboratory of Neuropharmacology and Translational Medicine of Zhejiang Province, Innovative Research Center for Basic Medicine on Autoimmune Diseases of Ministry of Education, Hangzhou, China
| | - Jie Yu
- College of Basic Medical Science, The Second Affiliated Hospital, Zhejiang Chinese Medical University, Key Laboratory of Neuropharmacology and Translational Medicine of Zhejiang Province, Innovative Research Center for Basic Medicine on Autoimmune Diseases of Ministry of Education, Hangzhou, China
| |
Collapse
|
16
|
Kakkadath M, Naidu D, Kanthlal SK, Sharun K. Combating Methotrexate Resistance in Cancer Treatment: A Review on Navigating Pathways and Enhancing Its Efficacy With Fat-Soluble Vitamins. SCIENTIFICA 2025; 2025:8259470. [PMID: 40270992 PMCID: PMC12017957 DOI: 10.1155/sci5/8259470] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/17/2024] [Accepted: 03/24/2025] [Indexed: 04/25/2025]
Abstract
Methotrexate (MTX), a potent analogue and antagonist of folic acid, is a first-line treatment for rheumatoid arthritis, IBD and cancer. The development of MTX resistance contributes to the reduced efficacy and development of adverse reactions, forcing clinicians to withdraw treatment early. This drawback requires combinational approaches to combat the resistance and enhance the efficacy and safety of MTX. To provide a brief overview of MTX resistance and strategies to mitigate its aftereffects in cancer therapy, a literature-based search was conducted using keywords such as cancer pathology, MTX mechanism and resistance, S100A4, folate uptake, folate efflux, P-glycoprotein, beta-catenin and anticancer properties of Vitamins A, D, E and K. Investigations encompassing in vitro studies, in vivo studies and clinical trials were reviewed to identify the mechanisms of resistance induced by MTX and the potential benefits of coadministering fat-soluble vitamins with existing anticancer drugs. Derivates of Vitamin A could target cancer stem cells and increase chemotherapy sensitivity in non-small cell lung cancer. Similarly, calcitriol and cytotoxic medications exhibit additive or synergistic effects. Existing research revealed that fat-soluble vitamins can inhibit drug transporters, such as P-glycoprotein, which inhibit drug efflux, improving chemotherapy efficacy in cancer. As personalised medicine continues to evolve, incorporating combination approaches with MTX and fat-soluble vitamins holds promise for enhancing treatment efficacy, which can counteract MTX resistance via multiple pathways and improve the safety profile.
Collapse
Affiliation(s)
- Muhsina Kakkadath
- Department of Pharmacology, Amrita School of Pharmacy, Amrita Vishwa Vidyapeetham, Kochi 682041, Kerala, India
| | - Disha Naidu
- Department of Pharmacology, Amrita School of Pharmacy, Amrita Vishwa Vidyapeetham, Kochi 682041, Kerala, India
| | - S. K. Kanthlal
- Department of Pharmacology, Amrita School of Pharmacy, Amrita Vishwa Vidyapeetham, Kochi 682041, Kerala, India
| | - Khan Sharun
- Graduate Institute of Medicine, Yuan Ze University, Taoyuan 32003, Taiwan
| |
Collapse
|
17
|
Khoroshun K, Bantel C, Hoffmann F, Jobski K. Methotrexate-related drug reactions on kidneys and liver in rheumatoid arthritis: an analysis of spontaneous reports in EudraVigilance. Arthritis Res Ther 2025; 27:80. [PMID: 40188149 PMCID: PMC11972469 DOI: 10.1186/s13075-025-03551-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2024] [Accepted: 03/27/2025] [Indexed: 04/07/2025] Open
Abstract
OBJECTIVE Methotrexate (MTX), a standard treatment for rheumatoid arthritis (RA), is known for its potential kidney and liver toxicity. Whether concomitant use of analgesics, possibly affecting the same organs, has an impact on the occurrence or course of adverse drug reactions (ADRs) remains unclear. METHODS We used all spontaneous reports (until 2022) of suspected ADRs associated with MTX in RA patients, from the EudraVigilance database, a spontaneous report system operated by the European Medicines Agency (EMA). We displayed case and treatment characteristics, stratified by the organ affected (kidneys, liver) and the outcome (fatal, non-fatal). RESULTS We included a total of 10,319 reports (mean age: 62.3 years, 72.6% female). 365 and 1,082 were related to ADRs involving the kidneys and liver, respectively. Patients with ADRs on the kidneys were older and comedication (e.g. non-steroidal anti-inflammatory drugs (NSAIDs), acetaminophen, metamizole and corticosteroids) was more common than in cases with ADRs on the liver. More patients with kidney- than liver-related ADRs had a fatal outcome (21.1% vs. 5.8%). In fatal cases with ADRs on the kidneys and with ADRs on the liver comedication was more common compared to non-fatal cases. CONCLUSION Liver dysfunction was reported nearly three times more often than renal impairment. However, the kidneys need to be especially watched for, since a fatal outcome was considerably more common in renal failure. More precise and standardized recommendations on renal function tests might be necessary to support physicians in the complex treatment of RA.
Collapse
Affiliation(s)
- Kai Khoroshun
- Department of Health Services Research, Carl von Ossietzky Universität Oldenburg, 26111, Oldenburg, Germany.
| | - Carsten Bantel
- University Department of Anesthesiology, Critical Care, Emergency and Pain Medicine, Klinikum Oldenburg, Oldenburg, Germany
| | - Falk Hoffmann
- Department of Health Services Research, Carl von Ossietzky Universität Oldenburg, 26111, Oldenburg, Germany
| | - Kathrin Jobski
- Department of Health Services Research, Carl von Ossietzky Universität Oldenburg, 26111, Oldenburg, Germany
| |
Collapse
|
18
|
Wang X, Wu J, Ye H, Zhao X, Zhu S. Comprehensive Parent-Metabolite PBPK/PD Modeling Insights Into Methotrexate Personalized Dosing Strategies in Patients With Rheumatoid Arthritis. CPT Pharmacometrics Syst Pharmacol 2025; 14:695-705. [PMID: 39778004 PMCID: PMC12001262 DOI: 10.1002/psp4.13305] [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: 10/15/2024] [Revised: 12/01/2024] [Accepted: 12/19/2024] [Indexed: 01/11/2025] Open
Abstract
Rheumatoid arthritis (RA) is a major public health concern, which can cause serious outcomes. Low-dose methotrexate (MTX) is a cornerstone in RA treatment, but there is significant heterogeneity in clinical response. To evaluate underlying sources of pharmacokinetic variability and clinical response of MTX, a physiologically based pharmacokinetic and pharmacodynamic (PBPK/PD) model was developed using PK-sim and Mobi (version 11.1). The PBPK model included metabolism and transportation by AXO1, FPGS, GGH, RFC, and MRP2, with renal and biliary excretion. We also developed various degrees of renal insufficiency populations with subsequent dosing optimizations. A total of 23 MTX plasma concentration-time profiles were used, with 97% of predicted plasma concentrations within a two-fold range compared to observed data. The PBPK/PD modeling and simulation demonstrated that variability in renal clearance and enzymes related to MTX are likely important drivers of PK variability and there is a quantitative relationship between MTX-PG3 and RA treatment response. The PBPK/PD model could be used to guide improvement in MTX dose regimens for RA patients.
Collapse
Affiliation(s)
- Xin Wang
- Department of PharmacyThe First Affiliated Hospital of Chongqing Medical UniversityChongqingChina
| | - Jiangfan Wu
- School of PharmacyChongqing Medical UniversityChongqingChina
| | - Hongjiang Ye
- Department of NeurosurgeryThe First Affiliated Hospital of Chongqing Medical UniversityChongqingChina
| | - Xiaofang Zhao
- School of PharmacyChongqing Medical UniversityChongqingChina
| | - Shenyin Zhu
- Department of PharmacyThe First Affiliated Hospital of Chongqing Medical UniversityChongqingChina
| |
Collapse
|
19
|
Hameed H, Afzal M, Khan MA, Javaid L, Shahzad M, Abrar K. Unraveling the role of withanolides as key modulators in breast cancer mitigation. Mol Biol Rep 2025; 52:331. [PMID: 40117002 DOI: 10.1007/s11033-025-10442-1] [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] [Accepted: 03/14/2025] [Indexed: 03/23/2025]
Abstract
Addressing the elaborated landscape of therapeutics of global health concern i.e. breast cancer, this comprehensive review explores the promising effects of withanolides, bioactive compounds derived from Withania somnifera, for the treatment of breast cancer. In the breast, random mutations can accumulate over time, eventually transforming it into a tumor cell as certain receptors may be overexpressed by BC cells, which elicits downstream signaling and causes the production of genes involved in angiogenesis, survival, growth and migration, and other critical cell cycle practices. Merging insights from recent studies, our exploration delves into the molecular mechanisms that highlight withanolide's potential in the intervention of breast cancer. The study of apoptotic pathways unveils the withanolide's distinctive as well as pro-apoptotic effects, hinting at its effect as a potent modulator of the progression of breast cancer cells. Beyond its independent potential, there is a discussion on its distinctive perspective over the other therapies. Inweaving together these threads of evidence illuminates channels for future research. This review acts as a guide for researchers and clinicians negotiating the challenges of incorporating withanolides into the changing landscape for the treatment of breast cancer by balancing optimism with perceptive interpretation.
Collapse
Affiliation(s)
- Huma Hameed
- Faculty of Pharmaceutical Sciences, University of Central Punjab (UCP), Lahore, 54000, Pakistan.
| | - Maham Afzal
- Faculty of Pharmaceutical Sciences, University of Central Punjab (UCP), Lahore, 54000, Pakistan
| | - Mahtab Ahmad Khan
- Faculty of Pharmaceutical Sciences, University of Central Punjab (UCP), Lahore, 54000, Pakistan
| | - Laiba Javaid
- Faculty of Pharmaceutical Sciences, University of Central Punjab (UCP), Lahore, 54000, Pakistan
| | - Maria Shahzad
- Faculty of Pharmaceutical Sciences, University of Central Punjab (UCP), Lahore, 54000, Pakistan
| | - Kamran Abrar
- Faculty of Pharmaceutical Sciences, University of Central Punjab (UCP), Lahore, 54000, Pakistan
| |
Collapse
|
20
|
Bilski R, Nuszkiewicz J. Antioxidant Therapies as Emerging Adjuncts in Rheumatoid Arthritis: Targeting Oxidative Stress to Enhance Treatment Outcomes. Int J Mol Sci 2025; 26:2873. [PMID: 40243461 PMCID: PMC11989177 DOI: 10.3390/ijms26072873] [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: 03/03/2025] [Revised: 03/14/2025] [Accepted: 03/20/2025] [Indexed: 04/18/2025] Open
Abstract
Rheumatoid arthritis (RA) is a chronic autoimmune disorder characterized by persistent inflammation and progressive joint destruction. Recent data underscore oxidative stress as a primary factor in the pathophysiology of rheumatoid arthritis, intensifying inflammatory processes and tissue damage via the overproduction of reactive oxygen species (ROS) and compromised antioxidant defenses. Current therapies, including disease-modifying antirheumatic drugs (DMARDs), primarily target immune dysregulation but fail to address oxidative stress, necessitating novel adjunctive treatment strategies. This review explores the potential of antioxidant-based therapies as complementary approaches to RA management. Natural compounds such as curcumin, resveratrol, sulforaphane, and propolis exhibit strong anti-inflammatory and antioxidative properties by modulating redox-sensitive pathways, including nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) and nuclear factor erythroid 2-related factor 2(Nrf2)/heme oxygenase (HO-1). N-acetylcysteine (NAC) replenishes intracellular glutathione, enhancing cellular resilience against oxidative stress. Additionally, molecular hydrogen (H2) selectively neutralizes harmful ROS, reducing oxidative damage and inflammation. The role of vitamin supplementation (D, B12, C, and K) in regulating immune responses and protecting joint structures is also discussed. This review aims to evaluate the efficacy and potential clinical applications of antioxidant therapies in RA, emphasizing their role in mitigating oxidative damage and improving treatment outcomes. While preliminary findings are promising, further clinical trials are needed to establish standardized dosing, long-term safety, and their integration into current RA treatment protocols.
