1
|
Zhang Q, Wang Y, Zhu J, Zou M, Zhang Y, Wu H, Jin T. Specialized pro-resolving lipid mediators: a key player in resolving inflammation in autoimmune diseases. Sci Bull (Beijing) 2025; 70:778-794. [PMID: 39837719 DOI: 10.1016/j.scib.2024.07.049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Revised: 06/21/2024] [Accepted: 07/16/2024] [Indexed: 01/23/2025]
Abstract
Uncontrolled hyperactivation of the immune system is the central mechanism underlying the pathogenesis of autoimmune diseases. Timely control of the inflammatory response is essential to prevent inflammation progression and organ damage. Specialized pro-resolving lipid mediators (SPMs) are autacoid molecules derived from essential polyunsaturated fatty acids during acute inflammatory responses. They promote the resolution of inflammation and orchestrate endogenous reparative responses. The SPM superfamily includes lipoxins, resolvins, protectins, and maresins, as well as novel conjugates involved in tissue regeneration. Much work has been done focusing on the actions of SPMs in autoimmunity and has identified their deficiencies and therapeutic effects in autoimmune diseases. In this review, we provide a brief introduction of SPMs, summarize their effects on key cells involved in innate and adaptive immunity, and highlight their role and therapeutic potential in autoimmune diseases.
Collapse
Affiliation(s)
- Qingxiang Zhang
- Department of Neurology, Neuroscience Center, The First Hospital of Jilin University, Changchun 130000, China
| | - Ying Wang
- Department of Neurology, Neuroscience Center, The First Hospital of Jilin University, Changchun 130000, China
| | - Jie Zhu
- Department of Neurology, Neuroscience Center, The First Hospital of Jilin University, Changchun 130000, China; Department of Neurobiology, Care Sciences & Society, Karolinska Institute, Karolinska University Hospital Solna, Stockholm 17176, Sweden
| | - Meijuan Zou
- Department of Neurology, Neuroscience Center, The First Hospital of Jilin University, Changchun 130000, China
| | - Yuxin Zhang
- Department of Neurology, Neuroscience Center, The First Hospital of Jilin University, Changchun 130000, China
| | - Hao Wu
- Department of Neurology, Neuroscience Center, The First Hospital of Jilin University, Changchun 130000, China
| | - Tao Jin
- Department of Neurology, Neuroscience Center, The First Hospital of Jilin University, Changchun 130000, China.
| |
Collapse
|
2
|
Düsterhöft S, Greve JN, Garbers C. Investigating plasticity within the interleukin-6 family with AlphaFold-Multimer. Comput Struct Biotechnol J 2025; 27:946-959. [PMID: 40151527 PMCID: PMC11946507 DOI: 10.1016/j.csbj.2025.02.030] [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: 01/08/2025] [Revised: 02/24/2025] [Accepted: 02/25/2025] [Indexed: 03/29/2025] Open
Abstract
Cytokines are important soluble mediators that are involved in physiological and pathophysiological processes. Among them, members of the interleukin-6 (IL-6) family of cytokines have gained remarkable attention, because especially the name-giving cytokine IL-6 has been shown to be an excellent target to treat inflammatory and autoimmune diseases. The IL-6 family consists of nine members, which activate their target cells via combinations of non-signaling α- and/or signal-transducing β-receptors. While some receptor combinations are exclusively used by a single cytokine, other cytokine receptor combinations are used by multiple cytokines. Research in recent years unraveled another level of complexity: several cytokine cannot only signal via their canonical receptors, but can bind to and signal via additional α- and/or β-receptors, albeit with less affinity. While several examples of such cytokine plasticity have been reported, a systematic analysis of this phenomenon is lacking. The development of artificial intelligence programs like AlphaFold allows the computational analysis of protein complexes in a systematic manner. Here, we develop a analysis pipeline for cytokine:cytokine receptor interaction and show that AlphaFold-Multimer correctly predicts the canonical ligands of the IL-6 family. However, AlphaFold-Multimer does not provide sufficient insight to conclusively predict alternative, low-affinity ligands for receptors within the IL-6 family.
Collapse
Affiliation(s)
- Stefan Düsterhöft
- Institute of Molecular Pharmacology, Medical Faculty, RWTH Aachen University, Aachen, Germany
| | - Johannes N. Greve
- Institute for Biophysical Chemistry, Hannover Medical School, Fritz-Hartmann-Centre for Medical Research, Hannover, Germany
| | - Christoph Garbers
- Institute of Clinical Biochemistry, Hannover Medical School, Hannover 30625, Germany
| |
Collapse
|
3
|
Iqbal MW, Shahab M, Ullah Z, Zheng G, Anjum I, Shazly GA, Mengistie AA, Sun X, Yuan Q. Integrating machine learning and structure-based approaches for repurposing potent tyrosine protein kinase Src inhibitors to treat inflammatory disorders. Sci Rep 2025; 15:1836. [PMID: 39805859 PMCID: PMC11730308 DOI: 10.1038/s41598-024-83767-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2024] [Accepted: 12/17/2024] [Indexed: 01/16/2025] Open
Abstract
Tyrosine-protein kinase Src plays a key role in cell proliferation and growth under favorable conditions, but its overexpression and genetic mutations can lead to the progression of various inflammatory diseases. Due to the specificity and selectivity problems of previously discovered inhibitors like dasatinib and bosutinib, we employed an integrated machine learning and structure-based drug repurposing strategy to find novel, targeted, and non-toxic Src kinase inhibitors. Different machine learning models including random forest (RF), k-nearest neighbors (K-NN), decision tree, and support vector machine (SVM), were trained using already available bioactivity data of Src kinase targeting compounds. The performance evaluation of these models demonstrated SVM as the best model, which was further utilized to shortlist 51 highly potent compounds by screening an FDA-approved library of 1040 drugs. Molecular docking and molecular dynamic simulation were subsequently employed to evaluate the binding affinity and stability of the proposed compounds. Orlistat, acarbose and afatinib were identified as the potent leads, demonstrating stable conformations and stronger interactions, validated by root mean square deviation (RMSD), root mean square fluctuation (RMSF), radius of gyration (RoG), and hydrogen bond analyses. Molecular Mechanics/Generalized Born Surface Area (MMGBSA) analysis validated their binding affinities by providing comparably lower binding free energies for orlistat (- 33.4743 ± 3.8908), acarbose (- 19.5455 ± 5.4702), and afatinib (- 36.4944 ± 5.4929) than the control, dasatinib (- 13.7785 ± 5.8058). Finally, toxicity analysis revealed orlistat and acarbose as the possible safer therapeutics by eliminating afatinib as it showed significant toxicity concerns. Our investigation supports the advance computational methods utilization in the field of drug discovery and suggest further experimental validation of proposed inhibitors of Src kinase for their safer use against inflammatory diseases. The ultimate aim of this study is to advance the development of effective treatments for inflammatory diseases, linked with Src overexpression.
Collapse
Affiliation(s)
- Muhammad Waleed Iqbal
- State Key Laboratory of Chemical Resources Engineering, Beijing University of Chemical Technology, Beijing, 100029, People's Republic of China
| | - Muhammad Shahab
- State Key Laboratory of Chemical Resources Engineering, Beijing University of Chemical Technology, Beijing, 100029, People's Republic of China
| | - Zakir Ullah
- State Key Laboratory of Chemical Resources Engineering, Beijing University of Chemical Technology, Beijing, 100029, People's Republic of China
| | - Guojun Zheng
- State Key Laboratory of Chemical Resources Engineering, Beijing University of Chemical Technology, Beijing, 100029, People's Republic of China
| | - Irfan Anjum
- Department of Basic Medical Sciences, Shifa College of Pharmaceutical Sciences, Shifa Tameer-e-Millat University, Islamabad, 44000, Pakistan
| | - Gamal A Shazly
- Department of Pharmaceutics, College of Pharmacy, King Saud University, Riyadh, 11451, Saudi Arabia
| | | | - Xinxiao Sun
- State Key Laboratory of Chemical Resources Engineering, Beijing University of Chemical Technology, Beijing, 100029, People's Republic of China.
| | - Qipeng Yuan
- State Key Laboratory of Chemical Resources Engineering, Beijing University of Chemical Technology, Beijing, 100029, People's Republic of China.
| |
Collapse
|
4
|
Xiong W, Li Y, Hu L, He G, Huang J. Risks of malignancies related to disease-modifying antirheumatic drugs in rheumatoid arthritis: a pharmacovigilance analysis using the FAERS database. Front Pharmacol 2024; 15:1458500. [PMID: 39605908 PMCID: PMC11598350 DOI: 10.3389/fphar.2024.1458500] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Accepted: 10/30/2024] [Indexed: 11/29/2024] Open
Abstract
Objectives Over the years when disease-modifying antirheumatic drugs (DMARDs) have been used in rheumatoid arthritis patients, reports of malignancies have emerged. This study aims to investigate the association between malignancies and DMARDs by using data extracted from the Food and Drug Administration Adverse Event Reporting System (FAERS). Methods FAERS data (January 2019 to December 2023) were reviewed. For each drug-event pair, the disproportionality analysis was conducted to evaluate the risk of malignancy. Multivariate logistic regression was implemented to mitigate potential biases. Moreover, the time to onset of malignancy was also evaluated. Results We conducted a detailed search for rheumatoid arthritis indications and identified a total of 17,412 adverse event reports associated with malignancies, with selective DMARDs designated as the role code "primary suspect". At the preferred term level, there were 198 positive signals, among which the lower limit of the 95% confidence interval for the information component is 3.55 for squamous cell carcinoma of the skin, 2.39 for breast cancer, and 2.27 for lymphoproliferative disorder. In comparison to other DMARDs, targeted synthetic DMARDs were associated with a broader range of malignancies at both preferred term and Standardized MedDRA Queries levels. The number of adverse events reported in female patients is approximately 2-3 times higher than men, and the median age across the population was approximately 62 years. In terms of onset time, the conventional synthetic DMRADs exhibited a relatively longer median time, ranging from 3.58 to 7.08 years, while the targeted synthetic DMARDs demonstrated a shorter median time of 0.83-1.67 years. Conclusion Our study uncovers varying degrees of malignancy risks related to DMARDs, with a significantly higher risk observed in targeted synthetic DMARDs. Additionally, novel malignancy signals, not documented in product labels, have been detected. In the future, further research will be necessary to validate our findings.
Collapse
Affiliation(s)
- Wan Xiong
- Department of Pharmacy, The Affiliated Changsha Hospital of Xiangya School of Medicine, Central South University, Changsha, China
- Department of Pharmacy, The First Hospital of Changsha, Changsha, China
| | - Yilin Li
- Department of Information and Digital Technology, PowerChina Zhongnan Engineering Corporation Limited, Changsha, China
| | - Lin Hu
- Department of Pharmacy, The Affiliated Changsha Hospital of Xiangya School of Medicine, Central South University, Changsha, China
- Department of Pharmacy, The First Hospital of Changsha, Changsha, China
| | - Gefei He
- Department of Pharmacy, The Affiliated Changsha Hospital of Xiangya School of Medicine, Central South University, Changsha, China
- Department of Pharmacy, The First Hospital of Changsha, Changsha, China
| | - Juanjuan Huang
- Department of Pharmacy, The Affiliated Changsha Hospital of Xiangya School of Medicine, Central South University, Changsha, China
- Department of Pharmacy, The First Hospital of Changsha, Changsha, China
| |
Collapse
|
5
|
Zheng DC, Hu JQ, Mai CT, Huang L, Zhou H, Yu LL, Xie Y. Liver X receptor inverse agonist SR9243 attenuates rheumatoid arthritis via modulating glycolytic metabolism of macrophages. Acta Pharmacol Sin 2024; 45:2354-2365. [PMID: 38987388 PMCID: PMC11489696 DOI: 10.1038/s41401-024-01315-7] [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: 12/06/2023] [Accepted: 05/15/2024] [Indexed: 07/12/2024]
Abstract
Liver X receptors (LXRs) which link lipid metabolism and inflammation, were overexpressed in experimental rheumatoid arthritis (RA) rats as observed in our previous studies, while suppression of LXRα by silybin ameliorates arthritis and abnormal lipid metabolism. However, the role of LXRs in RA remains undefined. In this study, we investigated the inhibition role of LXRs in the polarization and activation of M1 macrophage by using a special LXRs inverse agonist SR9243, which led to ameliorating the progression of adjuvant-induced arthritis (AIA) in rats. Mechanistically, SR9243 disrupted the LPS/IFN-γ-induced Warburg effect in M1 macrophages, while glycolysis inhibitor 2-DG attenuated the inhibition effect of SR9243 on M1 polarization and the cytokines expression of M1 macrophages including iNOS, TNF-α, and IL-6 in vitro. Furthermore, SR9243 downregulated key glycolytic enzymes, including LDH-A, HK2, G6PD, GLUT1, and HIF-1α in M1 macrophages, which is mediated by increased phosphorylation of AMPK (Thr172) and reduced downstream phosphorylation of mTOR (Ser2448). Importantly, gene silencing of LXRs compromises the inhibition effect of SR9243 on M1 macrophage polarization and activation. Collectively, for the first time, our findings suggest that the LXR inverse agonist SR9243 mitigates adjuvant-induced rheumatoid arthritis and protects against bone erosion by inhibiting M1 macrophage polarization and activation through modulation of glycolytic metabolism via the AMPK/mTOR/HIF-1α pathway.