Collapse
Affiliation(s)
- Rafał Bilski
- Department of Medical Biology and Biochemistry, Faculty of Medicine, Ludwik Rydygier Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Toruń, 24 Karłowicza St., 85-092 Bydgoszcz, Poland
| | - Jarosław Nuszkiewicz
- Department of Medical Biology and Biochemistry, Faculty of Medicine, Ludwik Rydygier Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Toruń, 24 Karłowicza St., 85-092 Bydgoszcz, Poland
| |
Collapse
|
21
|
Zou W, Wang B, Feng X, Lai C, Tan Q. Ultra-High-Performance Liquid Chromatography-Tandem Mass Spectrometry Combined With Network Pharmacology to Elucidate the Bioactive Ingredients and Potential Mechanism of Wu-Teng Decoction for Treatment of Rheumatoid Arthritis. Chem Biodivers 2025:e202403454. [PMID: 40035706 DOI: 10.1002/cbdv.202403454] [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: 12/27/2024] [Revised: 02/18/2025] [Accepted: 03/04/2025] [Indexed: 03/06/2025]
Abstract
Wu-Teng Decoction (WTD) is a significant in-hospital preparation widely used in clinical practice to treat rheumatoid arthritis (RA) in China, however, its active substances and underlying mechanisms remain unclear. In this study, the chemical constituents of WTD were analyzed using ultra-high-performance liquid chromatography-tandem mass spectrometry, identifying a total of 120 compounds, including flavonoids, phenylpropanoids, phenolic acids, alkaloids, etc. Subsequently, network pharmacology analyses revealed that 29 compounds were potential active compounds in WTD for the treatment of RA, as well as 48 core anti-RA targets, including tumor necrosis factor-α, V-Akt murine thymoma viral oncogene homolog 1, and albumin. Further analysis suggested that WTD treats RA via the phosphoinositide 3-kinase-Akt, mitogen-activated protein kinase, and Ras signaling pathways. Molecular docking analysis of the top five pivotal targets with the core active ingredients demonstrated suitable binding interactions at the active site of target proteins. The significant reduction of nitric oxide levels in lipopolysaccharides-induced RAW264.7 cells validated the anti-inflammation activity of WTD.
Collapse
Affiliation(s)
- Wei Zou
- Department of Medicinal Chemistry, School of Pharmaceutical Sciences, Guilin Medical University, Guilin, China
| | - Bangyong Wang
- Department of Medicinal Chemistry, School of Pharmaceutical Sciences, Guilin Medical University, Guilin, China
| | - Xiaoqing Feng
- Department of Medicinal Chemistry, School of Pharmaceutical Sciences, Guilin Medical University, Guilin, China
| | - Chunhua Lai
- Department of Traditional Chinese Medicine, Affiliated Hospital of Guilin Medical University, Guilin, China
- Guangxi Key Laboratory of Drug Discovery and Optimization, Guilin Medical University, Guilin, China
| | - Qingang Tan
- Department of Medicinal Chemistry, School of Pharmaceutical Sciences, Guilin Medical University, Guilin, China
- Guangxi Key Laboratory of Drug Discovery and Optimization, Guilin Medical University, Guilin, China
| |
Collapse
|
22
|
Tekade M, Sharma MC. Quality-by-Design (QbD) Assisted Synthesis of Nanoparticle for Efficient Loading, Stabilization, and Intracellular Delivery of Bioactive for the Treatment of Arthritis. Indian J Microbiol 2025; 65:477-504. [PMID: 40371029 PMCID: PMC12069780 DOI: 10.1007/s12088-024-01338-y] [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: 05/15/2024] [Accepted: 06/12/2024] [Indexed: 05/16/2025] Open
Abstract
Rheumatoid arthritis (RA) is an autoimmune disorder that induces joint inflammation, cartilage injury, and bone damage. Thus far, methotrexate (MTX) is a primary DMARD drug to treat RA. Despite high efficiency, its clinical application is compromised due to delivery-associated systemic side effects. This investigation reports a Quality-by-design (QbD; Box Behnken Strategy) assisted production of a novel, innovative, and multipurpose polycation-templated approach for producing stable albumin MTX nanoparticles (pT-AMTX-NP). This approach formed a highly biocompatible MTX formulation with reduced toxicity (1.81 ± 0.54% hemolysis) compared to plain MTX (13.19 ± 2.77%; SEM:1.965). pT-AMTX-NP was found to be nanometric (Particle size: 135.86 ± 5.17 nm; PDI: 0.27) with a net surface charge of ζ -10.15 ± 2.19 mV. With 4.01-fold cationization (TNBS assay), pT-AMTX-NP showed high drug loading (64.98 ± 1.25%) and sustained MTX release under physiological conditions (up to 48 h; p < 0.001). The nanoformulation followed the Higuchi release kinetics model (R2, 0.9957). Significantly reduced burst release by 70.61% (p = 0.0002) and 12.89% (p = 0.0115) compared to plain MTX and conventional MTX-formulation (AMTX-NP), respectively. Interestingly, pT-AMTX-NP showed pH-responsive drug release bio-environment-responsive architectural change. Cell line studies in lipopolysaccharide (LPS) stimulated RAW264.7 macrophage showed a significant reduction in intracellular nitrite level following pT-AMTX-NP treatment (p < 0.01). Cellular uptake and cell viability confirmed selective cellular uptake potential in inflamed cells. Furthermore, compared to the control, the high intracellular ROS-generation was noted with pT-AMTX-NP (2.1485-fold; p < 0.01). Furthermore, hemolysis assay and stability assessments were also conducted to determine the hemocompatibility and suitable conditions for the storage of nanoformulation. The outcome of this study suggests that the developed multipurpose nanoformulation is a superior therapeutics approach for improved RA treatment. Suggestively, the developed strategy can also be adopted to benefit other clinical situations that demand to counter inflammation, cytostatic as well as psoriatic conditions.
Collapse
Affiliation(s)
- Muktika Tekade
- School of Pharmacy, Devi Ahilya Vishwavidyalaya, Takshila Campus, Khandwa Road, Indore, Madhya Pradesh India
| | - Mukesh Chandra Sharma
- School of Pharmacy, Devi Ahilya Vishwavidyalaya, Takshila Campus, Khandwa Road, Indore, Madhya Pradesh India
| |
Collapse
|
23
|
Zhang Y, Sun Y, Liao H, Shi S. Multifunctional DNA nanomaterials: a new frontier in rheumatoid arthritis diagnosis and treatment. NANOSCALE 2025; 17:4974-4999. [PMID: 39745199 DOI: 10.1039/d4nr04013a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/28/2025]
Abstract
Rheumatoid arthritis (RA) remains a challenging autoimmune disease due to its complex and heterogeneous pathophysiology, which complicates therapeutic and diagnostic efforts. Advances in DNA nanotechnology have introduced DNA nanomaterials as promising tools to overcome these barriers. This review focuses on three primary categories of DNA nanomaterials applied in RA: DNA nanostructures, DNA aptamers, and DNA-modified nanoparticles. DNA nanostructures, such as tetrahedral framework nucleic acids (tFNAs) and DNA origami, demonstrate anti-inflammatory properties and facilitate precise, controlled drug delivery. DNA aptamers, functioning as molecular recognition ligands, surpass traditional antibodies with their high specificity, low immunogenicity, and thermal stability, offering significant potential in biomarker detection and therapeutic interventions. While DNA-modified nanoparticles, which integrate DNA with materials like gold or lipids, have shown significant progress in bioimaging and drug delivery in other fields, their application in RA remains limited and warrants further exploration. Furthermore, advancements in stimulus-responsive systems are being explored to enable controlled drug release, which could significantly improve the specificity and efficiency of DNA nanomaterials in therapeutic applications. Despite their immense potential, challenges such as stability under physiological conditions, safety concerns, and clinical regulatory complexities persist. Overcoming these limitations is essential. This review highlights how DNA nanomaterials, beyond serving as delivery platforms, are poised to redefine RA treatment and diagnosis, opening the door to more personalized and effective approaches.
Collapse
Affiliation(s)
- Yiyi Zhang
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan, China.
- Sichuan Provincial Engineering Research Center of Oral Biomaterials, Chengdu, Sichuan 610041, China
| | - Yue Sun
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan, China.
- Sichuan Provincial Engineering Research Center of Oral Biomaterials, Chengdu, Sichuan 610041, China
| | - Hang Liao
- Department of Cardiology, West China Hospital, Sichuan University, Chengdu, Sichuan 610015, China
| | - Sirong Shi
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan, China.
- Sichuan Provincial Engineering Research Center of Oral Biomaterials, Chengdu, Sichuan 610041, China
| |
Collapse
|
24
|
Li Y, Cai M, Qin Y, Dai X, Liang L, Li Z, Wen X, Jin H, Yang C, Chen Z. MTHFD2 promotes osteoclastogenesis and bone loss in rheumatoid arthritis by enhancing CKMT1-mediated oxidative phosphorylation. BMC Med 2025; 23:124. [PMID: 40016725 PMCID: PMC11866863 DOI: 10.1186/s12916-025-03945-y] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/08/2024] [Accepted: 02/12/2025] [Indexed: 03/01/2025] Open
Abstract
BACKGROUND Rheumatoid arthritis (RA) is a chronic autoimmune disease characterized by disrupted bone homeostasis. This study investigated the effect and underlying mechanisms of one-carbon metabolism enzyme methylenetetrahydrofolate dehydrogenase 2 (MTHFD2) on osteoclast differentiation and bone loss in RA. METHODS The expression of MTHFD2 was examined in CD14 + monocytes and murine bone marrow-derived macrophages (BMMs). RNA-sequencing was performed to evaluate the regulatory mechanisms of MTHFD2 on osteoclastogenesis. Extracellular flux assay, JC-1 staining, and transmission electron microscopy were used to detect mitochondrial function and energy metabolism changes during osteoclast formation. Collagen-induced arthritis (CIA) mice were used to evaluate the therapeutic effect of MTHFD2 knockdown on bone loss. Bone volume and osteoclast counts were quantified by μCT and histomorphometry. RESULTS Elevated MTHFD2 was observed in RA patients and CIA mice with a positive correlation to bone resorption parameters. During osteoclast formation, MTHFD2 was significantly upregulated in both human CD14 + monocytes and murine BMMs. The application of MTHFD2 inhibitor and MTHFD2 knockdown suppressed osteoclastogenesis, while MTHFD2 overexpression promoted osteoclast differentiation in vitro. RNA-sequencing revealed that MTHFD2 inhibition blocked oxidative phosphorylation (OXPHOS) in osteoclasts, leading to decreased adenosine triphosphate (ATP) production and mitochondrial membrane potential without affecting mitochondrial biogenesis. Mechanistically, inhibition of MTHFD2 downregulated the expression of mitochondrial creatine kinase 1 (CKMT1), which in turn affected phosphocreatine energy shuttle and OXPHOS during osteoclastogenesis. Further, a therapeutic strategy to knock down MTHFD2 in knee joint in vivo ameliorated bone loss in CIA mice. CONCLUSIONS Our findings demonstrate that MTHFD2 is upregulated in RA with relation to joint destruction. MTHFD2 promotes osteoclast differentiation and arthritic bone erosion by enhancing mitochondrial energy metabolism through CKMT1. Thus, targeting MTHFD2 may provide a potential new therapeutic strategy for tackling osteoclastogenesis and bone loss in RA.
Collapse
Affiliation(s)
- Yujing Li
- Department of Rheumatology and Immunology, the First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230001, China
| | - Minglong Cai
- Department of Rheumatology and Immunology, the First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230001, China
| | - Yi Qin
- Department of Rheumatology and Immunology, the First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230001, China
- Department of Internal Medicine 3, Rheumatology and Immunology, Friedrich Alexander University Erlangen-Nuremberg and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Xiaojuan Dai
- Department of Rheumatology and Immunology, the First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230001, China
| | - Liyuan Liang
- The MED-X Institute, Center for Cancer Precision Medicine, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an Jiaotong University, Building 21, Western China Science and Technology Innovation Harbor, Xi'an, 710000, China
| | - Zhenyu Li
- Department of Rheumatology and Immunology, the First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230001, China
| | - Xi Wen
- Department of Rheumatology and Immunology, the First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230001, China
| | - Huizhi Jin
- Department of Rheumatology and Immunology, the First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230001, China
| | - Chao Yang
- Department of Rheumatology and Immunology and The MED-X institute, Center for Immunological and Metabolic Diseases, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an Jiaotong University, Building 21, Western China Science and Technology Innovation Harbor, Xi'an, 710000, China.
| | - Zhu Chen
- Department of Rheumatology and Immunology, the First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230001, China.
| |
Collapse
|
25
|
Mohaddes AA, Saatchi MA, Afshari Chamanabadi M, Saatchi S, Rostami S, Askari VR. Quantum Health Accelerator ® Ameliorates CFA-Induced Animal Model of Rheumatoid Arthritis: Investigating the Role of Immunomodulatory and Anti-Oxidative Effects. Brain Sci 2025; 15:232. [PMID: 40149754 PMCID: PMC11940038 DOI: 10.3390/brainsci15030232] [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: 02/11/2025] [Accepted: 02/19/2025] [Indexed: 03/29/2025] Open
Abstract
Introduction: Rheumatoid arthritis (RA) is a systemic inflammatory and autoimmune disease characterized by joint swelling, pain, damage to the cartilage, and disability. In the present study, we aimed to evaluate the anti-oxidant, anti-inflammatory, and immune-modulatory properties of Quantum Health Accelerator® as water enriched with vital bio-quantum information/energy (EW) following complete Freund's adjuvant (CFA)-induced RA in rats. Methods: Forty adult male Wistar rats (180-220 g) were divided into five groups. Arthritis was induced on day one using a single subcutaneous injection of CFA into the left hind footpad of the rat. Rats were assigned to receive methotrexate (MTX, 2 mg/kg/week, intraperitoneally), EW (orally, instead of normal water ad libitum), or their combination for 29 days. The anti-RA activities were determined by paw edema, joint diameter, arthritis score, and several nociceptive behavioral tests (thermal hyperalgesia, cold allodynia, and tactile allodynia). The levels of inflammatory (TNF-α, CRP, RF, and anti-CCP), anti-inflammatory (IL-10), and oxidative stress (NO, MDA, and GSH) markers were measured in serum. In addition, the levels of IFN-γ, IL-4, IL-17, and TGF-β were assessed in the spleen-isolated lymphocytes. Results: We found that treatment with MTX, EW, and their combination remarkably ameliorated thermal hyperalgesia, cold allodynia, and tactile allodynia results following CFA-induced RA in rats. In addition, EW also notably attenuated arthritis score, joint diameter, inflammatory cytokines, and oxidative markers while propagating anti-inflammatory and anti-oxidative mediators. Conclusions: We reveal that EW possesses anti-arthritic effects, possibly through anti-oxidative, anti-inflammatory, and immunomodulatory properties. Collectively, EW may be a promising therapeutic agent for treating RA.