Collapse
Affiliation(s)
- De-Chong Zheng
- State Key Laboratory of Traditional Chinese Medicine Syndrome, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, 510006, China
- State Key Laboratory of Quality Research in Chinese Medicines, Macau University of Science and Technology, Macau, China
| | - Jia-Qin Hu
- State Key Laboratory of Traditional Chinese Medicine Syndrome, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, 510006, China
- State Key Laboratory of Quality Research in Chinese Medicines, Macau University of Science and Technology, Macau, China
| | - Chu-Tian Mai
- State Key Laboratory of Quality Research in Chinese Medicines, Macau University of Science and Technology, Macau, China
| | - Li Huang
- State Key Laboratory of Traditional Chinese Medicine Syndrome, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, 510006, China
| | - Hua Zhou
- State Key Laboratory of Traditional Chinese Medicine Syndrome, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, 510006, China
| | - Li-Li Yu
- State Key Laboratory of Quality Research in Chinese Medicines, Macau University of Science and Technology, Macau, China.
| | - Ying Xie
- State Key Laboratory of Traditional Chinese Medicine Syndrome, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, 510006, China.
| |
Collapse
|
6
|
Hao T, Tsang YP, Yin M, Mao Q, Unadkat JD. Dysregulation of Human Hepatic Drug Transporters by Proinflammatory Cytokines. J Pharmacol Exp Ther 2024; 391:82-90. [PMID: 39103232 DOI: 10.1124/jpet.123.002019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2023] [Revised: 07/23/2024] [Accepted: 07/29/2024] [Indexed: 08/07/2024] Open
Abstract
Proinflammatory cytokines, elevated during inflammation caused by infection and/or autoimmune disorders, result in reduced clearance of drugs eliminated primarily by cytochrome P450 enzymes (CYPs). However, the effect of cytokines on hepatic drug transporter expression or activity has not been well-studied. Here, using plated human hepatocytes (PHHs; n = 3 lots), we investigated the effect of interleukin (IL)-6, IL-1β, tumor necrosis factor-α (TNF-α), and interferon-γ (IFN-γ), on the mRNA expression and activity of hepatic drug transporters. PHHs were incubated for 72 hours at their pathophysiologically relevant plasma concentrations, both individually (0.01, 0.1, 1, 10 ng/ml) or as a cocktail (i.e., when each was combined at 0.1 or 1 ng/ml). Following cytokine cocktail exposure (1 ng/ml), significant downregulation of mRNA expression of organic anion transporting polypeptide 1B1 (OATP1B1), OATP1B3, sodium/taurocholate cotransporting polypeptide (NTCP), breast cancer resistance protein (BCRP), P-glycoprotein (P-gp), multidrug and toxin extrusion protein 1, multidrug resistance proteins (MRP) 2, 3, and 4 was observed. While the mRNA expression of organic anion transporter (OAT) 2 and organic cation transporter (OCT) 1 was downregulated in two lots, it was upregulated in one lot. In agreement (mostly), the 1 ng/ml cytokine cocktail reduced OATP1B1/3, OATP2B1, OAT2, OCT1, and NTCP activity by 75%, 44%, 82%, 47%, and 80%, respectively. Interestingly, upregulation of OAT2 and OCT1 mRNA in one donor did not translate into the same directional change in activity. Although significant interlot variability was observed, in general, the above effects, using individual cytokines, could be attributed to IL-1β, TNF-α, and IFN-γ. SIGNIFICANCE STATEMENT: To date, this is the first comprehensive study to investigate the effect of four major proinflammatory cytokines, both individually and as a cocktail, on the mRNA expression and activity of human hepatic drug transporters. The data obtained can be used in the future to predict transporter-mediated drug clearance changes during inflammation through physiologically based pharmacokinetic modeling and simulation.
Collapse
Affiliation(s)
- Tianran Hao
- Department of Pharmaceutics, School of Pharmacy, University of Washington, Seattle, Washington
| | - Yik Pui Tsang
- Department of Pharmaceutics, School of Pharmacy, University of Washington, Seattle, Washington
| | - Mengyue Yin
- Department of Pharmaceutics, School of Pharmacy, University of Washington, Seattle, Washington
| | - Qingcheng Mao
- Department of Pharmaceutics, School of Pharmacy, University of Washington, Seattle, Washington
| | - Jashvant D Unadkat
- Department of Pharmaceutics, School of Pharmacy, University of Washington, Seattle, Washington
| |
Collapse
|
7
|
Rohini S, Sharma UR, Vinutha M, Shreelaxmi D, Vada S, Janandri S, Haribabu T, Taj N, Gayathri SV, Ghara A, Mudagal MP. Rheumatoid arthritis-associated complications during pregnancy and its effect on offspring: comprehensive review. Inflammopharmacology 2024; 32:1-17. [PMID: 38691248 DOI: 10.1007/s10787-024-01482-3] [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: 02/22/2024] [Accepted: 04/05/2024] [Indexed: 05/03/2024]
Abstract
This study comprehensively explores the complexities of rheumatoid arthritis during pregnancy and its impact on offspring. Through an extensive review of existing literature, we investigate maternal and fetal risks, including adverse pregnancy outcomes and developmental issues in offspring. Utilizing reputable databases such as PubMed, Google Scholar, and Science Direct, we meticulously examined studies exploring the connection between rheumatoid arthritis and pregnancy complications, with a focus on outcomes for offspring. We excluded studies lacking sufficient data or peer review. Synthesizing findings from selected studies, we identified common themes and patterns, presenting results in a clear, organized manner. Our examination reveals a heightened likelihood of preterm birth and preeclampsia among pregnant individuals with rheumatoid arthritis, often correlated with disease activity. Furthermore, we highlight the impact on fetal and neonatal outcomes, such as low birth weight, underscoring the importance of meticulous disease management throughout pregnancy. Balancing the necessity of disease-modifying agents with potential risks, and consideration of medication safety is paramount. A multidisciplinary approach involving rheumatologists and obstetricians is crucial for optimizing outcomes. In conclusion, this synthesis underscores the nuanced challenges of rheumatoid arthritis in pregnancy. A comprehensive understanding and personalized, multidisciplinary approach to an organization is essential for informed decision-making in clinical practice. Our review contributes to ongoing discourse, providing insights for enhanced patient care and guiding future research endeavors.
Collapse
Affiliation(s)
- S Rohini
- Department of Pharmacology, Acharya & BM Reddy College of Pharmacy, Acharya Dr. Sarvepalli Radhakrishna Road, Achit Nagar (Post), Soldevanahalli, Bengaluru, 560090, India
| | - Uday Raj Sharma
- Department of Pharmacology, Acharya & BM Reddy College of Pharmacy, Acharya Dr. Sarvepalli Radhakrishna Road, Achit Nagar (Post), Soldevanahalli, Bengaluru, 560090, India.
| | - M Vinutha
- Department of Pharmacology, Acharya & BM Reddy College of Pharmacy, Acharya Dr. Sarvepalli Radhakrishna Road, Achit Nagar (Post), Soldevanahalli, Bengaluru, 560090, India
| | - D Shreelaxmi
- Department of Pharmacology, Acharya & BM Reddy College of Pharmacy, Acharya Dr. Sarvepalli Radhakrishna Road, Achit Nagar (Post), Soldevanahalli, Bengaluru, 560090, India
| | - Surendra Vada
- Department of Pharmacology, Acharya & BM Reddy College of Pharmacy, Acharya Dr. Sarvepalli Radhakrishna Road, Achit Nagar (Post), Soldevanahalli, Bengaluru, 560090, India
| | - Suresh Janandri
- Department of Pharmacology, Acharya & BM Reddy College of Pharmacy, Acharya Dr. Sarvepalli Radhakrishna Road, Achit Nagar (Post), Soldevanahalli, Bengaluru, 560090, India
| | - T Haribabu
- Department of Pharmacology, Acharya & BM Reddy College of Pharmacy, Acharya Dr. Sarvepalli Radhakrishna Road, Achit Nagar (Post), Soldevanahalli, Bengaluru, 560090, India
| | - Nageena Taj
- Department of Pharmacology, Acharya & BM Reddy College of Pharmacy, Acharya Dr. Sarvepalli Radhakrishna Road, Achit Nagar (Post), Soldevanahalli, Bengaluru, 560090, India
| | - S V Gayathri
- Department of Pharmacology, Acharya & BM Reddy College of Pharmacy, Acharya Dr. Sarvepalli Radhakrishna Road, Achit Nagar (Post), Soldevanahalli, Bengaluru, 560090, India
| | - Abhishek Ghara
- Department of Pharmacology, Acharya & BM Reddy College of Pharmacy, Acharya Dr. Sarvepalli Radhakrishna Road, Achit Nagar (Post), Soldevanahalli, Bengaluru, 560090, India
| | - Manjunatha P Mudagal
- Department of Pharmacology, Acharya & BM Reddy College of Pharmacy, Acharya Dr. Sarvepalli Radhakrishna Road, Achit Nagar (Post), Soldevanahalli, Bengaluru, 560090, India
| |
Collapse
|
8
|
Zhang Y, Zhang J, Liu Y, Ren S, Tao N, Meng F, Cao Q, Liu R. High fat diet increases the severity of collagen-induced arthritis in mice by altering the gut microbial community. Adv Rheumatol 2024; 64:44. [PMID: 38816873 DOI: 10.1186/s42358-024-00382-y] [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: 08/11/2023] [Accepted: 05/06/2024] [Indexed: 06/01/2024] Open
Abstract
OBJECTIVES Research has demonstrated that obesity may be associated with rheumatoid arthritis (RA). In addition, gut microbiota and its metabolites contribute to the occurrence and development of RA and obesity. However, the mechanism by which obesity affects RA remains unclear. In this study, we aimed to investigate whether gut microbiota and their metabolites alter the effects of high fat diet (HFD) on the severity of collagen-induced arthritis (CIA) in mice. METHODS Briefly, mice were divided into normal group (N), CIA model group (C), HFD group (T), and HFD CIA group (CT). Hematoxylin and Eosin staining(HE) and Safranin O-fast green staining were conducted, and levels of blood lipid and inflammatory cytokines were measured. 16S rDNA sequencing technique and liquid chromatography-mass spectrometry (LC-MS)-based metabolomics were performed to explore changes in the microbiota structure to further reveal the pathomechanism of HFD on CIA. RESULTS HFD aggravated the severity of CIA in mice. The CT group had the highest proportion of microbial abundance of Blautia, Oscillibacter, Ruminiclostridium-9, and Lachnospiraceae UCG 006 at the genus level, but had a lower proportion of Alistipes. Additionally, the fecal metabolic phenotype of the combined CT group shows significant changes, with differential metabolites enriched in 9 metabolic pathways, including primary bile acid biosynthesis, arginine biosynthesis, sphingolipid metabolism, purine metabolism, linoleic acid metabolism, oxytocin signaling pathway, aminoacyl-tRNA biosynthesis, the pentose phosphate pathway, and sphingolipid signaling pathway. Correlation analysis revealed that some of the altered gut microbiota genera were strongly correlated with changes in fecal metabolites, total cholesterol (TC), triglyceride (TG), and inflammatory cytokine levels. CONCLUSIONS This study shows that HFD may aggravate inflammatory reaction in CIA mice by altering the gut microbiota and metabolic pathways.