Collapse
Affiliation(s)
- Ali Akbar Mohaddes
- International Group of Ali Akbar Mohaddes Company, License NO 1090645, Dubai 35360-97797, United Arab Emirates; (A.A.M.); (M.A.S.); (M.A.C.); (S.S.); (S.R.)
| | - Mohammad Ali Saatchi
- International Group of Ali Akbar Mohaddes Company, License NO 1090645, Dubai 35360-97797, United Arab Emirates; (A.A.M.); (M.A.S.); (M.A.C.); (S.S.); (S.R.)
| | - Marziyeh Afshari Chamanabadi
- International Group of Ali Akbar Mohaddes Company, License NO 1090645, Dubai 35360-97797, United Arab Emirates; (A.A.M.); (M.A.S.); (M.A.C.); (S.S.); (S.R.)
| | - Saeed Saatchi
- International Group of Ali Akbar Mohaddes Company, License NO 1090645, Dubai 35360-97797, United Arab Emirates; (A.A.M.); (M.A.S.); (M.A.C.); (S.S.); (S.R.)
| | - Sadra Rostami
- International Group of Ali Akbar Mohaddes Company, License NO 1090645, Dubai 35360-97797, United Arab Emirates; (A.A.M.); (M.A.S.); (M.A.C.); (S.S.); (S.R.)
| | - Vahid Reza Askari
- Pharmacological Research Center of Medicinal Plants, Mashhad University of Medical Sciences, Mashhad 9177948564, Iran
| |
Collapse
|
26
|
Chowdary P, Puppala ER, Putta CL, Maddila JR, Pulavarthy V, Prasad VVSR, Rengan AK. Hyaluronic-Acid-Functionalized Tofacitinib Loaded Transethosomes for Targeted Drug Delivery in Rheumatoid Arthritis. ACS APPLIED BIO MATERIALS 2025; 8:1594-1606. [PMID: 39876608 DOI: 10.1021/acsabm.4c01743] [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: 01/30/2025]
Abstract
The Janus kinase inhibitor tofacitinib (TOF) is an FDA-approved drug for rheumatoid arthritis (RA) treatment, but its long-term oral use leads to significant systemic side effects. The present research aimed to conquer these challenges by formulating hyaluronic-acid-coated transethosomes (HA-TOF-TE), a novel system for targeted, topical delivery of TOF to reduce systemic toxicity and improve therapeutic efficacy. Transethosomes were synthesized via the cold sonication technique with HA functionalization enabling CD44 receptor-mediated targeting of inflamed synovial tissue. Optimized TOF-TE and HA-TOF-TE formulations showed particle sizes of 199.08 ± 4.2 and 151.5 ± 5.4 nm, zeta potentials of -27.1 ± 0.75 and -34.10 ± 0.89 mV, and entrapment efficiencies of 81.16 ± 0.84% and 79.19 ± 2.65%, respectively. The gels were assessed through in vitro drug release, ex vivo permeability, and in vivo effectiveness experiments using Freund's complete adjuvant (CFA) model. Ex vivo studies showed 2.02-fold and 1.61-fold increments in flux for TOF-TE and HA-TOF-TE, respectively, with superior skin retention for HA-TOF-TE. In vivo efficacy confirmed HA-TOF-TE's significant (P < 0.001) anti-inflammatory effect on arthritic rat paws, outperforming TOF-TE and FD gels. Cytokine analysis showed notable reductions in serum IL-1, IL-6, and PGE-2 levels after HA-TOF-TE treatment, closely approximating control values. Additionally, mRNA analysis demonstrated marked decreases in IL-6, CD44, and collagen II expression, indicating HA-TOF-TE's potential as an effective, targeted RA treatment, addressing the challenges of conventional TOF therapy and minimizing systemic side effects.
Collapse
Affiliation(s)
- Pavani Chowdary
- Department of Biomedical Engineering, Indian Institute of Technology Hyderabad, Kandi 502284, Telangana, India
- Department of Pharmaceutical Engineering, B V Raju Institute of Technology, Narsapur, Medak 502313, Telangana, India
| | - Eswara Rao Puppala
- Marlene and Stewart Greenebaum Comprehensive Cancer Centre, University of Maryland Baltimore School of Medicine, Baltimore, Maryland 21201, United States
| | - Chandra Lekha Putta
- Department of Biomedical Engineering, Indian Institute of Technology Hyderabad, Kandi 502284, Telangana, India
| | - Jagapathi Rao Maddila
- Department of Biomedical Engineering, Indian Institute of Technology Hyderabad, Kandi 502284, Telangana, India
| | - Vishnu Pulavarthy
- Department of Pharmaceutical Engineering, B V Raju Institute of Technology, Narsapur, Medak 502313, Telangana, India
| | - V V S Rajendra Prasad
- Centre for Molecular Cancer Research, Department of Pharmaceutical Chemistry and Analysis, Vishnu Institute of Pharmaceutical Education and Research, Narsapur, Medak 502313, Telangana, India
| | - Aravind Kumar Rengan
- Department of Biomedical Engineering, Indian Institute of Technology Hyderabad, Kandi 502284, Telangana, India
| |
Collapse
|
27
|
Mahmoud AM, Alfadl EMA, Ahmed ARH, Abouelella AMA, Alshazly O, Mohamed MFA, Allaf HE, Allam RM. Disclosing the impact of metformin and methotrexate in adjuvant arthritis in female rats: molecular docking and biochemical insights on visfatin. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025:10.1007/s00210-025-03823-7. [PMID: 39878818 DOI: 10.1007/s00210-025-03823-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/26/2024] [Accepted: 01/15/2025] [Indexed: 01/31/2025]
Abstract
Rheumatoid arthritis (RA) is one of the most common systemic autoimmune inflammatory diseases, with a progressive etiology that results in serious complications and a higher chance of early death. Visfatin, an adipokine, is correlated with disease pathologic features and becomes a key biomarker and therapeutic target for RA. This study aimed to evaluate the anti-arthritic activity of metformin (an antidiabetic drug with anti-inflammatory activities) and methotrexate (the first choice for disease-modifying antirheumatic drugs in RA, with diverse adverse effects) in complete Freund's adjuvant (CFA)-induced arthritis in female rats. Treatment outcomes were assessed using arthritis severity, serum levels of inflammatory markers, and pro-inflammatory adipokine (visfatin). In addition to radiological and histopathological examination, and docking analysis. Results showed that Met, MTX, and Met/MTX significantly (p ≤ 0.05) lowered paw swelling and arthritic score, as well as attenuated serum levels of rheumatoid factor (RF), C-reactive protein (CRP), and visfatin. The combined treatment gives the best results. The previously mentioned findings were further confirmed through radiological and histopathological examinations. In conclusion, the co-administration of metformin could potentiate the anti-arthritic activity of methotrexate, providing a medical strategy for arthritis management.
Collapse
Affiliation(s)
- Ahmed Mostafa Mahmoud
- Department of Physiology, Faculty of Medicine, Sohag University, Sohag, Egypt
- Department of Basic Medical Sciences, Aqaba Medical Sciences University, Aqaba, Jordan
| | - Esam Mohamed Abu Alfadl
- Department of Rheumatology and Rehabilitation, Faculty of Medicine, Sohag University, Sohag, Egypt
| | - Ahmed R H Ahmed
- Department of Pathology, Faculty of Medicine, Sohag University, Sohag, Egypt
| | - Azza M A Abouelella
- Department of Pharmacology, Faculty of Medicine, Sohag University, Sohag, Egypt
| | - Omar Alshazly
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Sohag University, Sohag, 82524, Egypt.
| | - Mamdouh F A Mohamed
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Sohag University, Sohag, 82524, Egypt
| | - Hasan El Allaf
- Department of Physiology, Faculty of Medicine, Sohag University, Sohag, Egypt
- Department of Basic Medical Sciences, Aqaba Medical Sciences University, Aqaba, Jordan
| | - Rasha M Allam
- Pharmacology Department, Medical and Clinical Research Institute, National Research Centre, Dokki, Cairo, 12622, Egypt.
| |
Collapse
|
28
|
Ye Y, Liang Y, Huang L, Cao X, Xia Z, Liang S. Daptomycin alleviates collagen-induced arthritis via suppressing inflammatory cytokines and NF-κB pathway. Int Immunopharmacol 2025; 144:113648. [PMID: 39571268 DOI: 10.1016/j.intimp.2024.113648] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2024] [Revised: 11/12/2024] [Accepted: 11/14/2024] [Indexed: 12/15/2024]
Abstract
Rheumatoid arthritis (RA) is a chronic autoimmune disease, and its pathogenic factors include bacterial infection and immune inflammation. Daptomycin (DAP) is a cyclic lipopeptide that has been approved to treat skin infections, bacteremia and right-sided endocarditis due to its effective antibacterial effects against most Gram-positive pathogenic bacteria. Herein, we focus on whether DAP exerts anti-inflammatory activity on RA to expand DAP drug indications. Our studies are first time to evaluate anti-inflammatory effect and reveal its molecular pharmacological mechanism of DAP on lipopolysaccharide-activated macrophage and collagen-induced arthritis (CIA) mice. In vitro cytological studies show that 10 µg/mL DAP inhibits secretion of IL-1β, IL-6 and TNF-α to alleviate cell inflammation, and the phosphorylation level of p65 (p-p65) of RAW 264.7 cells is decreased under DAP exposure. Administration with 10 mg/kg/2d DAP via intravenous injection on CIA mice significantly reduces arthritis symptom by inhibiting p-p65 level and decreasing serum level of IL-6, IL-1β and TNF-α. Moreover, our study is the first time to reveal new immunomodulatory effects of DAP on RA, which provides a pharmacological basis for new clinical applications of DAP for other immune-related diseases except to bacterial infections.
Collapse
Affiliation(s)
- Yang Ye
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, No.17, 3rd Section of People's South Road, Chengdu, 610041, PR China
| | - Yue Liang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, No.17, 3rd Section of People's South Road, Chengdu, 610041, PR China
| | - Lan Huang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, No.17, 3rd Section of People's South Road, Chengdu, 610041, PR China
| | - Xu Cao
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, No.17, 3rd Section of People's South Road, Chengdu, 610041, PR China
| | - Zijing Xia
- Department of Rheumatology, West China Hospital, Sichuan University, Chengdu 610041, PR China
| | - Shufang Liang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, No.17, 3rd Section of People's South Road, Chengdu, 610041, PR China.
| |
Collapse
|
29
|
Yielder R, Leibowitz K, Crum AJ, Manley P, Dalbeth N, Petrie KJ. Changing mindsets about methotrexate in the rheumatology clinic to reduce side effects and improve adherence: a randomized controlled trial. Ann Behav Med 2025; 59:kaae089. [PMID: 39715363 PMCID: PMC11761654 DOI: 10.1093/abm/kaae089] [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] [Indexed: 12/25/2024] Open
Abstract
BACKGROUND Patients' negative expectations about medication can exacerbate side effect burden leading to low adherence and persistence. A novel intervention involves targeting mindsets about non-severe symptoms; reframing them as encouraging signs of medication working. PURPOSE This study aimed to assess whether a brief symptom-mindset intervention can improve symptom experience and adherence in patients starting methotrexate to treat an inflammatory rheumatic disease. METHODS A randomized controlled trial was conducted with patients starting methotrexate. Participants were randomly assigned (1:1) to a mindset intervention or standard information control condition. Symptom mindset was assessed after 4 weeks to check intervention efficacy. The primary outcome was symptom experience after 4 weeks. Secondary outcomes were adherence and motivation to take methotrexate (4 weeks), as well as continuation and C-reactive protein (12 weeks). RESULTS Forty-seven participants were randomly assigned to the intervention (n = 24) or control group (n = 23). All participants completed the study. After 4 weeks, compared to the control group, intervention participants endorsed more positive symptom mindsets, experienced less symptom burden (mean difference -2.70 [95% CI, -4.50, -0.90] P = .005), fewer general symptoms (3.53 [-6.99, .79] P = .045) and a similar number of methotrexate-specific symptoms (-0.79 [-2.29, 0.71] P = .295). The intervention group had better motivation and adherence to methotrexate at 4 weeks and better continuation, and C-reactive protein at 12 weeks than the control group. There was no difference in side effect attribution. CONCLUSIONS In patients starting methotrexate, a mindset intervention reframing the role of non-severe side effects is a promising approach for improving symptom experience and early stage medication persistence.