Collapse
Affiliation(s)
- Yang Zhang
- The First Hospital of China Medical University, Shenyang, 110002, Liaoning, China
| | - Jie Zhang
- The First Hospital of China Medical University, Shenyang, 110002, Liaoning, China
| | - Yantong Liu
- The First Hospital of China Medical University, Shenyang, 110002, Liaoning, China
| | - Shuang Ren
- The First Hospital of China Medical University, Shenyang, 110002, Liaoning, China
| | - Ning Tao
- The First Hospital of China Medical University, Shenyang, 110002, Liaoning, China
| | - Fanyan Meng
- The First Hospital of China Medical University, Shenyang, 110002, Liaoning, China
| | - Qi Cao
- Liaoning University of Traditional Chinese Medicine, Shenyang, 110001, Liaoning, China
| | - Ruoshi Liu
- The First Hospital of China Medical University, Shenyang, 110002, Liaoning, China.
| |
Collapse
|
9
|
Wang Y, Guo R, Zou M, Jiang L, Kong L, Zhao S, Zhang X, Wang W, Xu B. Combined ROS Sensitive Folate Receptor Targeted Micellar Formulations of Curcumin Effective Against Rheumatoid Arthritis in Rat Model. Int J Nanomedicine 2024; 19:4217-4234. [PMID: 38766660 PMCID: PMC11100960 DOI: 10.2147/ijn.s458957] [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: 01/10/2024] [Accepted: 04/29/2024] [Indexed: 05/22/2024] Open
Abstract
Introduction Rheumatoid arthritis (RA) is an inflammatory immune-mediated disease that involves synovitis, cartilage destruction, and even joint damage. Traditional agents used for RA therapy remain unsatisfactory because of their low efficiency and obvious adverse effects. Therefore, we here established RA microenvironment-responsive targeted micelles that can respond to the increase in reactive oxygen species (ROS) levels in the joint and improve macrophage-specific targeting of loaded drugs. Methods We here prepared ROS-responsive folate-modified curcumin micelles (TK-FA-Cur-Ms) in which thioketal (TK) was used as a ROS-responsive linker for modifying polyethylene glycol 5000 (PEG5000) on the micellar surface. When micelles were in the ROS-overexpressing inflammatory microenvironment, the PEG5000 hydration layer was shed, and the targeting ligand FA was exposed, thereby enhancing cellular uptake by macrophages through active targeting. The targeting, ROS sensitivity and anti-inflammatory properties of the micelles were assessed in vitro. Collagen-induced arthritis (CIA) rats model was utilized to investigate the targeting, expression of serum inflammatory factors and histology change of the articular cartilage by micelles in vivo. Results TK-FA-Cur-Ms had a particle size of 90.07 ± 3.44 nm, which decreased to 78.87 ± 2.41 nm after incubation with H2O2. The micelles exhibited in vitro targeting of RAW264.7 cells and significantly inhibited inflammatory cytokine levels. Pharmacodynamic studies have revealed that TK-FA-Cur-Ms prolonged the drug circulation and exhibited augmented cartilage-protective and anti-inflammatory effects in vivo. Conclusion The unique ROS-responsive targeted micelles with targeting, ROS sensitivity and anti-inflammatory properties were successfully prepared and may offer an effective therapeutic strategy against RA.
Collapse
Affiliation(s)
- Yuanyuan Wang
- Department of Pharmacy, Affiliated Zhongshan Hospital of Dalian University, Dalian, People’s Republic of China
| | - Ruibo Guo
- School of Pharmacy, Liaoning University of Traditional Chinese Medicine, Dalian, People’s Republic of China
| | - Ming Zou
- Department of Pharmacy, Affiliated Zhongshan Hospital of Dalian University, Dalian, People’s Republic of China
| | - Lingling Jiang
- Department of Pharmacy, Affiliated Zhongshan Hospital of Dalian University, Dalian, People’s Republic of China
| | - Liang Kong
- School of Pharmacy, Liaoning University of Traditional Chinese Medicine, Dalian, People’s Republic of China
| | - Sen Zhao
- Department of Pharmacy, Affiliated Zhongshan Hospital of Dalian University, Dalian, People’s Republic of China
| | - Xuan Zhang
- Department of Pharmacy, Affiliated Zhongshan Hospital of Dalian University, Dalian, People’s Republic of China
| | - Wei Wang
- Department of Pharmacy, Affiliated Zhongshan Hospital of Dalian University, Dalian, People’s Republic of China
| | - Baoli Xu
- Department of Pharmacy, Affiliated Zhongshan Hospital of Dalian University, Dalian, People’s Republic of China
| |
Collapse
|
10
|
Li Z, Zhang X, Li K, Li F, Kou J, Wang Y, Wei X, Sun Y, Jing Y, Song Y, Yu Q, Yu H, Wang S, Chen S, Wang Y, Xie S, Zhu X, Zhan Y, Sun G, Ni Y. IL-36 antagonism blunts the proliferation and migration of oral squamous cell carcinoma cells. Cell Signal 2024; 117:111096. [PMID: 38346528 DOI: 10.1016/j.cellsig.2024.111096] [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: 12/07/2023] [Revised: 01/28/2024] [Accepted: 02/09/2024] [Indexed: 02/17/2024]
Abstract
IL-36 is known to mediate inflammation and fibrosis. Nevertheless, IL-36 signalling axis has also been implicated in cancer, although understanding of exact contribution of IL-36 to cancer progression is very limited, partly due to existence of multiple IL-36 ligands with agonistic and antagonistic function. Here we explored the role of IL-36 in oral squamous cell carcinoma (OSCC). Firstly, we analyzed expression of IL-36 ligands and receptor and found that the expression of IL-36γ was significantly higher in head and neck cancer (HNSCC) than that of normal tissues, and that the high expression of IL-36γ predicted poor clinical outcomes. Secondly, we investigated the direct effect of IL-36γ on OSCC cells and found that IL-36γ stimulated proliferation of OSCC cells with high expression of IL-36R expression. Interestingly, IL-36γ also promoted migration of OSCC cells with low to high IL-36R expression. Critically, both proliferation and migration of OSCC cells induced by IL-36γ were abrogated by anti-IL-36R mAb. Fittingly, RNA sequence analysis revealed that IL-36γ regulated genes involved in cell cycle and cell division. In summary, our results showed that IL-36γ can be a tumor-promoting factor, and targeting of IL-36R signalling may be a beneficial targeted therapy for patients with abnormal IL-36 signalling.
Collapse
Affiliation(s)
- Zihui Li
- Central Laboratory of Stomatology, Nanjing Stomatological Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China; Department of Oral and Maxillofacial Surgery, Nanjing Stomatological Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Xiaoxin Zhang
- Central Laboratory of Stomatology, Nanjing Stomatological Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Ke Li
- Central Laboratory of Stomatology, Nanjing Stomatological Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Fuyan Li
- Central Laboratory of Stomatology, Nanjing Stomatological Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Jiahao Kou
- Central Laboratory of Stomatology, Nanjing Stomatological Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Yuhan Wang
- Central Laboratory of Stomatology, Nanjing Stomatological Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Xiaoyue Wei
- Drug Discovery, Shanghai Huaota Biopharmaceutical Co. Ltd., Shanghai, China
| | - Yawei Sun
- Department of Oral and Maxillofacial Surgery, Nanjing Stomatological Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Yue Jing
- Central Laboratory of Stomatology, Nanjing Stomatological Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Yuxian Song
- Central Laboratory of Stomatology, Nanjing Stomatological Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - QiuYa Yu
- Central Laboratory of Stomatology, Nanjing Stomatological Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Haijia Yu
- Drug Discovery, Shanghai Huaota Biopharmaceutical Co. Ltd., Shanghai, China
| | - Shuai Wang
- Central Laboratory of Stomatology, Nanjing Stomatological Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Shi Chen
- Drug Discovery, Shanghai Huaota Biopharmaceutical Co. Ltd., Shanghai, China
| | - Yangtin Wang
- Drug Discovery, Shanghai Huaota Biopharmaceutical Co. Ltd., Shanghai, China
| | - Simin Xie
- Drug Discovery, Shanghai Huaota Biopharmaceutical Co. Ltd., Shanghai, China
| | - Xiangyang Zhu
- Drug Discovery, Shanghai Huaota Biopharmaceutical Co. Ltd., Shanghai, China
| | - Yifan Zhan
- Drug Discovery, Shanghai Huaota Biopharmaceutical Co. Ltd., Shanghai, China.
| | - Guowen Sun
- Department of Oral and Maxillofacial Surgery, Nanjing Stomatological Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China.
| | - Yanhong Ni
- Central Laboratory of Stomatology, Nanjing Stomatological Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China.
| |
Collapse
|
11
|
Hernandez-Suarez L, Diez-Martin E, Egiguren-Ortiz J, Fernandez R, Etxebarria A, Astigarraga E, Miguelez C, Ramirez-Garcia A, Barreda-Gómez G. Serological Antibodies against Kidney, Liver, and Spleen Membrane Antigens as Potential Biomarkers in Patients with Immune Disorders. Int J Mol Sci 2024; 25:2025. [PMID: 38396703 PMCID: PMC10888476 DOI: 10.3390/ijms25042025] [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: 12/20/2023] [Revised: 01/30/2024] [Accepted: 02/05/2024] [Indexed: 02/25/2024] Open
Abstract
Immune disorders arise from complex genetic and environmental factors, which lead to dysregulation at the cellular and inflammatory levels and cause tissue damage. Recent research highlights the crucial role of reactive antibodies in autoimmune diseases and graft rejection, but their complex determination poses challenges for clinical use. Therefore, our study aimed to ascertain whether the presence of reactive antibodies against membrane antigens in tissues from both animal models and humans could serve as biomarkers in patients with autoimmune disorders. To address this issue, we examined the binding profile of serological antibodies against a diverse panel of cell membranes from the spleen, liver, and kidney tissues of monkeys, rats, and humans. After developing the cell membrane microarrays, human sera were immunologically assayed. The study was first conducted on sera from two groups, healthy subjects and patients with inflammatory and autoimmune disorders, and then optimized for kidney transplant patient sera. A significant increase in antibody reactivity against specific monkey kidney and spleen membranes was observed in the serum of patients with lupus nephritis, while kidney transplant patients showed a significant enhancement against human tissues and human embryonic kidney 293 cells. These results show the potential importance for clinical and basic research purposes of studying the presence of specific IgG against membrane antigens in patients' serum as potential biomarkers of immune disorders. However, it is important to note that these results need to be verified in further studies with a larger sample size to confirm their relevance.
Collapse
Affiliation(s)
- Leidi Hernandez-Suarez
- Department of Research and Development, IMG Pharma Biotech S.L., 48170 Zamudio, Spain; (L.H.-S.); (E.D.-M.); (J.E.-O.); (R.F.); (A.E.); (E.A.)
- Department of Immunology, Microbiology and Parasitology, Faculty of Science and Technology, University of the Basque Country (UPV/EHU), 48940 Leioa, Spain;
| | - Eguzkiñe Diez-Martin
- Department of Research and Development, IMG Pharma Biotech S.L., 48170 Zamudio, Spain; (L.H.-S.); (E.D.-M.); (J.E.-O.); (R.F.); (A.E.); (E.A.)
- Department of Immunology, Microbiology and Parasitology, Faculty of Science and Technology, University of the Basque Country (UPV/EHU), 48940 Leioa, Spain;
| | - June Egiguren-Ortiz
- Department of Research and Development, IMG Pharma Biotech S.L., 48170 Zamudio, Spain; (L.H.-S.); (E.D.-M.); (J.E.-O.); (R.F.); (A.E.); (E.A.)