Collapse
Affiliation(s)
- Rachael Yielder
- Department of Psychological Medicine, Faculty of Medical and Health Sciences, University of Auckland, Auckland 1142, New Zealand
| | - Kari Leibowitz
- Department of Psychology, Stanford University, Stanford, CA 94305, United States
| | - Alia J Crum
- Department of Psychology, Stanford University, Stanford, CA 94305, United States
| | - Paul Manley
- Te Whatu Ora Health, Te Toka Tumai Auckland, Auckland 1023, New Zealand
| | - Nicola Dalbeth
- Department of Medicine, Faculty of Medical and Health Sciences, University of Auckland, Auckland 1142, New Zealand
| | - Keith J Petrie
- Department of Psychological Medicine, Faculty of Medical and Health Sciences, University of Auckland, Auckland 1142, New Zealand
| |
Collapse
|
30
|
Kakkadath M, Naidu D, Kanthlal SK, Sharun K. Combating Methotrexate Resistance in Cancer Treatment: A Review on Navigating Pathways and Enhancing Its Efficacy With Fat‐Soluble Vitamins. SCIENTIFICA 2025; 2025. [DOI: https:/doi.org/10.1155/sci5/8259470] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/17/2024] [Accepted: 03/24/2025] [Indexed: 05/13/2025]
Abstract
Methotrexate (MTX), a potent analogue and antagonist of folic acid, is a first‐line treatment for rheumatoid arthritis, IBD and cancer. The development of MTX resistance contributes to the reduced efficacy and development of adverse reactions, forcing clinicians to withdraw treatment early. This drawback requires combinational approaches to combat the resistance and enhance the efficacy and safety of MTX. To provide a brief overview of MTX resistance and strategies to mitigate its aftereffects in cancer therapy, a literature‐based search was conducted using keywords such as cancer pathology, MTX mechanism and resistance, S100A4, folate uptake, folate efflux, P‐glycoprotein, beta‐catenin and anticancer properties of Vitamins A, D, E and K. Investigations encompassing in vitro studies, in vivo studies and clinical trials were reviewed to identify the mechanisms of resistance induced by MTX and the potential benefits of coadministering fat‐soluble vitamins with existing anticancer drugs. Derivates of Vitamin A could target cancer stem cells and increase chemotherapy sensitivity in non–small cell lung cancer. Similarly, calcitriol and cytotoxic medications exhibit additive or synergistic effects. Existing research revealed that fat‐soluble vitamins can inhibit drug transporters, such as P‐glycoprotein, which inhibit drug efflux, improving chemotherapy efficacy in cancer. As personalised medicine continues to evolve, incorporating combination approaches with MTX and fat‐soluble vitamins holds promise for enhancing treatment efficacy, which can counteract MTX resistance via multiple pathways and improve the safety profile.
Collapse
|
31
|
Pimple P, Shah J, Singh P. Emerging Phytochemical Formulations for Management of Rheumatoid Arthritis: A Review. Curr Drug Deliv 2025; 22:15-40. [PMID: 38299275 DOI: 10.2174/0115672018270434240105110330] [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: 07/11/2023] [Revised: 12/02/2023] [Accepted: 12/17/2023] [Indexed: 02/02/2024]
Abstract
Rheumatoid arthritis (RA) is a T-cell-mediated chronic inflammatory disorder affecting 0.5-1% of the global population. The disease with unknown etiology causes slow destruction of joints, advancing to significant deterioration of an individual's quality of life. The present treatment strategy comprises the use of disease-modifying anti-rheumatic drugs (DMARDs) coupled with or without nonsteroidal anti-inflammatory drugs or glucocorticoids. Additionally, involves co-therapy of injectable biological DMARDs in case of persistent or recurrent arthritis. The availability of biological DMARDs and the implementation of the treat-to-target approach have significantly improved the outcomes for patients suffering from RA. Nevertheless, RA requires continuous attention due to inadequate response of patients, development of tolerance and severe side effects associated with long-term use of available treatment regimens. An estimated 60-90% of patients use alternative methods of treatment, such as herbal therapies, for the management of RA symptoms. Over the past few decades, researchers have exploring natural phytochemicals to alleviate RA and associated symptoms. Enormous plant-origin phytochemicals such as alkaloids, flavonoids, steroids, terpenoids and polyphenols have shown anti-inflammatory and immunomodulatory activity against RA. However, phytochemicals have certain limitations, such as high molecular weight, poor water solubility, poor permeability, poor stability and extensive first-pass metabolism, limiting absorption and bioavailability. The use of nanotechnology has aided to extensively improve the pharmacokinetic profile and stability of encapsulated drugs. The current review provides detailed information on the therapeutic potential of phytochemicals. Furthermore, the review focuses on developed phytochemical formulations for RA, with emphasis on clinical trials, regulatory aspects, present challenges, and future prospects.
Collapse
Affiliation(s)
- Prachi Pimple
- Department of Pharmaceutics, SVKM's Dr. Bhanuben Nanavati College of Pharmacy, Vile Parle, Mumbai 400056, India
| | - Jenny Shah
- Department of Pharmaceutics, SVKM's Dr. Bhanuben Nanavati College of Pharmacy, Vile Parle, Mumbai 400056, India
| | - Prabha Singh
- Department of Pharmaceutics, SVKM's Dr. Bhanuben Nanavati College of Pharmacy, Vile Parle, Mumbai 400056, India
| |
Collapse
|
32
|
Baek IW, Park KS, Kim KJ. An erythrocyte macrocytosis by methotrexate is associated with early initiation of biologic or targeted synthetic agents in patients with rheumatoid arthritis. JOURNAL OF RHEUMATIC DISEASES 2025; 32:30-37. [PMID: 39712246 PMCID: PMC11659665 DOI: 10.4078/jrd.2024.0073] [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: 06/19/2024] [Revised: 08/18/2024] [Accepted: 08/19/2024] [Indexed: 12/24/2024]
Abstract
Objective An association between increased erythrocyte mean corpuscular volume (MCV) and treatment response in patients with inflammatory arthritis receiving methotrexate (MTX) has been reported. We investigated the frequency of red blood cell (RBC) macrocytosis and its clinical implications regarding the initiation of biological or targeted synthetic disease-modifying anti-rheumatic drugs (b/tsDMARDs) in patients starting MTX for rheumatoid arthritis (RA). Methods RBC macrocytosis (MCV >100 fL) and clinical characteristics were retrospectively examined in 1,156 patients starting MTX for RA. Multivariable logistic regression analyses were performed to identify the independent predictors of RBC macrocytosis. The initiation of b/tsDMARDs was assessed using a multivariable Cox proportional hazards regression model. Results RBC macrocytosis was observed in 21.6% of RA patients over 35 [8, 89] months following MTX initiation and was persistent in 63.6% of the patients during MTX treatment. Anemia coexisted in only 20.0% of the patients with RBC macrocytosis. The occurrence of RBC macrocytosis was independently associated with age, MTX dose, and concomitant use of sulfasalazine or leflunomide (all p<0.001). A higher dose of MTX and double- or triple-DMARDs therapy were more frequently used in the group with RBC macrocytosis than in the group with normal MCV. Patients experiencing RBC macrocytosis were more likely to use b/tsDMARDs (hazard ratio 1.45 [95% confidence interval 1.13, 1.87], p=0.003). Conclusion RBC macrocytosis was possibly associated with the use of b/tsDMARD and could be a supplementary marker for assessing MTX resistance.
Collapse
Affiliation(s)
- In-Woon Baek
- Division of Rheumatology, Department of Internal Medicine, Ewha Womans University College of Medicine, Seoul, Korea
| | - Kyung-Su Park
- St. Vincent’s Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Ki-Jo Kim
- St. Vincent’s Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| |
Collapse
|
33
|
Tezcan D, Özer H, Topaloğlu ÖF, Hakbilen S, Durmaz MS, Yılmaz S, Öztürk M. Evaluation of liver parenchyma with shear wave elastography in patients with rheumatoid arthritis receiving disease-modifying antirheumatic drug therapy. JOURNAL OF CLINICAL ULTRASOUND : JCU 2025; 53:148-154. [PMID: 39304522 DOI: 10.1002/jcu.23847] [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: 08/16/2024] [Revised: 09/07/2024] [Accepted: 09/11/2024] [Indexed: 09/22/2024]
Abstract
BACKGROUND Methotrexate (MTX) and leflunomide (LEF) play fundamental roles in rheumatoid arthritis (RA) treatment and require proper monitoring of side effects. Concerns about MTX/LEF-related liver fibrosis (LF) in patients with RA remain unclear. This study investigated liver stiffness using two-dimensional shear wave elastography (2D-SWE) in RA patients undergoing disease-modifying antirheumatic drug (DMARD) therapy. Moreover, 2D-SWE was employed to evaluate the correlations between liver stiffness, cumulative MTX and LEF doses and risk factors for substantial LF. METHODS We recruited 222 participants from the Department of Rheumatology. The participants were divided into healthy controls (n = 78) and patients with RA (n = 144). Pearson's correlation analysis was performed to assess the correlations between liver stiffness and the cumulative dose of MTX/LEF and other clinical and laboratory variables. RESULTS The mean elasticity modulus was 4.79 ± 0.92 kPa, excluding the presence of significant fibrosis. Mean 2D-SWE values were significantly lower in healthy controls than in RA treated with MTX and LEF. The cut-off ≥3.8 kPa 2D-SWE values with the sensitivity of 86.1%, specifity of 83.3%. 2D-SWE values were not significantly different across the strata of the cumulative MTX subgroups. CONCLUSIONS MTX and LEF increase liver stiffness but may be considered low risk for the development of LF.
Collapse
Affiliation(s)
- Dilek Tezcan
- Department of Internal Medicine, Division of Rheumatology, Gülhane Faculty of Medicine, University of Health Sciences Turkey, Ankara, Turkey
| | - Halil Özer
- Division of Radiology, Selcuk University Faculty of Medicine, Konya, Turkey
| | | | - Selda Hakbilen
- Division of Rheumatology, Selcuk University Faculty of Medicine, Konya, Turkey
| | | | - Sema Yılmaz
- Division of Rheumatology, Selcuk University Faculty of Medicine, Konya, Turkey
| | - Mehmet Öztürk
- Division of Radiology, Selcuk University Faculty of Medicine, Konya, Turkey
| |
Collapse
|
34
|
Ammon T, Zeiträg J, Mayr V, Benedicic M, Holthoff H, Ungerer M. Citrullinated Autoantigen-Specific T and B Lymphocytes in Rheumatoid Arthritis: Focus on Follicular T Helper Cells and Expansion by Coculture. ACR Open Rheumatol 2025; 7:e11785. [PMID: 39846262 PMCID: PMC11755120 DOI: 10.1002/acr2.11785] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2024] [Revised: 11/12/2024] [Accepted: 12/03/2024] [Indexed: 01/24/2025] Open
Abstract
OBJECTIVE Rheumatoid arthritis (RA) is characterized by circulating anti-cyclic citrullinated peptide (CCP) autoantibodies (ACPAs), resulting in inflammation of the joints and other organs. We have established novel assays to assess immune cell subpopulations, including citrullinated antigen-specific (CAS) autoreactive B and T lymphocytes, in patients with RA. METHODS AND RESULTS We found that activated CD25+ T cells were markedly increased in patients with RA compared to healthy controls. Novel combinations of major histocompatibility complex class II citrulline epitope tetramers were developed, which enabled robust detection of CAS T cells and showed increases of CAS-naive T helper cells, Th1.17 cells, CAS total circulating T follicular helper (cTfh) cells, and cTfh1 cells in ACPA+ patients with RA. In addition, an innovative assay using dual labeling with CCP-biotin probes allowed for reproducible identification of primary CAS B cells after enrichment with advantages over existing detection methods. Furthermore, patient-derived immune cells were successfully expanded. Primary RA B cells were successfully cultured on novel feeder cell lines, whereas T cells were expanded ex vivo in the presence of interleukin-2 and citrullinated peptides, and subsequent alterations in cell frequencies were assessed. CONCLUSION Novel assays were established to reliably detect CAS T and B cells in patients with RA, and specific CAS-naive T helper cells, Th1.17 cells, cTfh cells, and cTfh1 cells were observed more frequently in RA. Based on these results, new coculture systems of disease-relevant cells are developed to simulate human secondary lymphoid tissues ex vivo. This technology will serve as a platform to identify therapies that modulate disease-specific immune cells.