- Department of Pharmacology, Faculty of Medicine and Nursing, University of the Basque Country (UPV/EHU), 48940 Leioa, Spain;
| | - Roberto Fernandez
- Department of Research and Development, IMG Pharma Biotech S.L., 48170 Zamudio, Spain; (L.H.-S.); (E.D.-M.); (J.E.-O.); (R.F.); (A.E.); (E.A.)
| | - Aitor Etxebarria
- Department of Research and Development, IMG Pharma Biotech S.L., 48170 Zamudio, Spain; (L.H.-S.); (E.D.-M.); (J.E.-O.); (R.F.); (A.E.); (E.A.)
| | - Egoitz Astigarraga
- Department of Research and Development, IMG Pharma Biotech S.L., 48170 Zamudio, Spain; (L.H.-S.); (E.D.-M.); (J.E.-O.); (R.F.); (A.E.); (E.A.)
| | - Cristina Miguelez
- Department of Pharmacology, Faculty of Medicine and Nursing, University of the Basque Country (UPV/EHU), 48940 Leioa, Spain;
- Neurodegenerative Diseases Group, BioBizkaia Health Research Institute, 48940 Barakaldo, Spain
| | - Andoni Ramirez-Garcia
- Department of Immunology, Microbiology and Parasitology, Faculty of Science and Technology, University of the Basque Country (UPV/EHU), 48940 Leioa, Spain;
| | - Gabriel Barreda-Gómez
- Department of Research and Development, IMG Pharma Biotech S.L., 48170 Zamudio, Spain; (L.H.-S.); (E.D.-M.); (J.E.-O.); (R.F.); (A.E.); (E.A.)
| |
Collapse
|
12
|
Fang W, Liu J, Zhang F, Pang C, Li X. A novel cholesterol metabolism-related ferroptosis pathway in hepatocellular carcinoma. Discov Oncol 2024; 15:7. [PMID: 38191842 PMCID: PMC10774324 DOI: 10.1007/s12672-023-00822-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Accepted: 11/09/2023] [Indexed: 01/10/2024] Open
Abstract
BACKGROUND Emerging studies have reported the contribution of cholesterol to hepatocellular carcinoma (HCC) progression. However, the specific role and mechanism of cholesterol metabolism on spontaneous and progressive HCC development from the point of view of ferroptosis are still worth exploring. The present study aimed to reveal a novel mechanism of cholesterol metabolism-related ferroptosis in hepatocellular carcinoma cells. METHODS Two microarray datasets (GSE25097, GSE22058) related to HCC were downloaded from Gene Expression Omnibus (GEO) datasets. Metabolomics analysis was performed by ultra performance liquid chromatography - tandem mass spectrometer (UPLC-MS/MS). The cholesterol-related proteins were downloaded from HMBD. Ferroptosis-related genes were extracted from FerrDb database. Data sets were separated into two groups. GSE25097 was used to identify ferroptosis-related genes, and GSE22058 was used to verify results. During these processes, chemical-protein interaction (CPI), protein-protein interaction (PPI), the Gene Ontology (GO), and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analyses were conducted. Multivariate logistic regression analysis was used to test the associated pathway. RESULTS We identified 8 differentially expressed ferroptosis-related genes (HAMP, PTGS2, IL1B, ALOX15B, CDKN2A, RRM2, NQO1 and KIF20A) and 4 differentially expressed cholesterol-related genes (LCAT, CH25H, CEL and CYP7A1). Furthermore, based on the predicted results with STITCH, we identified indomethacin and IL1B as the essential node for cholesterol-mediated ferroptosis in hepatocellular carcinoma cell. Multivariate logistic regression analysis showed the activities of plasma IL1B in liver cancer patients enrolled have been significantly affected by the level of plasma cholesterol (P < 0.001) and the test result of IL1B is a predictor variable causing the changes of serum Fe levels (P < 0.001). CONCLUSIONS Our findings shed new light on the association between cholesterol metabolism and ferroptosis in HCC, and suggest that IL1B is the necessary node for cholesterol to lead to ferroptosis process in HCC. Also, we identified the potential role of indomethacin in adjuvant therapy of HCC with complications of abnormal cholesterol metabolism.
Collapse
Affiliation(s)
- Weiwei Fang
- Department of Blood Transfusion, State Key Laboratory of Molecular Oncology, National Clinical Research Center for Cancer/Cancer Hospital, Institute/University, National Cancer Center, Chinese Academy of Medical Sciences and Peking Union Medical College, No.17, Nanli Road, Pan Jia Yuan, Beijing, 100021, China
| | - Jianyong Liu
- Department of Urology, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Institute/University/Hospital, Chinese Academy of Medical Sciences, No.1, Dahua Road, Dong Dan, 100730, Beijing, China
- Graduate School of Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, 100730, China
| | - Fanguo Zhang
- Excellence Future International Consulting Co, Ltd, Beijing, 101100, China
| | - Cheng Pang
- Department of Urology, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Institute/University/Hospital, Chinese Academy of Medical Sciences, No.1, Dahua Road, Dong Dan, 100730, Beijing, China.
| | - Xiying Li
- Department of Blood Transfusion, State Key Laboratory of Molecular Oncology, National Clinical Research Center for Cancer/Cancer Hospital, Institute/University, National Cancer Center, Chinese Academy of Medical Sciences and Peking Union Medical College, No.17, Nanli Road, Pan Jia Yuan, Beijing, 100021, China.
| |
Collapse
|
13
|
Saharan SS, Nagar P, Creasy KT, Stock EO, Feng J, Malloy MJ, Kane JP. Optimization of Smoking Classification by Applying Neural Network with Variable Importance Using Cytokine Biomarkers. PROCEEDINGS. INTERNATIONAL CONFERENCE ON COMPUTATIONAL SCIENCE AND COMPUTATIONAL INTELLIGENCE 2023; 2023:661-670. [PMID: 39512263 PMCID: PMC11542929 DOI: 10.1109/csci62032.2023.00115] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/15/2024]
Abstract
Cigarette smoking is a preventable epidemic that is a leading cause of death. It increases the risk of coronary heart disease, stroke, lung cancer, chronic obstructive lung diseases etc., multifold. Smoking tobacco is not only injurious to oneself but also to those who are exposed second hand. Smoking induces endothelial dysfunction via inflammatory cytokines that can be quantified precisely. Cytokines can be leveraged as powerful predictive biomarkers for identifying risk of potential diseases. Current advances in biomarker research are providing substantive evidence of the roles of cytokines in disease. This is driving precision-based diagnosis and translational therapeutic interventions. Innovative machine algorithms (ML) are pioneering transformative changes in the field of medical research. This research implements the Neural Networks (NN) algorithm to classify smokers versus non-smokers using 63 cytokines as predictor features. In addition to the fact that NN is a generative algorithm, which makes it a very powerful tool to achieve the objective of this differentiation, techniques like cross validation and hyperparameter tuning improve the efficacy of the algorithm. The study identified the 10 most impactful predictor features that contributed to the classification and then used these to characterize smokers versus non-smokers. Primarily, the study constructed and investigated two classifiers, of which the first implemented NN using the entire set of 63 cytokines and the second using 10 most informative cytokines. The performance of the first classifier, implemented using 63 cytokines, evaluated by area under receiver operating characteristic (AUROC), was extremely good with an AUROC score of .949 and 95% Confidence Interval (CI) (.923,.974). The second classifier that used the 10 most impactful cytokines with regard to the classification, demonstrated an exemplary performance, with an AUROC score of .995 and a 95% CI (.991,1). The 10 most impactful cytokines from the aspect of smoker versus non-smoker differentiation, listed in order of importance, include: I-TAC, IL-22, IL-2R, IL-3, HGF, IL-18, G-CSF-CSF-3, MIF, SDF-1alpha, MMP-1. To gain a deeper understanding of the effect of smoking on cytokine levels, a 2-sample independent t test was performed, ascertaining the statistical significance of the 63 cytokine levels in smokers versus non-smokers. Machine Learning using biomarkers such as cytokines will enhance the ability to predict the advent of a disease and its outcome, and lead to novel treatment strategies.
Collapse
Affiliation(s)
- Seema Singh Saharan
- Department of Clinical Pharmacy University of California San Francisco, USA
- UCSF Kane Lab San Francisco, USA
- UC Berkeley Extension Berkeley, USA
| | - Pankaj Nagar
- Department of Statistics University of Rajasthan Jaipur, India
| | - Kate Townsend Creasy
- Cardiovascular Research Institute, Department of Medicine University of California San Francisco, USA
| | - Eveline O Stock
- Cardiovascular Research Institute, Department of Medicine University of California San Francisco, USA
| | - James Feng
- Cardiovascular Research Institute, Department of Medicine University of California San Francisco, USA
| | - Mary J Malloy
- Cardiovascular Research Institute, Department of Medicine University of California San Francisco, USA
| | - John P Kane
- Cardiovascular Research Institute, Department of Medicine University of California San Francisco, USA
| |
Collapse
|
14
|
Yang L, Zhao W, Gong X, Yue D, Liu Y, Tian Y, Dong K. Exploring potential network pharmacology-and molecular docking-based mechanism of melittin in treating rheumatoid arthritis. Medicine (Baltimore) 2023; 102:e34728. [PMID: 37565866 PMCID: PMC10419517 DOI: 10.1097/md.0000000000034728] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Accepted: 07/21/2023] [Indexed: 08/12/2023] Open
Abstract
BACKGROUND Rheumatoid arthritis (RA) is a type of difficult-to-cure arthralgia with a worldwide prevalence. It severely affects people's living standards. For a long time, bee venom has been used to treat RA and has shown good results. Melittin is the main active component of bee venom used for RA treatment, but the molecular mechanism of melittin in RA treatments remains unclear. METHODS Potential melittin and RA targets were obtained from relevant databases, and common targets of melittin and RA were screened. The STRING database was used to build the PPI network and screen the core targets after visualization. The core targets were enriched by Gene Ontology functional annotation and Kyoto Encyclopedia of Genes and Genomes pathway. Finally, the binding of melittin to target proteins was evaluated through simulated molecular docking, which verified the reliability of the prediction results of network pharmacology. RESULTS In total, 138 melittin targets and 5795 RA targets were obtained from relevant databases, and 90 common targets were obtained through intersection. Eighteen core targets, such as STAT3, AKT1, tumor necrosis factor, and JUN, were screened out. Enrichment analysis results suggested that melittin plays an anti-RA role mainly through tumor necrosis factor, interleukin-17, toll-like receptors, and advanced glycation end products-RAGE signaling pathways, and pathogenic bacterial infection. Molecular docking results suggested that melittin has good docking activity with core target proteins. CONCLUSION RA treatment with melittin is the result of a multi-target and multi-pathway interaction. This study offers a theoretical basis and scientific evidence for further exploring melittin in RA therapy.
Collapse
Affiliation(s)
- Linfu Yang
- Yunnan Provincial Engineering and Research Center for Sustainable Utilization of Honey Bee Resources, Eastern Bee Research Institute, College of Animal Science and Technology, Yunnan Agricultural University, Kunming, China
| | - Wenzheng Zhao
- Yunnan Provincial Engineering and Research Center for Sustainable Utilization of Honey Bee Resources, Eastern Bee Research Institute, College of Animal Science and Technology, Yunnan Agricultural University, Kunming, China
| | - Xueyang Gong
- Yunnan Provincial Engineering and Research Center for Sustainable Utilization of Honey Bee Resources, Eastern Bee Research Institute, College of Animal Science and Technology, Yunnan Agricultural University, Kunming, China
| | - Dan Yue
- Yunnan Provincial Engineering and Research Center for Sustainable Utilization of Honey Bee Resources, Eastern Bee Research Institute, College of Animal Science and Technology, Yunnan Agricultural University, Kunming, China
| | - Yiqiu Liu
- Yunnan Provincial Engineering and Research Center for Sustainable Utilization of Honey Bee Resources, Eastern Bee Research Institute, College of Animal Science and Technology, Yunnan Agricultural University, Kunming, China
| | - Yakai Tian
- Yunnan Provincial Engineering and Research Center for Sustainable Utilization of Honey Bee Resources, Eastern Bee Research Institute, College of Animal Science and Technology, Yunnan Agricultural University, Kunming, China
| | - Kun Dong
- Yunnan Provincial Engineering and Research Center for Sustainable Utilization of Honey Bee Resources, Eastern Bee Research Institute, College of Animal Science and Technology, Yunnan Agricultural University, Kunming, China
| |
Collapse
|
15
|
Gligorić E, Igić R, Teofilović B, Grujić-Letić N. Phytochemical Screening of Ultrasonic Extracts of Salix Species and Molecular Docking Study of Salix-Derived Bioactive Compounds Targeting Pro-Inflammatory Cytokines. Int J Mol Sci 2023; 24:11848. [PMID: 37511606 PMCID: PMC10380267 DOI: 10.3390/ijms241411848] [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: 06/28/2023] [Revised: 07/17/2023] [Accepted: 07/20/2023] [Indexed: 07/30/2023] Open
Abstract
Willow bark (Salix spp., Salicaceae) is a traditional analgesic and antirheumatic herbal medicine. The aim of this study was to evaluate and compare the phytochemical and antioxidant profiles of leaf and bark extracts of six species of the genus Salix obtained by ultrasound-assisted extraction (UAE) and to examine the inhibitory potential of target bioactive compounds against two inflammatory mediators, tumor necrosis factor alpha (TNF-α) and interleukin 6 (IL-6), through in silico molecular docking. The total phenolic and flavonoid content of the extracts was estimated using spectrophotometric methods and the antioxidant activity using 2,2-diphenyl-1-picrylhydrazyl (DPPH•) and hydroxyl radical (•OH) scavenging assays. Chemical profiling of extracts was carried out using high-performance liquid chromatography coupled with a diode array detector (HPLC-DAD). Principal component analysis (PCA) was performed to differentiate the sample extracts based on their phytochemical profiles and amounts of target bioactive compounds. Chemical composition varied among the analyzed willow species and also among the plant organs of the same species. The major bioactive compounds of the extracts were salicin, chlorogenic acid, rutin and epicatechin. The extracts exhibited significant DPPH● and ●OH scavenging activities. Results of molecular docking revealed that chlorogenic acid had the highest binding affinity toward TNF-α and IL-6. UAE extracts represent valuable sources of antioxidant and anti-inflammatory compounds.