Collapse
|
35
|
Zhao C, Hao X, Cai W, Zeng LF, Zhao W, Li X. Colchicine combination therapy increases treatment tolerance in patients with arthritis: A systematic review and meta-analysis. PLoS One 2024; 19:e0316126. [PMID: 39775222 PMCID: PMC11684588 DOI: 10.1371/journal.pone.0316126] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2024] [Accepted: 12/05/2024] [Indexed: 01/11/2025] Open
Abstract
BACKGROUND Arthritis seriously affects people's quality of life, and there is an urgent clinical need to improve the efficacy of medications as well as to reduce the adverse effects induced by treatment. Combined colchicine therapy is gradually being embraced in clinical care, but the evidence remains insufficient. METHODS English databases were searched from the establishment to September 4, 2024. Eleven eligible Randomized controlled trials (RCTs) were included. The quality of the literature was assessed by the risk of bias tool in the Cochrane Handbook. Relative risk (RR) and Cohen's d (SMD) were used for categorical and continuous variables, respectively, at 95% confidence interval (CI), and Stata 17.0 software was used for statistical analysis. Sensitivity analyses were used to verify the stability of the analyzed results, and heterogeneity analyses were used to explore the sources of heterogeneity in the studies. Funnel plots and Egger's test were used to assess publication bias. RESULTS Eleven eligible RCTs were included in this study. Compared with conventional treatment, combined colchicine treatment improved patient's global assessment results (SMD = 1.24, 95% CI [0.01, 2.47], P = 0.05, I2 = 0]), stiffness (SMD = -0.81, 95% CI [-1.43, -0.19], P = 0.01, I2 = 63.91%]) and did not increase adverse effects (RR = 0.79, 95% CI [0.31, 1.27], P = 0.36, I2 = 0.00%). However, combined colchicine treatment did not improve visual analog scores (VAS) (SMD = -0.96, 95% CI [-2.85, 0.93], P = 0.13, I2 = 97.99%]), Western Ontario and McMaster Universities Arthritis Index (WOMAC) pain (SMD = 0.01, 95% CI [-0.24, 0.27], P = 0.91, I2 = 0]), WOMAC function (SMD = -0.01, 95% CI [-0.36, 0.16], P = 0.44, I2 = 0]), Total WOMAC scale (SMD = -0.05, 95% CI [-0.33, 0.22], P = 0.70, I2 = 0]), physician 's global assessment (SMD = 0.36, 95% CI [-2.27, 3.00], P = 0.79, I2 = 97.04%]) and Modified Clinical Health Assessment Questionnaire (ModHAD) (SMD = -1.72, 95% CI [-4.90,1.45], P = 0.29, I2 = 99.11%]). CONCLUSION Compared with colchicine alone, combination therapy improves patients' quality of life without increasing the incidence of adverse events.
Collapse
Affiliation(s)
- Changwei Zhao
- Changchun University of Chinese Medicine, Changchun, China
- The Affiliated Hospital of Changchun University of Chinese Medicine, Changchun, China
| | - Xiaogang Hao
- The Affiliated Hospital of Changchun University of Chinese Medicine, Changchun, China
| | - Wenjun Cai
- Third Affiliated Clinical Hospital to Changchun University of Chinese Medicine, Changchun, China
| | - Ling-Feng Zeng
- The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Wenhai Zhao
- The Affiliated Hospital of Changchun University of Chinese Medicine, Changchun, China
| | - Xiangxin Li
- The Affiliated Hospital of Changchun University of Chinese Medicine, Changchun, China
| |
Collapse
|
36
|
Dixit T, Vaidya A, Ravindran S. Therapeutic potential of antibody-drug conjugates possessing bifunctional anti-inflammatory action in the pathogenies of rheumatoid arthritis. Arthritis Res Ther 2024; 26:216. [PMID: 39695738 PMCID: PMC11656801 DOI: 10.1186/s13075-024-03452-0] [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: 08/10/2024] [Accepted: 12/05/2024] [Indexed: 12/20/2024] Open
Abstract
In an age where there is a remarkable upsurge in developing precision medicines, antibody-drug conjugates (ADCs) have emerged as a progressive therapeutic strategy. ADCs typically consist of monoclonal antibodies (mAb) conjugated to the cytotoxic payloads by utilizing a linker, combining the benefits of definitive target specificity of mAbs and potent killing impact of payload to achieve precise and efficient elimination of target cells. In addition to their well-established role in oncology, ADCs are currently demonstrating encouraging potential in addressing the unmet requirements in the treatment of autoimmune conditions such as rheumatoid arthritis (RA). Prevalent long-term autoimmune disease RA costs billions of dollars annually but still, there is a lack of precision-targeted therapeutics with minimal side effects. This review provides an overview of the RA pathogenesis, pre-existing therapies, and their limitations, the introduction of ADCs in RA treatment, the mechanism of ADCs, and a summary of ADCs in preclinical and clinical trials. Based on the literature we also propose a strategy in ADC synthesis, which may increase the efficiency in targeting multifactorial diseases like RA. We propose to utilize DMARDs (Disease-modifying anti-rheumatic drugs), the first-line treatment for RA, as a payload for ADC synthesis. DMARDs are the only class of medication that limits the disease progression, but their efficacy is limited due to off-target toxicities. Hence, utilizing them as payload will help to deliver them directly at the targeted site, reducing their off-target toxicity, which in turn will increase their efficiency in targeting disease. Also, as mAbs are not sufficient to achieve remission, they are given in combinations with DMARDs. Hence, synthesizing ADCs may reduce the multiple and higher dosages given to patients, which in turn may enhance patient compliance.
Collapse
Affiliation(s)
- Tanu Dixit
- Symbiosis School of Biological Sciences (SSBS), Faculty of Medical & Health Sciences, Symbiosis International (Deemed University), Lavale, Pune, 412115, India
| | - Anuradha Vaidya
- Symbiosis School of Biological Sciences (SSBS), Faculty of Medical & Health Sciences, Symbiosis International (Deemed University), Lavale, Pune, 412115, India
| | - Selvan Ravindran
- Symbiosis School of Biological Sciences (SSBS), Faculty of Medical & Health Sciences, Symbiosis International (Deemed University), Lavale, Pune, 412115, India.
| |
Collapse
|
37
|
Fiaschini N, Hanieh PN, Ariaudo D, Cimino R, Abbate C, Romano E, Cavalieri F, Venanzi M, Palumbo V, Scimeca M, Bernardini R, Mattei M, Migliore A, Rinaldi A. Application of Microsponge Drug Platform to Enhance Methotrexate Administration in Rheumatoid Arthritis Therapy. Pharmaceutics 2024; 16:1593. [PMID: 39771571 PMCID: PMC11676977 DOI: 10.3390/pharmaceutics16121593] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Revised: 12/09/2024] [Accepted: 12/11/2024] [Indexed: 01/11/2025] Open
Abstract
BACKGROUND/OBJECTIVES This study aimed to develop a novel nanotechnological slow-release drug delivery platform based on hyaluronic acid Microsponge (MSP) for the subcutaneous administration of methotrexate (MTX) in the treatment of rheumatoid arthritis (RA). RA is a chronic autoimmune disease characterized by joint inflammation and damage, while MTX is a common disease-modifying antirheumatic drug (DMARD), the conventional use of which is limited by adverse effects and the lack of release control. METHODS MSP were synthesized as freeze-dried powder to increase their stability and allow for a facile reconstitution prior to administration and precise MTX dosing. RESULTS A highly stable and rounded-shaped micrometric MSP, characterized by an open porosity inner structure, achieved both a high MTX loading efficiency and a slow release of MTX after injection. Our drug release assays indeed demonstrated a characteristic drug release profile consisting of a very limited burst release in the first few hours, followed by a slow release of MTX sustained for over a month. By means of a preclinical rat model of RA, the administration of MTX-loaded MSP proved to nearly double the therapeutic efficacy compared to sole MTX, according to a steep reduction in arthritic score compared to control groups. The preclinical study was replicated twice to confirm this improvement in performance and the safety profile of the MSP. CONCLUSIONS This study suggests that the MSP drug delivery platform holds significant potential for clinical use in improving RA therapy by enabling the sustained slow release of MTX, thereby enhancing therapeutic outcomes and minimizing side effects associated with conventional burst-release drug administration.
Collapse
Affiliation(s)
- Noemi Fiaschini
- Nanofaber S.r.l., Via Anguillarese 301, 00123 Rome, Italy; (N.F.); (P.N.H.)
| | | | - Daniela Ariaudo
- Department of Chemical Science and Technologies, University of Rome Tor Vergata, Via Della Ricerca Scientifica 1, 00133 Rome, Italy; (D.A.); (R.C.); (C.A.); (F.C.); (M.V.)
| | - Rita Cimino
- Department of Chemical Science and Technologies, University of Rome Tor Vergata, Via Della Ricerca Scientifica 1, 00133 Rome, Italy; (D.A.); (R.C.); (C.A.); (F.C.); (M.V.)
| | - Carlo Abbate
- Department of Chemical Science and Technologies, University of Rome Tor Vergata, Via Della Ricerca Scientifica 1, 00133 Rome, Italy; (D.A.); (R.C.); (C.A.); (F.C.); (M.V.)
| | - Elena Romano
- Centre of Advance Microscopy P. Albertano, Department of Biology, University of Rome Tor Vergata, Via della Ricerca Scientifica Snc, 00133 Rome, Italy;
| | - Francesca Cavalieri
- Department of Chemical Science and Technologies, University of Rome Tor Vergata, Via Della Ricerca Scientifica 1, 00133 Rome, Italy; (D.A.); (R.C.); (C.A.); (F.C.); (M.V.)
| | - Mariano Venanzi
- Department of Chemical Science and Technologies, University of Rome Tor Vergata, Via Della Ricerca Scientifica 1, 00133 Rome, Italy; (D.A.); (R.C.); (C.A.); (F.C.); (M.V.)
| | - Valeria Palumbo
- Department of Experimental Medicine, University of Rome Tor Vergata, 00133 Rome, Italy; (V.P.); (M.S.)
| | - Manuel Scimeca
- Department of Experimental Medicine, University of Rome Tor Vergata, 00133 Rome, Italy; (V.P.); (M.S.)
| | - Roberta Bernardini
- Department of Clinical Sciences and Translational Medicine, University of Rome Tor Vergata, 00133 Rome, Italy;
| | - Maurizio Mattei
- Interdepartmental Center for Comparative Medicine, Alternative Techniques and Aquaculture (CIMETA), University of Rome Tor Vergata, Via Montpellier 1, 00133 Rome, Italy;
| | - Alberto Migliore
- San Pietro Fatebenefratelli Hospital, Via Cassia 600, 00189 Rome, Italy
| | - Antonio Rinaldi
- Nanofaber S.r.l., Via Anguillarese 301, 00123 Rome, Italy; (N.F.); (P.N.H.)
| |
Collapse
|
38
|
Yang X, Li J, Xu C, Zhang G, Che X, Yang J. Potential mechanisms of rheumatoid arthritis therapy: Focus on macrophage polarization. Int Immunopharmacol 2024; 142:113058. [PMID: 39236455 DOI: 10.1016/j.intimp.2024.113058] [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: 07/18/2024] [Revised: 08/29/2024] [Accepted: 08/30/2024] [Indexed: 09/07/2024]
Abstract
Rheumatoid arthritis (RA) is an autoimmune inflammatory disease that affects multiple organs and systems in the human body, often leading to disability. Its pathogenesis is complex, and the long-term use of traditional anti-rheumatic drugs frequently results in severe toxic side effects. Therefore, the search for a safer and more effective antirheumatic drug is extremely important for the treatment of RA. As important immune cells in the body, macrophages are polarized. Under pathological conditions, macrophages undergo proliferation and are recruited to diseased tissues upon stimulation. In the local microenvironment, they polarize into different types of macrophages in response to specific factors and perform unique functions and roles. Previous studies have shown that there is a link between macrophage polarization and RA, indicating that certain active ingredients can ameliorate RA symptoms through macrophage polarization. Notably, Traditional Chinese medicine (TCM) monomer component and compounds demonstrate a particular advantage in this process. Building upon this insight, we reviewed and analyzed recent studies to offer valuable and meaningful insights and directions for the development and application of anti-rheumatic drugs.