Collapse
Affiliation(s)
- Emilia Gligorić
- Department of Pharmacy, Faculty of Medicine, University of Novi Sad, Hajduk Veljkova 3, 21000 Novi Sad, Serbia
| | - Ružica Igić
- Department of Biology and Ecology, Faculty of Sciences, University of Novi Sad, Trg Dositeja Obradovića 3, 21000 Novi Sad, Serbia
| | - Branislava Teofilović
- Department of Pharmacy, Faculty of Medicine, University of Novi Sad, Hajduk Veljkova 3, 21000 Novi Sad, Serbia
| | - Nevena Grujić-Letić
- Department of Pharmacy, Faculty of Medicine, University of Novi Sad, Hajduk Veljkova 3, 21000 Novi Sad, Serbia
| |
Collapse
|
16
|
Mu L, Xu H, Hong Y, Zhou W, Wang L, Liu P, Chen M, Hu Y. Chemical compositions of Souliea vaginata (Maxim) Franch rhizome and their potential therapeutic effects on collagen-induced arthritis in rats. JOURNAL OF ETHNOPHARMACOLOGY 2023; 310:116416. [PMID: 36990303 DOI: 10.1016/j.jep.2023.116416] [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: 11/01/2022] [Revised: 03/15/2023] [Accepted: 03/20/2023] [Indexed: 06/19/2023]
Abstract
ETHNOPHARMACOLOGICAL REVEVANCE Rheumatoid arthritis (RA) is a global prevalent chronic autoimmune inflammatory disease and acceptable safety drugs are lack for its treatment. The rhizomes of Souliea vaginata (Maxim) Franch (SV) possess anti-inflammatory functions and are used as substitution of Coptis chinensis Franch. SV is also traditional Chinese medicine and Tibetan medicine for the treatment of conjunctivitis, enteritis and rheumatic. For searching complementary and alternative anti-RA drugs, it is necessary to characterize the potential anti-arthritic activity of SV and underlying mechanism involved. AIM OF THE STUDY The aim of the study was to test the chemical compositions, evaluate the anti-arthritic effects and underlying mechanisms of SV. MATERIALS AND METHODS The chemical compositions of SV were analyzed using liquid chromatography-ion trap-time of flight tandem mass spectrometry (LCMS-IT-TOF). From day 11 to day 31, SV (0.5, 1.0 and 1.5 g/kg body weight) and Tripterygium glycosidorum (TG, 10 mg/kg body weight) were administered orally to the CIA model rats once a day. Thickness of paw and body weights were measured once every two days from day 1 to day 31. Histopathological changes were measured using hematoxylin-eosin (HE) staining. Effects of SV on the levels of IL-2, TNF-α, IFN-γ, IL-4 and IL-10 in serum of CIA rats were measured by enzyme-linked immunosorbent assay (ELISA) kits. CD3+, CD4+, CD8+ and CD4+CD25+ T cells populations were measured using flow cytometric analysis. To evaluate the possible hepatotoxicity and nephrotoxicity, the serum levels of alanine aminotransferase (ALT), aspartate aminotransferase (AST), blood urea (UREA) and creatinine (CREA) in CIA rats were also tested using blood auto analyzer. RESULTS 34 compounds were identified from SV based on LCMS-IT-TOF, and triterpenoids are major anti-arthritic compositions. SV significantly relieved CIA rats' paw swelling without obvious influence on the body weight growth. SV decreased the serum levels of IL-2, TNF-α and IFN-γ in CIA rat, and increased the serum levels of IL-4 and IL-10. SV significantly increased and decreased the percentages of CD4+ and CD8+, with no significant effects on CD3+ in lymphocytes of CIA model rats. Moreover, SV simultaneously decreased thymus and spleen indexes and no hepatotoxicity and nephrotoxicity was observed after short-term treatment. CONCLUSION These findings suggest that SV possesses preventive and therapeutic effect on RA by modulating the inflammatory cytokines, T-lymphocyte, thymus and spleen indexes and shows no hepatotoxicity and nephrotoxicity.
Collapse
Affiliation(s)
- LiHua Mu
- Department of Pharmacy, Medical Supplies Center of Chinese PLA General Hospital, Beijing, 100853, China
| | - HaiYan Xu
- Department of Pharmacy, Medical Supplies Center of Chinese PLA General Hospital, Beijing, 100853, China
| | - Yan Hong
- Department of Obstetrics and Gynecology, The First Medical Centre of Chinese PLA General Hospital, Beijing, 100853, China
| | - WenBin Zhou
- Key Laboratory of Ethnomedicine of Ministry of Education, Minzu University of China, Beijing, 100081, China
| | - LiHua Wang
- Department of Pharmacy, Medical Supplies Center of Chinese PLA General Hospital, Beijing, 100853, China
| | - Ping Liu
- Department of Pharmacy, Medical Supplies Center of Chinese PLA General Hospital, Beijing, 100853, China
| | - MengLi Chen
- Department of Pharmacy, Medical Supplies Center of Chinese PLA General Hospital, Beijing, 100853, China.
| | - Yuan Hu
- Department of Pharmacy, Medical Supplies Center of Chinese PLA General Hospital, Beijing, 100853, China.
| |
Collapse
|
17
|
Ferrara V, Toti A, Lucarini E, Parisio C, Micheli L, Ciampi C, Margiotta F, Crocetti L, Vergelli C, Giovannoni MP, Di Cesare Mannelli L, Ghelardini C. Protective and Pain-Killer Effects of AMC3, a Novel N-Formyl Peptide Receptors (FPRs) Modulator, in Experimental Models of Rheumatoid Arthritis. Antioxidants (Basel) 2023; 12:1207. [PMID: 37371936 DOI: 10.3390/antiox12061207] [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: 05/03/2023] [Revised: 05/26/2023] [Accepted: 05/31/2023] [Indexed: 06/29/2023] Open
Abstract
Rheumatoid arthritis is an autoimmune disorder that causes chronic joint pain, swelling, and movement impairment, resulting from prolonged inflammation-induced cartilage and bone degradation. The pathogenesis of RA, which is still unclear, makes diagnosis and treatment difficult and calls for new therapeutic strategies to cure the disease. Recent research has identified FPRs as a promising druggable target, with AMC3, a novel agonist, showing preclinical efficacy in vitro and in vivo. In vitro, AMC3 (1-30 µM) exhibited significant antioxidant effects in IL-1β (10 ng/mL)-treated chondrocytes for 24 h. AMC3 displayed a protective effect by downregulating the mRNA expression of several pro-inflammatory and pro-algic genes (iNOS, COX-2, and VEGF-A), while upregulating genes essential for structural integrity (MMP-13, ADAMTS-4, and COLIAI). In vivo, AMC3 (10 mg kg-1) prevented hypersensitivity and restored postural balance in CFA-injected rats after 14 days. AMC3 attenuated joint alterations, reduced joint inflammatory infiltrate, pannus formation, and cartilage erosion. Chronic AMC3 administration reduced transcriptional changes of genes causing excitotoxicity and pain (EAATs and CCL2) and prevented morphological changes in astrocytes, including cell body hypertrophy, processes length, and thickness, caused by CFA in the spinal cord. This study demonstrates the usefulness of AMC3 and establishes the groundwork for further research.
Collapse
Affiliation(s)
- Valentina Ferrara
- Department of Neuroscience, Psychology, Drug Research and Child Health-NEUROFARBA-Pharmacology and Toxicology Section, University of Florence, 50139 Florence, Italy
| | - Alessandra Toti
- Department of Neuroscience, Psychology, Drug Research and Child Health-NEUROFARBA-Pharmacology and Toxicology Section, University of Florence, 50139 Florence, Italy
| | - Elena Lucarini
- Department of Neuroscience, Psychology, Drug Research and Child Health-NEUROFARBA-Pharmacology and Toxicology Section, University of Florence, 50139 Florence, Italy
| | - Carmen Parisio
- Department of Neuroscience, Psychology, Drug Research and Child Health-NEUROFARBA-Pharmacology and Toxicology Section, University of Florence, 50139 Florence, Italy
| | - Laura Micheli
- Department of Neuroscience, Psychology, Drug Research and Child Health-NEUROFARBA-Pharmacology and Toxicology Section, University of Florence, 50139 Florence, Italy
| | - Clara Ciampi
- Department of Neuroscience, Psychology, Drug Research and Child Health-NEUROFARBA-Pharmacology and Toxicology Section, University of Florence, 50139 Florence, Italy
| | - Francesco Margiotta
- Department of Neuroscience, Psychology, Drug Research and Child Health-NEUROFARBA-Pharmacology and Toxicology Section, University of Florence, 50139 Florence, Italy
| | - Letizia Crocetti
- Department of Neuroscience, Psychology, Drug Research and Child Health-NEUROFARBA-Pharmaceutical and Nutraceutical Section, University of Florence, 50139 Florence, Italy
| | - Claudia Vergelli
- Department of Neuroscience, Psychology, Drug Research and Child Health-NEUROFARBA-Pharmaceutical and Nutraceutical Section, University of Florence, 50139 Florence, Italy
| | - Maria Paola Giovannoni
- Department of Neuroscience, Psychology, Drug Research and Child Health-NEUROFARBA-Pharmaceutical and Nutraceutical Section, University of Florence, 50139 Florence, Italy
| | - Lorenzo Di Cesare Mannelli
- Department of Neuroscience, Psychology, Drug Research and Child Health-NEUROFARBA-Pharmacology and Toxicology Section, University of Florence, 50139 Florence, Italy
| | - Carla Ghelardini
- Department of Neuroscience, Psychology, Drug Research and Child Health-NEUROFARBA-Pharmacology and Toxicology Section, University of Florence, 50139 Florence, Italy
| |
Collapse
|
18
|
Luo Y, Shen Y, Zong L, Xie J, Dai L, Luo X. Anti-rheumatoid arthritis potential of Rhododendron molle G. Don leaf extract in adjuvant induced arthritis rats. JOURNAL OF ETHNOPHARMACOLOGY 2023; 307:116175. [PMID: 36702447 DOI: 10.1016/j.jep.2023.116175] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Revised: 01/12/2023] [Accepted: 01/15/2023] [Indexed: 06/18/2023]
Abstract
AIM OF THE STUDY The aim of this study was to test the anti-rheumatic arthritis effects of Rhododendron molle G. Don leaf extract in arthritis rats and inflammatory RAW 264.7 cells. Preliminary analysis and comparison of potential medicinal components of three polar extracts by HPLC and UHPLC-Q-TOF-MS. MATERIALS AND METHODS SD rats were subcutaneously injected with complete Freund's adjuvant (CFA) to induce inflammation on the right hind paw. RAW 264.7 cells were induced by lipopolysaccharide (LPS) to established cell inflammatory model. The volume of rat hind paw was measured with a volume meter to detect swelling, and the weight of rats was measured with an electronic balance. The severity of arthritis in rats was evaluated by arthritis score. The pathological sections of rat hind paw joints were observed by hematoxylin-eosin staining, and the contents of IL-6 and IL-1β in serum were detected. qRT-PCR was used to detect the expression of IL-1β, IL-6, TNF-α and COX-2 genes in RAW 264.7 cells. The release of nitric oxide was measured by Griess reaction. The expression levels of IL-6 and IL-1β were detected by Western-Blot. RESULTS and discussion: The chloroform extract from R. molle leaves (CERL), Ethyl acetate extract from R. molle leaves (EERL), n-butanol extract from R. molle leaves (BERL) could significantly inhibit hind paws swelling and reduce arthritis index in arthritis rats. And it showed dose dependence. Compared with tripterygium glycosides (TG) tablets, an effective drug of RA treatment, CERL have better anti-RA effect after administration. In addition, the three kinds of the polar extracts of Rhododendron molle leaves (PERL) had lower toxicity, with the LD50 279.87, 239.65, 500.08 (mg/kg) respectively, while TG group's LD50 was 96.00 (mg/kg). In vitro experiments showed that the three PERLs can significantly inhibit the level of pro-inflammatory factors and inflammatory mediator, such as TNF-α, IL-1β, IL-6, COX-2 and NO, which were consistent with their anti-RA ability. Among the three kinds of PERLs, CERL showed the best inhibitory activity. CONCLUSION The R. molle leaf is a potential medicinal part for the treatment of RA. This study explored the anti-RA and anti-inflammatory activities of CERL, EERL, BERL, which laid a foundation for further promoting the clinical application of R. molle.