Collapse
Affiliation(s)
- Xinyu Yang
- College of Acupuncture and Massage, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Jinling Li
- College of Acupuncture and Massage, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Chengchao Xu
- College of Rehabilitation Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Guangheng Zhang
- Department of First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Xinzhen Che
- Department of First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Jiguo Yang
- College of Acupuncture and Massage, Shandong University of Traditional Chinese Medicine, Jinan, China.
| |
Collapse
|
39
|
Hemajha L, Singh S, Biji CA, Balde A, Benjakul S, Nazeer RA. A review on inflammation modulating venom proteins/peptide therapeutics and their delivery strategies: A review. Int Immunopharmacol 2024; 142:113130. [PMID: 39278056 DOI: 10.1016/j.intimp.2024.113130] [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: 05/24/2024] [Revised: 09/02/2024] [Accepted: 09/06/2024] [Indexed: 09/17/2024]
Abstract
Inflammation is an initial biological reaction that occurs in response to infection caused by foreign pathogens or injury. This process involves a tightly controlled series of signaling events at the molecular and cellular levels, with the ultimate goal of restoring tissue balance and protecting against invading pathogens. Malfunction in the process of inflammation can result in a diverse array of diseases, such as cardiovascular, neurological, and autoimmune disorders. Therefore, the management of inflammation is of utmost importance in modern medicine. Nonsteroidal anti-inflammatory drugs (NSAIDs) and corticosteroids have long been the mainstays of pharmacological treatment for inflammation, effectively alleviating symptoms in many patients. Recently, toxins and venom, formerly seen as mostly harmful to the human body, have been recognized as possible medicinal substances for treating inflammation. Organisms that are venomous, such as spiders, scorpions, snakes, and certain marine species, have developed a wide range of powerful toxins that can effectively disable or discourage predators. Remarkably, the majority of these poisons and venoms consist of proteins and peptides, which are acknowledged as significant bioactive compounds with medicinal potential. The goal of this review is to investigate the medicinal potential of peptides derived from venoms and their complex mechanism of action in suppressing inflammation. This review also discusses various challenges and future prospects for effective venom delivery.
Collapse
Affiliation(s)
- Lakshmikanthan Hemajha
- Biopharmaceuticals Lab, Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur, Chennai 603203, Tamilnadu, India
| | - Simran Singh
- Biopharmaceuticals Lab, Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur, Chennai 603203, Tamilnadu, India
| | - Catherin Ann Biji
- Biopharmaceuticals Lab, Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur, Chennai 603203, Tamilnadu, India
| | - Akshad Balde
- Biopharmaceuticals Lab, Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur, Chennai 603203, Tamilnadu, India
| | - Soottawat Benjakul
- International Center of Excellence in Seafood Science and Innovation, Faculty of Agro Industry, Prince of Songkla University, Hat Yai, Songkhla 90110, Thailand; Department of Food and Nutrition, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Rasool Abdul Nazeer
- Biopharmaceuticals Lab, Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur, Chennai 603203, Tamilnadu, India.
| |
Collapse
|
40
|
Zewail MB, Doghish AS, El-Husseiny HM, Mady EA, Mohammed OA, Elbadry AMM, Elbokhomy AS, Bhnsawy A, El-Dakroury WA. Lipid-based nanocarriers: an attractive approach for rheumatoid arthritis management. Biomater Sci 2024; 12:6163-6195. [PMID: 39484700 DOI: 10.1039/d4bm01058b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2024]
Abstract
Lipid nanoparticles (LNPs) have emerged as transformative tools in modern drug delivery, offering unparalleled potential in enhancing the efficacy and safety of various therapeutics. In the context of rheumatoid arthritis (RA), a disabling autoimmune disorder characterized by chronic inflammation, joint damage, and limited patient mobility, LNPs hold significant promise for revolutionizing treatment strategies. LNPs offer several advantages over traditional drug delivery systems, including improved pharmacokinetics, enhanced tissue penetration, and reduced systemic toxicity. This article concisely summarizes the pathogenesis of RA, its associated risk factors, and therapeutic techniques and their challenges. Additionally, it highlights the noteworthy advancements made in managing RA through LNPs, including liposomes, niosomes, bilosomes, cubosomes, spanlastics, ethosomes, solid lipid nanoparticles, lipid micelles, lipid nanocapsules, nanostructured lipid carriers, etc. It also delves into the specific functional attributes of these nanocarrier systems, focusing on their role in treating and monitoring RA.
Collapse
Affiliation(s)
- Moataz B Zewail
- School of Chemical Engineering, Faculty of Sciences, Engineering and Technology, The University of Adelaide, Adelaide, SA, 5005, Australia
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Badr University in Cairo (BUC), Badr City, Cairo 11829, Egypt.
| | - Ahmed S Doghish
- Department of Biochemistry, Faculty of Pharmacy, Badr University in Cairo (BUC), Badr City, 17 Cairo, 11829, Egypt
- Biochemistry and Molecular Biology Department, Faculty of Pharmacy (Boys), Al-Azhar University, Nasr City 11231, Cairo, Egypt
| | - Hussein M El-Husseiny
- Laboratory of Veterinary Surgery, Department of Veterinary Medicine, Faculty of Agriculture, Tokyo University of Agriculture and Technology, 3-5-8 Saiwai Cho, Fuchu-shi, Tokyo 183-8509, Japan
- Department of Surgery, Anesthesiology, and Radiology, Faculty of Veterinary Medicine, Benha University, Moshtohor, Toukh, Elqaliobiya, 13736, Egypt
- Laboratory of Veterinary Physiology, Department of Veterinary Medicine, Tokyo University of Agriculture and Technology, Tokyo 183-8509, Japan
- Institute of Global Innovation Research, Tokyo University of Agriculture and Technology, 3-8-1 Harumi-cho, Fuchu-shi, Tokyo 183-8538, Japan
| | - Eman A Mady
- Laboratory of Veterinary Physiology, Department of Veterinary Medicine, Faculty of Agriculture, 10 Tokyo University of Agriculture and Technology, 3-5-8 Saiwai Cho, Fuchu-shi, Tokyo 183-8509, Japan
- Department of Animal Hygiene, Behavior, and Management, Faculty of Veterinary Medicine, Benha University, Moshtohor, Toukh, Elqaliobiya 13736, Egypt
| | - Osama A Mohammed
- Department of Pharmacology, College of Medicine, University of Bisha, Bisha 61922, Saudi Arabia
| | - Abdullah M M Elbadry
- Badr University in Cairo Research Center, Badr University in Cairo, Badr City, Cairo 11829, Egypt
| | - Amir S Elbokhomy
- Faculty of Pharmacy, Badr University in Cairo (BUC), Badr City, Cairo 11829, Egypt
| | - Abdelmenem Bhnsawy
- Faculty of Pharmacy, Badr University in Cairo (BUC), Badr City, Cairo 11829, Egypt
| | - Walaa A El-Dakroury
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Badr University in Cairo (BUC), Badr City, Cairo 11829, Egypt.
| |
Collapse
|
41
|
Jia M, Ren W, Wang M, Liu Y, Wang C, Zhang Z, Xu M, Ding N, Li C, Yang H. Surface saturation of drug-loaded hollow manganese dioxide nanoparticles with human serum albumin for treating rheumatoid arthritis. Drug Deliv 2024; 31:2380538. [PMID: 39044468 PMCID: PMC11271085 DOI: 10.1080/10717544.2024.2380538] [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: 09/20/2023] [Accepted: 07/03/2024] [Indexed: 07/25/2024] Open
Abstract
Rheumatoid arthritis (RA) is a chronic inflammatory joint disease accompanied by energy depletion and accumulation of reactive oxygen species (ROS). Inorganic nanoparticles (NPs) offer great promise for the treatment of RA because they mostly have functions beyond being drug carriers. However, conventional nanomaterials become coated with a protein corona (PC) or lose their cargo prematurely in vivo, reducing their therapeutic efficacy. To avoid these problems, we loaded methotrexate (MTX) into hollow structured manganese dioxide nanoparticles (H-MnO2 NPs), then coated them with a 'pseudo-corona' of human serum albumin (HSA) at physiological concentrations to obtain HSA-MnO2@MTX NPs. Efficacy of MTX, MnO2@MTX, and HSA-MnO2@MTX NPs was compared in vitro and in vivo. Compared to MnO2@MTX, HSA-coated NPs were taken up better by lipopolysaccharide-activated RAW264.7 and were more effective at lowering levels of pro-inflammatory cytokines and preventing ROS accumulation. HSA-MnO2@MTX NPs were also more efficient at blocking the proliferation and migration of fibroblast-like synoviocytes from rats with collagen-induced arthritis. In this rat model, HSA-MnO2@MTX NPs showed better biodistribution than other treatments, specifically targeting the ankle joint. Furthermore, HSA-MnO2@MTX NPs reduced swelling in the paw, regulated pro-inflammatory cytokine production, and limited cartilage degradation and signs of inflammation. These results establish the therapeutic potential of HSA-MnO2@MTX NPs against RA.
Collapse
Affiliation(s)
- Ming Jia
- Department of Pharmaceutical Sciences, School of Pharmacy, Southwest Medical University, Luzhou, China
- Nanchong Institute for Food and Drug Control, Nanchong, China
| | - Wei Ren
- National Traditional Chinese Medicine Clinical Research Base and Drug Research Center of the Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, China
| | - Minrui Wang
- School of Basic Medical Sciences, Southwest Medical University, Luzhou, China
- Nucleic Acid Medicine of Luzhou Key Laboratory, Southwest Medical University, Luzhou, China
| | - Yan Liu
- Department of Pharmaceutical Sciences, School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Chenglong Wang
- Department of Pharmaceutical Sciences, School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Zongquan Zhang
- Department of Pharmaceutical Sciences, School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Maochang Xu
- Department of Pharmaceutical Sciences, School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Nianhui Ding
- School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Chunhong Li
- Department of Pharmaceutical Sciences, School of Pharmacy, Southwest Medical University, Luzhou, China
- Nucleic Acid Medicine of Luzhou Key Laboratory, Southwest Medical University, Luzhou, China
| | - Hong Yang
- Sichuan Clinical Research Center for Birth Defects, Luzhou, China
- The Affiliated Hospital of Southwest Medical University, Luzhou, China
| |
Collapse
|
42
|
Poon EK, Liu L, Wu KC, Lim J, Sweet MJ, Lohman RJ, Iyer A, Fairlie DP. A novel inhibitor of class IIa histone deacetylases attenuates collagen-induced arthritis. Br J Pharmacol 2024; 181:4804-4821. [PMID: 39223784 DOI: 10.1111/bph.17306] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2023] [Revised: 06/15/2024] [Accepted: 06/27/2024] [Indexed: 09/04/2024] Open
Abstract
BACKGROUND AND PURPOSE Most inhibitors of histone deacetylases (HDACs) are not selective and are cytotoxic. Some have anti-inflammatory activity in disease models, but cytotoxicity prevents long-term uses in non-fatal diseases. Inhibitors selective for class IIa HDACs are much less cytotoxic and may have applications in management of chronic inflammatory diseases. EXPERIMENTAL APPROACH LL87 is a novel HDAC inhibitor examined here for HDAC enzyme selectivity. It was also investigated in macrophages for cytotoxicity and for inhibition of lipopolysaccharide (LPS)-stimulated cytokine secretion. In a rat model of collagen-induced arthritis, LL87 was investigated for effects on joint inflammation in Dark Agouti rats. Histological, immunohistochemical, micro-computed tomography and molecular analyses characterise developing arthritis and anti-inflammatory efficacy. KEY RESULTS LL87 was significantly more inhibitory against class IIa than class I or IIb HDAC enzymes. In macrophages, LL87 was not cytotoxic and reduced both LPS-induced secretion of pro-inflammatory cytokines, and IL6-induced class IIa HDAC activity. In rats, LL87 attenuated paw swelling and clinical signs of arthritis, reducing collagen loss and histological damage in ankle joints. LL87 decreased immune cell infiltration, especially pro-inflammatory macrophages and osteoclasts, into synovial joints and significantly reduced expression of pro-inflammatory cytokines and tissue-degrading proteases. CONCLUSION AND IMPLICATIONS A novel inhibitor of class IIa HDACs has been shown to have an anti-inflammatory and anti-arthritic profile distinct from current therapies. It is efficacious in reducing macrophage infiltration and joint inflammation in a chronic model of rat arthritis.
Collapse
Affiliation(s)
- Eunice K Poon
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland, Australia
- Australian Research Council Centre of Excellence for Innovations in Peptide and Protein Science, The University of Queensland, Brisbane, Queensland, Australia
| | - Ligong Liu
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland, Australia
| | - Kai-Chen Wu
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland, Australia
- Australian Research Council Centre of Excellence for Innovations in Peptide and Protein Science, The University of Queensland, Brisbane, Queensland, Australia
| | - Junxian Lim
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland, Australia
- Australian Research Council Centre of Excellence for Innovations in Peptide and Protein Science, The University of Queensland, Brisbane, Queensland, Australia
| | - Matthew J Sweet
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland, Australia
| | - Rink-Jan Lohman
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland, Australia
| | - Abishek Iyer
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland, Australia
| | - David P Fairlie
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland, Australia
- Australian Research Council Centre of Excellence for Innovations in Peptide and Protein Science, The University of Queensland, Brisbane, Queensland, Australia
| |
Collapse
|
43
|
Zhao X, Wu S, Ni S, Zhong Y, Qin X, Zhang K, Qu K, Zhu L, Wu W. Tannic Acid-Based Biomimetic Nanomedicine with Pathological Reactive Oxygen Species-Responsive Cargo Release for Relieving Inflammation in Rheumatoid Arthritis. ACS APPLIED MATERIALS & INTERFACES 2024; 16:59789-59802. [PMID: 39448903 DOI: 10.1021/acsami.4c11494] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/26/2024]
Abstract
Rheumatoid arthritis (RA) is a chronic disease characterized by immune cell infiltration and cartilage damage. The local lesion of RA shows severe oxidative stress and proinflammatory cytokine secretion. For drug therapy, the efficacy of agents, such as methotrexate (MTX), may be greatly limited, resulting from the low bioavailability, immune clearance, and toxic side effects. A nanocarrier (TA-PBA NPs) was developed with anti-inflammatory and antioxidant activities, combined with MTX to prepare nanomedicine (MTX NPs) for synergistic treatment of RA. Moreover, inspired by the biological functions homing to inflammation lesion of macrophages, the biomimetic nanomedicine camouflaged with macrophage membrane (MM@MTX NPs) was constructed. TA-PBA NPs could timely promote MTX release in response to the overaccumulated ROS to exhibit high anti-inflammatory and antioxidant activities for alleviating RA progression. The experimental results confirmed that MM@MTX NPs could significantly reduce the secretion of proinflammatory cytokines (TNF-α) while significantly increasing the typical anti-inflammatory cytokines (IL-10), promote the phenotype transformation of macrophages from M1 to M2, and up-regulate the Nrf2-keap1 pathway-related proteins (HO-1 and NRF2) to positively regulate the local inflammation for effectively inhibiting RA development. Thus, MM@MTX NPs represent a possible candidate as a safe and efficient nanotherapy platform for RA management.