Collapse
Affiliation(s)
- Yuqiang Luo
- College of Life Science, Jiangxi Normal University, Nanchang, 330022, China.
| | - Yu Shen
- College of Life Science, Jiangxi Normal University, Nanchang, 330022, China.
| | - Luye Zong
- College of Life Science, Jiangxi Normal University, Nanchang, 330022, China.
| | - Jiankun Xie
- College of Life Science, Jiangxi Normal University, Nanchang, 330022, China.
| | - Liangfang Dai
- College of Life Science, Jiangxi Normal University, Nanchang, 330022, China.
| | - Xiangdong Luo
- College of Life Science, Jiangxi Normal University, Nanchang, 330022, China.
| |
Collapse
|
19
|
Yu Z, Tsapis N, Fay F, Chen L, Karpus A, Shi X, Cailleau C, García Pérez S, Huang N, Vergnaud J, Mignani S, Majoral JP, Fattal E. Amphiphilic Phosphorus Dendrons Associated with Anti-inflammatory siRNA Reduce Symptoms in Murine Collagen-Induced Arthritis. Biomacromolecules 2023; 24:667-677. [PMID: 36599673 DOI: 10.1021/acs.biomac.2c01117] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Small interfering RNA (siRNA) holds promise for treating rheumatoid arthritis by inhibiting major cytokines such as tumor necrosis factor-α (TNF-α). We developed original cationic amphiphilic phosphorus dendrons to produce dendriplexes associated with TNF-α siRNA. The dendrons were made of 10 pyrrolidinium end groups and a C17 aliphatic chain. The dendriplexes demonstrated the ability to protect siRNA from nuclease degradation and to promote macrophage uptake. Moreover, they led to potent inhibition of TNF-α expression in the lipopolysaccharide-activated mouse macrophage cell line RAW264.7 in vitro model. A significant anti-inflammatory effect in the murine collagen-induced arthritis model was observed through arthritis scoring and histological observations. These results open up essential perspectives in using this original amphiphilic dendron to reduce the disease burden and improve outcomes in chronic inflammatory diseases.
Collapse
Affiliation(s)
- Zhibo Yu
- Université Paris-Saclay, CNRS, Institut Galien Paris-Saclay, Orsay91400, France
| | - Nicolas Tsapis
- Université Paris-Saclay, CNRS, Institut Galien Paris-Saclay, Orsay91400, France
| | - François Fay
- Université Paris-Saclay, CNRS, Institut Galien Paris-Saclay, Orsay91400, France
| | - Liang Chen
- Laboratoire de Chimie de Coordination, CNRS, Université de Toulouse, Toulouse Cedex 431077, France.,Université Toulouse, 118 Route de Narbonne, Toulouse Cedex431077, France
| | - Andrii Karpus
- Laboratoire de Chimie de Coordination, CNRS, Université de Toulouse, Toulouse Cedex 431077, France.,Université Toulouse, 118 Route de Narbonne, Toulouse Cedex431077, France
| | - Xiangyang Shi
- College of Chemistry, Chemical Engineering and Biotechnology, Donghua University, Shanghai201620, PR China
| | - Catherine Cailleau
- Université Paris-Saclay, CNRS, Institut Galien Paris-Saclay, Orsay91400, France
| | - Samuel García Pérez
- Rheumatology & Immuno-mediated Diseases Research Group (IRIDIS), Galicia Sur Health Research Institute (IIS Galicia Sur), Sergas-Uvigo36213, Spain
| | - Nicolas Huang
- Université Paris-Saclay, CNRS, Institut Galien Paris-Saclay, Orsay91400, France
| | - Juliette Vergnaud
- Université Paris-Saclay, CNRS, Institut Galien Paris-Saclay, Orsay91400, France
| | - Serge Mignani
- Laboratoire de Chimie et Biochimie Pharmacologiques et Toxicologique, University Paris Descartes, Pres Sorbonne Paris Cité, CNRS UMR 860, 45 Rue des Saints Peres, Paris75006, France.,CQM─Centro de Química da Madeira, Universidade da Madeira, Funchal9020-105, Portugal
| | - Jean-Pierre Majoral
- Laboratoire de Chimie de Coordination, CNRS, Université de Toulouse, Toulouse Cedex 431077, France
| | - Elias Fattal
- Université Paris-Saclay, CNRS, Institut Galien Paris-Saclay, Orsay91400, France
| |
Collapse
|
20
|
Zeng W, Fang Y, Mo S, Shen C, Yang H, Luo G, Xiao L, Zhan R, Yan P. The Underling Mechanisms Exploration of Rubia cordifolia L. Extract Against Rheumatoid Arthritis by Integrating Network Pharmacology and Metabolomics. Drug Des Devel Ther 2023; 17:439-457. [PMID: 36818604 PMCID: PMC9930591 DOI: 10.2147/dddt.s388932] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Accepted: 02/02/2023] [Indexed: 02/13/2023] Open
Abstract
Purpose Rubia cordifolia L. (RC) is a classic herbal medicine for the treatment of rheumatoid arthritis (RA) and has been used since ancient times. The ethanol extract of Rubia cordifolia L. (RCE) showed obvious anti-RA effects in our previous study. However, further potential mechanisms require more exploration. We aimed to investigate the mechanism of RCE for the treatment of RA by integrating metabolomics and network pharmacology in this study. Methods An adjuvant-induced arthritis (AIA) rat model was established, and we evaluated the therapeutic effects of RCE. Metabolomics of serum and urine was used to identify the differential metabolites. Network pharmacology was applied to determine the key metabolites and potential targets. Finally, the potential targets and compounds of RCE were verified by molecular docking. Results The results indicated that RCE suppressed foot swelling and alleviated joint damage and also had anti-inflammatory properties by inhibiting the expressions of tumor necrosis factor (TNF)-α, Interleukin (IL)-1β, prostaglandin E2 (PGE2), and P65. Ten and seven differential metabolites were found in the serum and urine, respectively, of rats. Six key targets, ie, phospholipase A2 group IIA (PLA2G2A), phospholipase A2 group X (PLA2G10), cytidine deaminase (CDA), uridine-cytidine kinase 2 (UCK2), charcot-leyden crystal galectin (CLC), and 5',3'-nucleotidase, mitochondrial (NT5M), were discovered by network pharmacology and metabolite analysis and were found to be related to glycerophospholipid metabolism and pyrimidine metabolism. Molecular docking confirmed that the favorable compounds showed affinities with the key targets, including alizarin, 6-hydroxyrubiadin, ruberythric acid, and munjistin. Conclusion This study revealed the underlying mechanisms of RCE and provided evidence that will allow researchers to further investigate the functions and components of RCE against RA.
Collapse
Affiliation(s)
- Weiya Zeng
- College of Traditional Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, People’s Republic of China,Key Laboratory of Chinese Medicinal Resources from Lingnan (Guangzhou University of Chinese Medicine), Ministry of Education, Guangzhou, People’s Republic of China,Joint Laboratory of Nation Engineering Research Center for the Pharmaceutics of Traditional Chinese Medicines, Guangzhou, People’s Republic of China
| | - Yuan Fang
- College of Traditional Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, People’s Republic of China,Key Laboratory of Chinese Medicinal Resources from Lingnan (Guangzhou University of Chinese Medicine), Ministry of Education, Guangzhou, People’s Republic of China,Joint Laboratory of Nation Engineering Research Center for the Pharmaceutics of Traditional Chinese Medicines, Guangzhou, People’s Republic of China
| | - Suifen Mo
- College of Traditional Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, People’s Republic of China,Key Laboratory of Chinese Medicinal Resources from Lingnan (Guangzhou University of Chinese Medicine), Ministry of Education, Guangzhou, People’s Republic of China,Joint Laboratory of Nation Engineering Research Center for the Pharmaceutics of Traditional Chinese Medicines, Guangzhou, People’s Republic of China
| | - Caihong Shen
- College of Traditional Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, People’s Republic of China,Key Laboratory of Chinese Medicinal Resources from Lingnan (Guangzhou University of Chinese Medicine), Ministry of Education, Guangzhou, People’s Republic of China,Joint Laboratory of Nation Engineering Research Center for the Pharmaceutics of Traditional Chinese Medicines, Guangzhou, People’s Republic of China
| | - Huiling Yang
- College of Traditional Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, People’s Republic of China,Key Laboratory of Chinese Medicinal Resources from Lingnan (Guangzhou University of Chinese Medicine), Ministry of Education, Guangzhou, People’s Republic of China,Joint Laboratory of Nation Engineering Research Center for the Pharmaceutics of Traditional Chinese Medicines, Guangzhou, People’s Republic of China
| | - Guihua Luo
- College of Traditional Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, People’s Republic of China,Key Laboratory of Chinese Medicinal Resources from Lingnan (Guangzhou University of Chinese Medicine), Ministry of Education, Guangzhou, People’s Republic of China,Joint Laboratory of Nation Engineering Research Center for the Pharmaceutics of Traditional Chinese Medicines, Guangzhou, People’s Republic of China
| | - Luhua Xiao
- College of Traditional Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, People’s Republic of China,Key Laboratory of Chinese Medicinal Resources from Lingnan (Guangzhou University of Chinese Medicine), Ministry of Education, Guangzhou, People’s Republic of China,Joint Laboratory of Nation Engineering Research Center for the Pharmaceutics of Traditional Chinese Medicines, Guangzhou, People’s Republic of China
| | - Ruoting Zhan
- College of Traditional Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, People’s Republic of China,Key Laboratory of Chinese Medicinal Resources from Lingnan (Guangzhou University of Chinese Medicine), Ministry of Education, Guangzhou, People’s Republic of China,Joint Laboratory of Nation Engineering Research Center for the Pharmaceutics of Traditional Chinese Medicines, Guangzhou, People’s Republic of China,Correspondence: Ruoting Zhan; Ping Yan, Guangzhou University of Chinese Medicine, No. 232, Outer Ring East Road, Guangzhou, Guangdong, People’s Republic of China, Tel/Fax +86 20-39358045, Email ;
| | - Ping Yan
- College of Traditional Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, People’s Republic of China,Key Laboratory of Chinese Medicinal Resources from Lingnan (Guangzhou University of Chinese Medicine), Ministry of Education, Guangzhou, People’s Republic of China,Joint Laboratory of Nation Engineering Research Center for the Pharmaceutics of Traditional Chinese Medicines, Guangzhou, People’s Republic of China
| |
Collapse
|
21
|
Hannan A, Akhtar B, Sharif A, Anjum F, Pasha I, Khan A, Akhtar MF, Saleem A. Quercetin-loaded chitosan nanoparticles ameliorate adjuvant-induced arthritis in rats by regulating anti-oxidant enzymes and downregulating pro- and inflammatory cytokines. Inflammopharmacology 2023; 31:287-300. [PMID: 36542211 DOI: 10.1007/s10787-022-01118-4] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2022] [Accepted: 12/10/2022] [Indexed: 12/24/2022]
Abstract
Rheumatoid arthritis (RA) is an inflammatory condition and associated with the symmetrical synovitis of the joints and cause joint pain. The use of anti-rheumatic drugs is associated with many adverse effects. Quercetin, an important polyphenolic flavonoid, possess anti-inflammatory and anti-rheumatic effects. Quercetin use is limited due to poor absorption and bioavailability. Nanomedicines are used for the targeted drug delivery, hence it reduces the adverse effects of the drug. Based upon these factors, quercetin-loaded chitosan nanoparticles (Q-NPs) were prepared by solvent evaporation method, characterized and their better anti-rheumatic effect with mechanistic insights was validated in Freund's complete adjuvant (FCA)-induced arthritic rats along with safety studies. The animals were divided into five groups, each containing 5 animals. Group I was normal control, group II was arthritic control, while groups III, IV and V were administered with quercetin (15 mg/Kg) and Q-NPs (10 and 20 mg/Kg), respectively. The reduction in ankle diameter, serum oxidative stress markers as well as pro- and inflammatory cytokines, e.g., tumor necrosis factor (TNFα), interleukin (IL-6) were determined. The prepared Q-NPs showed hydrodynamic size of 83.9 nm, polydispersity index of 0.687, entrapment efficiency 90.5% as well as no interaction between quercetin and chitosan in Fourier transform infrared spectroscopy (FTIR). A significant reduction (p < 0.001) in ankle diameter was observed after administration of high-dose Q-NPs (4.32 ± 0.14 cm to 5.13 ± 0.62 cm). There was also reduction (p < 0.001) in levels of TNFα and IL-6 following high-dose Q-NPs (72.56 ± 2.30 and 308.19 ± 11.5 pg). The effect on biochemical tests, hematological parameters and oxidative stress parameters was also found to be significant. Histopathological changes of kidney, liver and ankle also confirmed the anti-rheumatic effect of high-dose Q-NPs. The study concludes that administration of Q-NPs (20 mg/Kg) may be used for the treatment of FCA-induced RA in rats.