Collapse
Affiliation(s)
- Xiong Zhao
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing 400044, China
| | - Shuai Wu
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing 400044, China
| | - Sheng Ni
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing 400044, China
| | - Yuan Zhong
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing 400044, China
| | - Xian Qin
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing 400044, China
- Chongqing Municipality Clinical Research Center for Endocrinology and Metabolic Diseases, Chongqing University Three Gorges Hospital, Chongqing 404000, China
| | - Kun Zhang
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing 400044, China
- Chongqing Municipality Clinical Research Center for Endocrinology and Metabolic Diseases, Chongqing University Three Gorges Hospital, Chongqing 404000, China
| | - Kai Qu
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing 400044, China
- Chongqing Municipality Clinical Research Center for Endocrinology and Metabolic Diseases, Chongqing University Three Gorges Hospital, Chongqing 404000, China
| | - Li Zhu
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing 400044, China
| | - Wei Wu
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing 400044, China
- JinFeng Laboratory, Chongqing 401329, China
| |
Collapse
|
44
|
Al-Nasser MM, Al-Saeedi MJ, Alhowaiti SA, Shinwari Z, Alhamlan FS, Alothaid H, Alkahtani S, Al-Qahtani AA. Combination of Methotrexate and Resveratrol Reduces Pro-Inflammatory Chemokines in Human THP-1 Cells. J Inflamm Res 2024; 17:8085-8098. [PMID: 39507267 PMCID: PMC11539838 DOI: 10.2147/jir.s482503] [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: 08/02/2024] [Accepted: 10/08/2024] [Indexed: 11/08/2024] Open
Abstract
Introduction Methotrexate (MTX) is a widely used anti-metabolite drug in cancer therapy, but its efficacy is often hindered by reactive oxygen species (ROS)-induced cellular toxicity. Resveratrol, a natural polyphenol, possesses antioxidant and anticancer properties. Therefore, this in vitro study aimed to investigate the synergistic anti-proliferative and anti-inflammatory effects of MTX and resveratrol in human THP-1 cells. Methods THP-1 cells were differentiated into macrophage-like cells using phorbol 12-myristate 13-acetate. In vitro experiments assessed the impact of various concentrations of MTX and resveratrol on cell viability and proliferation using the MTT assay. Concentration-effect curves were generated to elucidate their relationship. Gene expression was analyzed by RT-qPCR, while chemokine expression was measured via ELISA. Phagocytic and migratory activities were also evaluated. Results Differentiated THP-1 cells were treated with MTX and resveratrol and stimulated with dimethyl sulfoxide (DMSO) and lipopolysaccharide (LPS) as inflammatory stimuli. The combination of MTX and resveratrol exhibited an anti-proliferative effect in THP-1 cells (p = 0.03). The concentration-effect curve revealed IC50 values of 49.15 µg at 24 hours (R = 0.8236) and 2.029e-005 µg at 48 hours (R = 0.97) for MTX, and 20.17 µg at 48 hours (R = 1.000) and 55.38 µg at 96 hours (R = 0.9666) for resveratrol. Co-treatment with MTX and resveratrol significantly reduced mRNA and chemokine expression of CCL2, CCL3, CCL4, CCL5, and CXCL10 (p < 0.05). Moreover, decreased phagocytic and migratory activities were confirmed by phagocytosis and migration assays (p < 0.05). Conclusion The combination of MTX and resveratrol effectively attenuated pro-inflammatory activity in THP-1 cells, as evidenced by the downregulation of mRNA and chemokine expression. These findings suggest that the synergistic effects of MTX and resveratrol hold promise for enhancing cancer therapeutics.
Collapse
Affiliation(s)
- Moonerah M Al-Nasser
- Department of Zoology, College of Science, King Saud University, Riyadh, Saudi Arabia
| | - Mashael J Al-Saeedi
- Department of Infection and Immunity, Research Centre, King Faisal Specialist Hospital & Research Centre, Riyadh, Saudi Arabia
| | - Saltana A Alhowaiti
- Department of Infection and Immunity, Research Centre, King Faisal Specialist Hospital & Research Centre, Riyadh, Saudi Arabia
| | - Zakia Shinwari
- Therapeutics & Biomarker Discovery for Clinical Applications, Stem Cell & Tissue Re-Engineering Program, King Faisal Specialist Hospital and Research Centre, Riyadh, Saudi Arabia
| | - Fatimah S Alhamlan
- Department of Infection and Immunity, Research Centre, King Faisal Specialist Hospital & Research Centre, Riyadh, Saudi Arabia
- Department of Microbiology and Immunology, College of Medicine, Alfaisal University, Riyadh, Saudi Arabia
| | - Hani Alothaid
- Department of Basic Sciences, Faculty of Applied Medical Sciences, Al-Baha University, Al-Baha, Saudi Arabia
| | - Saad Alkahtani
- Department of Zoology, College of Science, King Saud University, Riyadh, Saudi Arabia
| | - Ahmed A Al-Qahtani
- Department of Infection and Immunity, Research Centre, King Faisal Specialist Hospital & Research Centre, Riyadh, Saudi Arabia
- Department of Microbiology and Immunology, College of Medicine, Alfaisal University, Riyadh, Saudi Arabia
| |
Collapse
|
45
|
Ge S, Wang X, Zhao X, Yuan L, Bao X, Sun C, Gong Z, Guo J, Yuan S, Hu D, Yang J, Yuan B, Zhang G. Responsive Multi-Arm PEG-Modified COF Nanocomposites: Dynamic Photothermal, pH/ROS Dual-Responsive, Targeted Carriers for Rheumatoid Arthritis Treatment. Adv Healthc Mater 2024; 13:e2401744. [PMID: 38885286 DOI: 10.1002/adhm.202401744] [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: 05/11/2024] [Revised: 06/12/2024] [Indexed: 06/20/2024]
Abstract
Rheumatoid arthritis (RA) is a chronic immune disease characterized by the infiltration of immune cells and the proliferation of fibroblast-like synoviocytes (FLS) at the joint site, leading to inflammation and joint destruction. However, the available treatment options targeting both inflammatory and proliferative FLS are limited. Herein, this work presents three covalent organic frameworks (COFs) photothermal composite systems modified with multi-armed polyethylene glycols (PEG) for the treatment of RA. These systems exhibit a dual response under low pH and high reactive oxygen species (ROS) conditions at the site of inflammation, with a specific focus on delivering the protein drug ribonuclease A (RNase A). Notably, molecular docking studies reveal the interaction between RNase A and NF-κB p65 protein, and Western blotting confirm its inhibitory effect on NF-κB activity. In vitro and in vivo experiments verify the significant reduction in joint swelling and deformities in adjuvant-induced arthritis (AIA) rats after treatment with RNase A delivered by multi-armed PEG-modified COF ligands, restoring joint morphology to normal. These findings underscore the promising therapeutic potential of COFs for the treatment of RA, highlighting their unique capabilities in addressing both inflammatory and proliferative aspects of the disease and expanding the scope of biomedical applications for COFs.
Collapse
MESH Headings
- Animals
- Polyethylene Glycols/chemistry
- Arthritis, Rheumatoid/metabolism
- Arthritis, Rheumatoid/pathology
- Arthritis, Rheumatoid/drug therapy
- Arthritis, Rheumatoid/therapy
- Rats
- Reactive Oxygen Species/metabolism
- Nanocomposites/chemistry
- Nanocomposites/therapeutic use
- Hydrogen-Ion Concentration
- Metal-Organic Frameworks/chemistry
- Metal-Organic Frameworks/pharmacology
- Ribonuclease, Pancreatic/metabolism
- Ribonuclease, Pancreatic/chemistry
- Arthritis, Experimental/pathology
- Arthritis, Experimental/drug therapy
- Arthritis, Experimental/therapy
- Arthritis, Experimental/metabolism
- Humans
- Drug Carriers/chemistry
- Male
- Molecular Docking Simulation
- Synoviocytes/metabolism
- Synoviocytes/drug effects
Collapse
Affiliation(s)
- Saisai Ge
- Department of Pharmacology, School of Basic Medical Sciences, Anhui Medical University, Hefei, 230032, China
| | - Xinyue Wang
- Department of Pharmacology, School of Basic Medical Sciences, Anhui Medical University, Hefei, 230032, China
| | - Xinru Zhao
- Department of Pharmacology, School of Basic Medical Sciences, Anhui Medical University, Hefei, 230032, China
| | - Lingling Yuan
- Department of Pharmacology, School of Basic Medical Sciences, Anhui Medical University, Hefei, 230032, China
| | - Xuewei Bao
- Department of Pharmacology, School of Basic Medical Sciences, Anhui Medical University, Hefei, 230032, China
| | - Caidie Sun
- Department of Pharmacology, School of Basic Medical Sciences, Anhui Medical University, Hefei, 230032, China
| | - Zehua Gong
- Department of Pharmacology, School of Basic Medical Sciences, Anhui Medical University, Hefei, 230032, China
| | - Jun Guo
- Department of Pharmacology, School of Basic Medical Sciences, Anhui Medical University, Hefei, 230032, China
| | - Siyu Yuan
- Department of Pharmacology, School of Basic Medical Sciences, Anhui Medical University, Hefei, 230032, China
| | - Danyou Hu
- Department of Microbiology, School of Basic Medical Sciences, Anhui Medical University, Hefei, 230032, China
| | - Jing Yang
- Experimental Teaching Center for Preventive Medicine, School of Public Health, Anhui Medical University, Hefei, 230032, China
| | - Bin Yuan
- Department of Pharmacology, School of Basic Medical Sciences, Anhui Medical University, Hefei, 230032, China
| | - Guiyang Zhang
- Department of Pharmacology, School of Basic Medical Sciences, Anhui Medical University, Hefei, 230032, China
| |
Collapse
|
46
|
Ohashi Y, Takahashi N, Sobue Y, Suzuki M, Sato R, Maeda M, Kihira D, Kishimoto K, Terabe K, Asai S, Imagama S. Associations of frailty with RA-ILD and poor control of disease activity in patients with rheumatoid arthritis: A multi-center retrospective observational study. J Orthop Sci 2024; 29:1496-1502. [PMID: 38044213 DOI: 10.1016/j.jos.2023.11.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 10/10/2023] [Accepted: 11/23/2023] [Indexed: 12/05/2023]
Abstract
BACKGROUND This study aimed to investigate factors associated with frailty in rheumatoid arthritis (RA) patients. METHODS A total of 656 RA patients were evaluated using data from an observational study in 2022. Among these patients, 152 with frailty were assigned to the frailty group, and 504 without frailty were assigned to the non-frailty group. Patient characteristics were compared between the two groups by univariate analysis, and factors associated with frailty were assessed by logistic regression analysis. Patient characteristics were also compared between patients with RA-associated interstitial lung disease (RA-ILD) (n = 102) and those without RA-ILD (n = 554). RESULTS The frailty group was older (mean: 73.6 vs. 66.8 years) and had a higher DAS28-ESR (3.67 vs. 2.66), a higher HAQ-DI (1.13 vs. 0.32), and a higher rate of RA-ILD (25.0 vs. 12.7 %) than the non-frailty group. Age (OR: 1.03, 95 % CI: 1.01-1.05), HAQ-DI (3.22, 2.28-4.56), DAS28-ESR (1.44, 1.19-1.75), and RA-ILD (2.21, 1.24-3.94) were associated with frailty. RA patients with RA-ILD were older (73.3 vs. 67.5 years) and had a higher DAS28-ESR (3.30 vs. 2.80), a higher HAQ-DI (1.19 vs. 0.32), a higher proportion of frail patients (37.3 vs. 20.6 %), lower MTX use (26.5 vs. 62.9 %), and higher steroid use (44.1 vs. 26.8 %) than those without RA-ILD. CONCLUSIONS Maintaining reasonable control of disease activity is necessary for RA patients, including those with RA-ILD, to recover from frailty.