Collapse
Affiliation(s)
- Abdul Hannan
- Institute of Physiology and Pharmacology, University of Agriculture, Faisalabad, Pakistan
| | - Bushra Akhtar
- Department of Pharmacy, University of Agriculture, Faisalabad, Pakistan.
| | - Ali Sharif
- Institute of Pharmacy, Faculty of Pharmaceutical and Allied Health Sciences, Lahore College for Women University, Lahore, Pakistan
| | - Fozia Anjum
- Department of Chemistry, Government College University, Faisalabad, Pakistan
| | - Imran Pasha
- National Institute of Food Science and Technology, University of Agriculture, Faisalabad, Pakistan
| | - Ahrar Khan
- Shandong Vocational Animal Science and Veterinary College, Weifang, China
| | - Muhammad Furqan Akhtar
- Riphah Institute of Pharmaceutical Sciences, Riphah International University Lahore Campus, Lahore, Pakistan
| | - Ammara Saleem
- Department of Pharmacology, Faculty of Pharmaceutical Sciences, Government College University, Faisalabad, Pakistan
| |
Collapse
|
22
|
Abstract
INTRODUCTION Rheumatoid Arthritis (RA) remains a challenge for rheumatologists and patients despite implementation of intensive treat-to-target strategies in shared decision with patients and an increasing availability of drugs. Janus kinase inhibitors (JAKi) are a new generation of oral targeted drugs. Filgotinib preferentially inhibits JAK1 and is the latest JAKi to be approved for use in RA. AREAS COVERED This narrative review focuses on drug characteristics, efficacy, and safety of filgotinib in patients with RA, summarizing available literature. Trial data are detailed, put into perspective for practice and discussed in regulatory perspective. EXPERT OPINION Preclinical studies demonstrate preferential inhibition of JAK1 and a promising pharmacokinetic profile with few drug-drug interactions. Increase in hemoglobin in line with preferential inhibition of JAK1 over JAK2 is seen in early-phase clinical trials. A phase III program demonstrates efficacy in several disease stages, numerically higher with 200 mg versus 100 mg daily. In the overall RA population such dose-related effect is not observed for safety except for herpes zoster and increases in lipids and creatine phosphokinase. This reassuring safety profile is to be confirmed in future practice. It also needs to be unraveled if JAK1 preferential inhibition plays a key role in this safety profile.
Collapse
Affiliation(s)
- Rene Westhovens
- Emeritus Professor KU Leuven, Skeletal Biology and Engineering Research Center Department of Development and Regeneration, Leuven, Belgium
| |
Collapse
|
23
|
Phytochemical Evaluation and Anti-Inflammatory Potential of Miconia albicans (Sw.) Triana Extracts. Molecules 2022; 27:molecules27185954. [PMID: 36144693 PMCID: PMC9500825 DOI: 10.3390/molecules27185954] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Revised: 09/02/2022] [Accepted: 09/07/2022] [Indexed: 11/17/2022] Open
Abstract
The plant Miconia albicans (Sw.) Triana has been popularly used in Brazil to treat chronic inflammatory disturbances, such as osteoarthritis. This disease affects 250 million people worldwide, and is associated with intense pain and loss of articular function. There is a lack of information about the phytochemistry and bioactivity of M. albicans. Therefore, this study determined the chemical composition of some extracts and evaluated their cytotoxicity, along with their antioxidant and anti-inflammatory, activities using in vitro models. Aqueous and ethanolic extracts were prepared. Afterwards, a liquid–liquid partition was developed using chloroform, ethyl acetate, and n-butanol. The extracts were characterized by LC–MS, and their biological activities were evaluated on epithelial cells (Vero), tumoral hepatic cells (Hep-G2), and THP-1 macrophages. LC–MS analyses identified several flavonoids in all fractions, such as quercetin, myricetin, and their glycosides. The crude extracts and n-butanol fractions did not present cytotoxicity to the cells. The non-toxic fractions presented significant antioxidant activity when evaluated in terms of DPPH scavenging activity, lipid peroxidation, and ROS inhibition. THP-1 macrophages treated with the n-butanol fraction (250 µg/mL) released fewer pro-inflammatory cytokines, even in the presence of LPS. In the future, it will be necessary to identify the phytochemicals that are responsible for anti-inflammatory effects for the discovery of new drugs. In vivo studies on M. albicans extracts are still required to confirm their possible mechanisms of action.
Collapse
|
24
|
Gu X, Zhao L, Tan J, Zhang Q, Fu L, Li J. Characterization of a novel β-agarase from Antarctic macroalgae-associated bacteria metagenomic library and anti-inflammatory activity of the enzymatic hydrolysates. Front Microbiol 2022; 13:972272. [PMID: 36118221 PMCID: PMC9478344 DOI: 10.3389/fmicb.2022.972272] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2022] [Accepted: 08/01/2022] [Indexed: 11/13/2022] Open
Abstract
An agarase gene (aga1904) that codes a protein with 640 amino acids was obtained from the metagenomic library of macroalgae-associated bacteria collected from King George Island, Antarctica. Gene aga1904 was expressed in Escherichia coli BL21 (DE3) and recombinant Aga1904 was purified by His Bind Purification kit. The optimal temperature and pH for the activity of Aga1904 were 50°C and 6.0, respectively. Fe3+ and Cu2+ significantly inhibited the activity of Aga1904. The Vmax and Km values of recombinant Aga1904 were 108.70 mg/ml min and 6.51 mg/ml, respectively. The degradation products of Aga1904 against agarose substrate were mainly neoagarobiose, neoagarotetraose, and neoagarohexaose analyzed by thin layer chromatography. The cellular immunoassay of enzymatic hydrolysates was subsequently carried out, and the results showed that agaro-oligosaccharides dominated by neoagarobiose significantly inhibited key pro-inflammatory markers including, nitric oxide (NO), interleukins 6 (IL-6), and tumor necrosis factor α (TNF-α). This work provides a promising candidate for development recombinant industrial enzyme to prepare agaro-oligosaccharides, and paved up a new path for the exploitation of natural anti-inflammatory agent in the future.
Collapse
Affiliation(s)
- Xiaoqian Gu
- Key Laboratory of Ecological Environment Science and Technology, First Institute of Oceanography, Ministry of Natural Resources, Qingdao, China
- CAS and Shandong Province Key Laboratory of Experimental Marine Biology, Center for Ocean Mega-Science, Institute of Oceanology, Chinese Academy of Sciences, Qingdao, China
| | - Luying Zhao
- Key Laboratory of Ecological Environment Science and Technology, First Institute of Oceanography, Ministry of Natural Resources, Qingdao, China
| | - Jiaojiao Tan
- CAS and Shandong Province Key Laboratory of Experimental Marine Biology, Center for Ocean Mega-Science, Institute of Oceanology, Chinese Academy of Sciences, Qingdao, China
| | - Qian Zhang
- Key Laboratory of Ecological Environment Science and Technology, First Institute of Oceanography, Ministry of Natural Resources, Qingdao, China
| | - Liping Fu
- Key Laboratory of Ecological Environment Science and Technology, First Institute of Oceanography, Ministry of Natural Resources, Qingdao, China
| | - Jiang Li
- Key Laboratory of Ecological Environment Science and Technology, First Institute of Oceanography, Ministry of Natural Resources, Qingdao, China
- *Correspondence: Jiang Li,
| |
Collapse
|
25
|
Chen C, Yin Y, Shi G, Zhou Y, Shao S, Wei Y, Wu L, Zhang D, Sun L, Zhang T. A highly selective JAK3 inhibitor is developed for treating rheumatoid arthritis by suppressing γc cytokine-related JAK-STAT signal. SCIENCE ADVANCES 2022; 8:eabo4363. [PMID: 35984890 PMCID: PMC9390995 DOI: 10.1126/sciadv.abo4363] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Accepted: 07/07/2022] [Indexed: 05/28/2023]
Abstract
Janus kinases (JAKs) play a critical role in immune responses by relaying signals from more than 50 cytokines, making them attractive therapeutic targets for autoimmune diseases. Although approved JAK inhibitors have demonstrated clinical efficacy, they target a broad spectrum of cytokines, which results in side effects. Therefore, next-generation inhibitors maintain efficacy, while sparing adverse events need to be developed. Among members of the JAK family, JAK3 only regulates a narrow spectrum of γc cytokines and becomes a potentially ideal target. Here, a highly JAK3-selective inhibitor Z583 is developed, which showed a potent inhibition of JAK3 with an IC50 of 0.1 nM and exhibited a 4500-fold selectivity for JAK3 than other JAK subtypes. Furthermore, Z583 completely inhibited the γc cytokine signaling and sufficiently blocked the development of inflammatory response in RA model, while sparing hematopoiesis. Collectively, the highly selective JAK3 inhibitor Z583 is a promising candidate with significant therapeutic potential for autoimmune diseases.
Collapse
Affiliation(s)
- Chengjuan Chen
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Yuan Yin
- School of Science, China Pharmaceutical University, Nanjing 210009, China
| | - Gaona Shi
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Yu Zhou
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Shuai Shao
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Yazi Wei
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Lei Wu
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Dayong Zhang
- School of Science, China Pharmaceutical University, Nanjing 210009, China
| | - Lan Sun
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Tiantai Zhang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| |
Collapse
|
26
|
Concerted regulation of OPG/RANKL/ NF‑κB/MMP-13 trajectories contribute to ameliorative capability of prodigiosin and/or low dose γ-radiation against adjuvant- induced arthritis in rats. Int Immunopharmacol 2022; 111:109068. [PMID: 35944459 DOI: 10.1016/j.intimp.2022.109068] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Revised: 07/08/2022] [Accepted: 07/14/2022] [Indexed: 11/22/2022]
Abstract
BACKGROUND Prodigiosin (PDG) is a microbial red dye with antioxidant and anti-inflammatory properties, although its effect on rheumatoid arthritis (RA) remains uncertain. Also, multiple doses of low dose γ- radiation (LDR) have been observed to be as a successful intervention for RA. Thus, the purpose of this study was to investigate the ameliorative potential of PDG and/or LDR on adjuvant-induced arthritis (AIA) in rats. METHODS The anti-inflammatory and anti-arthritic effects of PDG and/or LDR were examined in vitro and in vivo, respectively. In the AIA model, the arthritic indexes, paw swelling degrees, body weight gain, and histopathological assessment in AIA rats were assayed. The impact of PDG (200 µg/kg; p.o) and/or LDR (0.5 Gy) on the levels of pro- and anti-inflammatory cytokines (IL-1β, TNF-α, IL-6, IL-18, IL-17A, and IL-10) as well as the regulation of osteoprotegrin (OPG)/ receptor activator of nuclear factor κB ligand (RANKL)/ nuclear factor-κB (NF-κB)/MMP-13 pathways was determined. Methotrexate (MTX; 0.05 mg/kg; twice/week, i.p) was administered concurrently as a standard anti-arthritic drug. RESULTS PDG and/or LDR markedly diminished the arthritic indexes, paw edema, weigh loss in AIA rats, alleviated the pathological alterations in joints, reduced the levels of pro-inflammatory cytokines IL-1β, TNF-α, IL-6, IL-18, IL-17A, and RANKL in serum and synovial tissues, while increasing anti-inflammatory cytokines IL-10 and OPG levels. Moreover, PDG and/or LDR down-regulated the expression of RANKL, NF-κBp65, MMP13, caspase-3, and decreased the RANKL/OPG ratio, whereas OPG and collagen II were enhanced in synovial tissues. CONCLUSION PDG and/or LDR exhibited obvious anti-RA activity on AIA.