Collapse
Affiliation(s)
- Yoshifumi Ohashi
- Department of Orthopedic Surgery, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, Aichi 466-8550, Japan; Department of Orthopedic Surgery, Yokkaichi Municipal Hospital, 2-2-37 Shibata, Yokkaichi, Mie 453-8511, Japan.
| | - Nobunori Takahashi
- Department of Orthopedic Surgery, Aichi Medical University, Graduate School of Medicine, 1-1 Karimata yazako, Nagakute, Aichi 480-1195, Japan.
| | - Yasumori Sobue
- Department of Orthopedic Surgery, Japan Red Cross, Aichi Medical Center, Nagoya Daiichi Hospital, 3-35 Michishita-cho, Nakamura-ku, Nagoya, Aichi 453-8511, Japan
| | - Mochihito Suzuki
- Department of Orthopedic Surgery, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, Aichi 466-8550, Japan; Department of Orthopedic Surgery, Japan Community Health Care Organization, Kani Tono Hospital, 1221-5 Tsuchida, Kani, Gifu 509-0206, Japan
| | - Ryo Sato
- Department of Orthopedic Surgery, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, Aichi 466-8550, Japan
| | - Masataka Maeda
- Department of Orthopedic Surgery, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, Aichi 466-8550, Japan
| | - Daisuke Kihira
- Department of Orthopedic Surgery, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, Aichi 466-8550, Japan
| | - Kenji Kishimoto
- Department of Orthopedic Surgery, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, Aichi 466-8550, Japan
| | - Kenya Terabe
- Department of Orthopedic Surgery, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, Aichi 466-8550, Japan
| | - Shuji Asai
- Department of Orthopedic Surgery, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, Aichi 466-8550, Japan
| | - Shiro Imagama
- Department of Orthopedic Surgery, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, Aichi 466-8550, Japan
| |
Collapse
|
47
|
Liu Y, Qu Y, Liu C, Zhang D, Xu B, Wan Y, Jiang P. Neutrophil extracellular traps: Potential targets for the treatment of rheumatoid arthritis with traditional Chinese medicine and natural products. Phytother Res 2024; 38:5067-5087. [PMID: 39105461 DOI: 10.1002/ptr.8311] [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: 02/27/2024] [Revised: 07/06/2024] [Accepted: 07/19/2024] [Indexed: 08/07/2024]
Abstract
Rheumatoid arthritis (RA) is a chronic systemic autoimmune disease. Abnormal formation of neutrophil extracellular traps (NETs) at the synovial membrane leads to the release of many inflammatory cytokines, including IL-1β, IL-6, and TNF-α. Elastase, histone H3, and myeloperoxidase, which are carried by NETs, damage the soft tissues of the joints and aggravate the progression of RA. The balance of NET formation coordinates the pro-inflammatory and anti-inflammatory effects and plays a key role in the development of RA. Therefore, when NETs are used as effector targets, highly targeted drugs with fewer side effects can be developed to treat RA without damaging the host immune system. Currently, an increasing number of studies have shown that traditional Chinese medicines and natural products can regulate the formation of NETs through multiple pathways to counteract RA, which shows great potential for the treatment of RA and has a promising future for clinical application. In this article, we review the latest biological progress in understanding NET formation, the mechanism of NETs in RA, and the potential targets or pathways related to the modulation of NET formation by Chinese medicines and natural products. This review provides a relevant basis for the use of Chinese medicines and natural products as natural adjuvants in the treatment of RA.
Collapse
Affiliation(s)
- Yuan Liu
- The first Clinical Medical College, Shandong University of Chinese Traditional Medicine, Jinan, China
| | - Yuan Qu
- The first Clinical Medical College, Shandong University of Chinese Traditional Medicine, Jinan, China
| | - Chuanguo Liu
- Experimental Center, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Di Zhang
- Rheumatology and Immunology Department, The Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Bing Xu
- Rheumatology and Immunology Department, The Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Yakun Wan
- School of Rehabilitation Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Ping Jiang
- The first Clinical Medical College, Shandong University of Chinese Traditional Medicine, Jinan, China
- Rheumatology and Immunology Department, The Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| |
Collapse
|
48
|
Li H, Duan X, Wu F, Yin L. Simultaneous Determination of Methotrexate and 7-Hydroxy-Methotrexate by UHPLC-MS 3 Assay Coupled With Multiple Stage Fragmentation to Enhance Sensitivity. J Sep Sci 2024; 47:e70020. [PMID: 39513194 DOI: 10.1002/jssc.70020] [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: 08/19/2024] [Revised: 10/09/2024] [Accepted: 10/23/2024] [Indexed: 11/15/2024]
Abstract
A selective ultra-high performance liquid chromatography tandem mass spectrometry cubed (UHPLC/MS3) assay for simultaneous determination of methotrexate (MTX) and 7-hydroxy-methotrexate (7-OH-MTX) in human plasma was developed and validated. After protein precipitation with methanol, chromatographic separation of MTX, MTX-d3, and 7-OH-MTX was performed on a Waters AcQuity UPLC-BEH column (2.1 × 50 mm I.D., 1.7 µm) with gradient elution. MRM3 transition was used for the detection of MTX (m/z 455.2→308.0→175.1) and 7-OH-MTX (m/z 471.3→191.0→148.1). The linear range of UHPLC/MS3 assay for the determination of MTX and 7-OH-MTX was 0.5-300 ng/mL (R2 ≥ 0.99) and 5-1500 ng/mL (R2 ≥ 0.99), respectively. The enhanced selectivity and sensitivity are the novelties of the developed UHPLC/MS3 assay. The analytical method was successfully applied to simultaneous determination of MTX and its major metabolite 7-OH-MTX in real human plasma samples.
Collapse
Affiliation(s)
- Huaqiang Li
- College of Biological and Food Engineering, Guangdong University of Petrochemical Technology, Maoming, Guangdong, P.R. China
| | - Xujian Duan
- School of Chemical Engineering, Ocean and Life Sciences, Dalian University of Technology, Panjin, Liaoning, P.R. China
- BYD Auto Industry Company Limited, Shenzhen, P.R. China
| | - Feifei Wu
- College of Biological and Food Engineering, Guangdong University of Petrochemical Technology, Maoming, Guangdong, P.R. China
| | - Lei Yin
- School of Chemical Engineering, Ocean and Life Sciences, Dalian University of Technology, Panjin, Liaoning, P.R. China
- Department of Laboratory Medicine, The First Hospital of Jilin University, Changchun, Jilin Province, P.R. China
| |
Collapse
|
49
|
Wang JX, Zhang JH, Guo M, Huang WR, Zheng HL, Liao YQ, Yan YX, Lin ZL, Qiu NF, Dai CT, Yu XB, Yu Y. Safety Evaluation of a Potential Anti-Rheumatoid Arthritis Candidate, Levamisole. J Inflamm Res 2024; 17:7837-7849. [PMID: 39494209 PMCID: PMC11531285 DOI: 10.2147/jir.s477013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Accepted: 10/24/2024] [Indexed: 11/05/2024] Open
Abstract
INTRODUCTION Given the limitations and adverse effects of current rheumatoid arthritis (RA) treatments, there is an urgent need for safer and more effective therapeutic options. Levamisole (LVM) is a non-specific immunomodulator with potential for treating skin diseases, tumors, and autoimmune disorders. Recognizing LVM's potential despite its controversial reputation, this study aimed to investigate its safety profile and therapeutic efficacy towards RA. METHODS To evaluate the potential toxicity of LVM, a 28-day oral administration was conducted in SD rats, assessing general toxicity and neurotoxicity using serum biochemical indicators, the Morris water maze test, transmission electron microscopy, and H&E staining. Subsequently, the therapeutic effects of LVM on RA were evaluated. RESULTS The results showed that 30 mg/kg LVM has promising therapeutic effects in the treatment of RA with negligible toxicity from 45 mg/kg to 180 mg/kg. DISCUSSIONS This study provides valuable preclinical data on the safety and efficacy of LVM, laying the groundwork for future clinical applications and potentially offering a safer and more effective treatment option for RA patients.
Collapse
Affiliation(s)
- Jun-Xiang Wang
- School of Pharmacy & Fujian Center for New Drug Safety Evaluation, Fujian Medical University, Fuzhou, 350122, People’s Republic of China
| | - Jia-Hui Zhang
- School of Pharmacy & Fujian Center for New Drug Safety Evaluation, Fujian Medical University, Fuzhou, 350122, People’s Republic of China
| | - Mu Guo
- School of Pharmacy & Fujian Center for New Drug Safety Evaluation, Fujian Medical University, Fuzhou, 350122, People’s Republic of China
| | - Wei-Rui Huang
- School of Pharmacy & Fujian Center for New Drug Safety Evaluation, Fujian Medical University, Fuzhou, 350122, People’s Republic of China
| | - Han-Lu Zheng
- School of Pharmacy & Fujian Center for New Drug Safety Evaluation, Fujian Medical University, Fuzhou, 350122, People’s Republic of China
| | - Yi-Qi Liao
- School of Pharmacy & Fujian Center for New Drug Safety Evaluation, Fujian Medical University, Fuzhou, 350122, People’s Republic of China
| | - Ying-Xue Yan
- School of Pharmacy & Fujian Center for New Drug Safety Evaluation, Fujian Medical University, Fuzhou, 350122, People’s Republic of China
| | - Zhao-Long Lin
- School of Pharmacy & Fujian Center for New Drug Safety Evaluation, Fujian Medical University, Fuzhou, 350122, People’s Republic of China
| | - Neng-Fu Qiu
- School of Pharmacy & Fujian Center for New Drug Safety Evaluation, Fujian Medical University, Fuzhou, 350122, People’s Republic of China
| | - Cui-Ting Dai
- Department of Pathology, The People’s Hospital Affiliated to Fujian University of Traditional Chinese Medicine, Fuzhou, 350004, People’s Republic of China
| | - Xiang-Bin Yu
- School of Pharmacy & Fujian Center for New Drug Safety Evaluation, Fujian Medical University, Fuzhou, 350122, People’s Republic of China
| | - Yue Yu
- School of Pharmacy & Fujian Center for New Drug Safety Evaluation, Fujian Medical University, Fuzhou, 350122, People’s Republic of China
| |
Collapse
|
50
|
Chen Y, Chen QW, Fu FS, Gu HY, Yu A, Zhang XZ. Bone Destruction-Chemotactic Osteoprogenitor Cells Deliver Liposome Nanomedicines for the Treatment of Osteosarcoma and Osteoporosis. ACS NANO 2024; 18:29864-29879. [PMID: 39424791 DOI: 10.1021/acsnano.4c10053] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/21/2024]
Abstract
Therapeutic efficacy of skeletal diseases is usually limited by unfavorable drug delivery due to incapable bone targeting and low bone affinity of conventional drug carriers, as well as relatively reduced vascularization and dense structure of bone tissues. Due to CXC chemokine receptor 4 (CXCR4)/CXC chemokine ligand 12 (CXCL12) signal axis-guided recruitment, osteoprogenitor cells (OPCs) can actively migrate to bone disease nidus. Here, drugs-loaded nanoliposomes are prepared and decorated onto OPCs by biotin-streptavidin linkage for precise bone disease targeting and effective drug delivery. In mouse models of tibia defect and orthotopic osteosarcoma, superior targeting property of OPCs-based drug delivery systems toward diseased bone niduses is verified. By encapsulating antitumor and antiosteoporosis drugs into nanoliposomes, OPCs-based drug delivery systems effectively inhibit disease development and restore bone destruction in mouse models of orthotopic osteosarcoma and ovariectomized osteoporosis. This study reveals a cell-based drug delivery system for precise bone disease targeting and highly effective drug delivery, which will find great potential as a universal drug delivery platform for targeting treatment of various bone diseases.
Collapse
Affiliation(s)
- Yu Chen
- Department of Orthopedic Trauma and Microsurgery, Zhongnan Hospital of Wuhan University, Wuhan 430071, P. R. China
| | - Qi-Wen Chen
- Key Laboratory of Biomedical Polymers of Ministry of Education & Department of Chemistry, Wuhan University, Wuhan 430072, P. R. China
- Department of Pharmaceutical Engineering, College of Chemistry and Chemical Engineering, Central South University, Changsha 410083, P. R. China
| | - Fang-Sheng Fu
- Department of Orthopedic Trauma and Microsurgery, Zhongnan Hospital of Wuhan University, Wuhan 430071, P. R. China
| | - Hui-Yun Gu
- Department of Orthopedic Trauma and Microsurgery, Zhongnan Hospital of Wuhan University, Wuhan 430071, P. R. China
| | - Aixi Yu
- Department of Orthopedic Trauma and Microsurgery, Zhongnan Hospital of Wuhan University, Wuhan 430071, P. R. China
| | - Xian-Zheng Zhang
- Department of Orthopedic Trauma and Microsurgery, Zhongnan Hospital of Wuhan University, Wuhan 430071, P. R. China
- Key Laboratory of Biomedical Polymers of Ministry of Education & Department of Chemistry, Wuhan University, Wuhan 430072, P. R. China
| |
Collapse
|