Collapse
|
27
|
Sakyi SA, Owusu‐Yeboah M, Obirikorang C, Dadzie Ephraim RK, Kwarteng A, Opoku S, Afranie BO, Senu E, Boateng AO, Boakye DK, Buckman TA, Amoani B. Profiling vitamin D, its mediators and proinflammatory cytokines in rheumatoid arthritis: A case-control study. Immun Inflamm Dis 2022; 10:e676. [PMID: 35894711 PMCID: PMC9274797 DOI: 10.1002/iid3.676] [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: 04/25/2022] [Revised: 06/25/2022] [Accepted: 06/27/2022] [Indexed: 11/24/2022] Open
Abstract
INTRODUCTION The active form of vitamin D has immunomodulatory and anti-inflammatory effect. Vitamin D is implicated in pathogenesis of rheumatoid arthritis (RA) and its deficiency leads to increased inflammation. Moreover, its production is dependent on concentration of calcium, phosphorus, and parathyroid hormone (PTH). Cytokines mediates inflammation in RA synovium. This study evaluated vitamin D, its mediators and proinflammatory cytokines among RA patients. METHODS In a case-control study, 78 RA patients from Komfo Anokye Teaching Hospital rheumatology clinic and 60 healthy blood donors were recruited. Chemistry analyzer and enzyme-linked immunosorbent assay kits were used to measure biochemical parameters and cytokines. RESULTS We found significantly higher levels of interleukin (IL)-1β, interferon gamma (IFN-γ), and tumor necrosis factor-α (TNF-α) in RA patients compared with controls (p < .05). There was a significant positive correlation between intact parathyroid hormone (iPTH) and IL-10 (r = .30, p < .05) and a negative correlation between IL-6 (r = -0.28, p > .05), IL-1β (r = -0.25, p > .05), TNF-α (r = -0.26, p > .05), IFN-γ (r = -0.24, p > .05), and iPTH. There was a significant negative correlation between IL-1β (r = -0.33, p < .05), IFN- γ (r = -0.29, p < .05), and calcium. CONCLUSION Reduced PTH, calcium, and phosphorus is associated with higher levels of proinflammatory cytokines which may worsen RA disease condition. Vitamin D is therefore not an independent regulator of proinflammatory cytokines in RA.
Collapse
Affiliation(s)
- Samuel A. Sakyi
- Department of Molecular Medicine, School of Medicine and DentistryKwame Nkrumah University of Science and TechnologyKumasiGhana
| | - Mavis Owusu‐Yeboah
- Department of Molecular Medicine, School of Medicine and DentistryKwame Nkrumah University of Science and TechnologyKumasiGhana
| | - Christian Obirikorang
- Department of Molecular Medicine, School of Medicine and DentistryKwame Nkrumah University of Science and TechnologyKumasiGhana
| | - Richard K. Dadzie Ephraim
- Department of Medical Laboratory Sciences, Faculty of Allied HealthUniversity of Cape CoastCape CoastGhana
| | - Alexander Kwarteng
- Department of Biochemistry and BiotechnologyKwame Nkrumah University of Science and TechnologyKumasiGhana
| | - Stephen Opoku
- Department of Molecular Medicine, School of Medicine and DentistryKwame Nkrumah University of Science and TechnologyKumasiGhana
- Department of Medical Diagnostics, Faculty of Allied Health Sciences, College of Health SciencesKwame Nkrumah University of Science and TechnologyKumasiGhana
| | - Bright O. Afranie
- Department of Molecular Medicine, School of Medicine and DentistryKwame Nkrumah University of Science and TechnologyKumasiGhana
| | - Ebenezer Senu
- Department of Molecular Medicine, School of Medicine and DentistryKwame Nkrumah University of Science and TechnologyKumasiGhana
| | - Andy O. Boateng
- Department of Molecular Medicine, School of Medicine and DentistryKwame Nkrumah University of Science and TechnologyKumasiGhana
| | - Derrick K. Boakye
- Department of Medical Diagnostics, Faculty of Allied Health Sciences, College of Health SciencesKwame Nkrumah University of Science and TechnologyKumasiGhana
| | - Tonnies A. Buckman
- Department of Molecular Medicine, School of Medicine and DentistryKwame Nkrumah University of Science and TechnologyKumasiGhana
| | - Benjamin Amoani
- Department of Biomedical Science, School of Allied Health SciencesUniversity of Cape CoastCape CoastGhana
| |
Collapse
|
28
|
Zahedipour F, Hosseini SA, Henney NC, Barreto GE, Sahebkar A. Phytochemicals as inhibitors of tumor necrosis factor alpha and neuroinflammatory responses in neurodegenerative diseases. Neural Regen Res 2022; 17:1675-1684. [PMID: 35017414 PMCID: PMC8820712 DOI: 10.4103/1673-5374.332128] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Revised: 01/15/2021] [Accepted: 06/20/2021] [Indexed: 12/06/2022] Open
Abstract
Inflammatory processes and proinflammatory cytokines have a key role in the cellular processes of neurodegenerative diseases and are linked to the pathogenesis of functional and mental health disorders. Tumor necrosis factor alpha has been reported to play a major role in the central nervous system in Alzheimer's disease, Parkinson's disease and amyotrophic lateral sclerosis and many other neurodegenerative diseases. Therefore, a potent proinflammatory/proapoptotic tumor necrosis factor alpha could be a strong candidate for targeted therapy. Plant derivatives have now become promising candidates as therapeutic agents because of their antioxidant and chemical characteristics, and anti-inflammatory features. Recently, phytochemicals including flavonoids, terpenoids, alkaloids, and lignans have generated interest as tumor necrosis factor alpha inhibitor candidates for a number of diseases involving inflammation within the nervous system. In this review, we discuss how phytochemicals as tumor necrosis factor alpha inhibitors are a therapeutic strategy targeting neurodegeneration.
Collapse
Affiliation(s)
- Fatemeh Zahedipour
- Department of Medical Biotechnology, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
- Student Research Committee, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Seyede Atefe Hosseini
- Department of Medical Biotechnology, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
- Student Research Committee, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Neil C. Henney
- Pharmacy & Biomolecular Sciences, Liverpool John Moores University, Liverpool, UK
| | - George E. Barreto
- Department of Biological Sciences, University of Limerick, Limerick, Ireland
- Health Research Institute, University of Limerick, Limerick, Ireland
| | - Amirhossein Sahebkar
- Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
- Polish Mother's Memorial Hospital Research Institute (PMMHRI), Lodz, Poland
- School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
29
|
Heimfarth L, Rezende MM, Pereira EWM, Passos FRS, Monteiro BS, Santos TKB, Lima NT, Souza ICL, de Albuquerque Junior RLC, de Souza Siqueira Lima P, de Souza Araújo AA, Quintans Júnior LJ, Kim B, Coutinho HDM, de Souza Siqueira Quintans J. Pharmacological effects of a complex α-bisabolol/β-cyclodextrin in a mice arthritis model with involvement of IL-1β, IL-6 and MAPK. Biomed Pharmacother 2022; 151:113142. [PMID: 35623175 DOI: 10.1016/j.biopha.2022.113142] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Revised: 05/03/2022] [Accepted: 05/15/2022] [Indexed: 11/15/2022] Open
Abstract
Inflammatory arthritis is the most prevalent chronic inflammatory disease worldwide. The pathology of the disease is characterized by increased inflammation and oxidative stress, which leads to chronic pain and functional loss in the joints. Conventional anti-arthritic drugs used to relieve pain and other arthritic symptoms often cause severe side effects. α-bisabolol (BIS) is a sesquiterpene that exhibits high anti-inflammatory potential and a significant antinociceptive effect. This study evaluates the anti-arthritic, anti-inflammatory and antihyperalgesic effects of BIS alone and in a β-cyclodextrin (βCD/BIS) inclusion complex in a CFA-induced arthritis model. Following the intra-articular administration of CFA, male mice were treated with vehicle, BIS and βCD/BIS (50 mg/kg, p.o.) or a positive control and pain-related behaviors, knee edema and inflammatory and oxidative parameters were evaluated on days 4, 11, 18 and/or 25. Ours findings shows that the oral administration of BIS and βCD/BIS significantly attenuated spontaneous pain-like behaviors, mechanical hyperalgesia, grip strength deficit and knee edema induced by repeated injections of CFA, reducing the joint pain and functional disability associated with arthritis. BIS and βCD/BIS also inhibited the generation of inflammatory and oxidative markers in the knee and blocked MAPK in the spinal cord. In addition, ours results also showed that the incorporation of BIS in cyclodextrin as a drug delivery system improved the pharmacological profile of this substance. Therefore, these results contribute to the pharmacological knowledge of BIS and demonstrated that this terpene appears to be able to mitigate deleterious symptoms of arthritis.
Collapse
Affiliation(s)
- Luana Heimfarth
- Multiuser Health Center Facility (CMulti-Saúde), Federal University of Sergipe, São Cristóvão, SE, Brazil; Health Sciences Graduate Program (PPGCS), Federal University of Sergipe, São Cristóvão, SE, Brazil; Laboratory of Neurosciences and Pharmacological Assays (LANEF), Department of Physiology, Federal University of Sergipe, São Cristóvão, Brazil
| | - Marília Matos Rezende
- Multiuser Health Center Facility (CMulti-Saúde), Federal University of Sergipe, São Cristóvão, SE, Brazil; Health Sciences Graduate Program (PPGCS), Federal University of Sergipe, São Cristóvão, SE, Brazil; Laboratory of Neurosciences and Pharmacological Assays (LANEF), Department of Physiology, Federal University of Sergipe, São Cristóvão, Brazil
| | - Erik Willyame Menezes Pereira
- Multiuser Health Center Facility (CMulti-Saúde), Federal University of Sergipe, São Cristóvão, SE, Brazil; Health Sciences Graduate Program (PPGCS), Federal University of Sergipe, São Cristóvão, SE, Brazil; Laboratory of Neurosciences and Pharmacological Assays (LANEF), Department of Physiology, Federal University of Sergipe, São Cristóvão, Brazil
| | - Fabiolla Rocha Santos Passos
- Multiuser Health Center Facility (CMulti-Saúde), Federal University of Sergipe, São Cristóvão, SE, Brazil; Health Sciences Graduate Program (PPGCS), Federal University of Sergipe, São Cristóvão, SE, Brazil; Laboratory of Neurosciences and Pharmacological Assays (LANEF), Department of Physiology, Federal University of Sergipe, São Cristóvão, Brazil
| | - Brenda Souza Monteiro
- Laboratory of Neurosciences and Pharmacological Assays (LANEF), Department of Physiology, Federal University of Sergipe, São Cristóvão, Brazil
| | - Tiffany Karoline Barroso Santos
- Laboratory of Neurosciences and Pharmacological Assays (LANEF), Department of Physiology, Federal University of Sergipe, São Cristóvão, Brazil
| | - Natália Teles Lima
- Laboratory of Neurosciences and Pharmacological Assays (LANEF), Department of Physiology, Federal University of Sergipe, São Cristóvão, Brazil
| | - Isana Carla Leal Souza
- Laboratory of Morphology and Experimental Pathology, Research and Technology Institute, Tiradentes University (UNIT), Aracaju, SE, Brazil
| | | | - Pollyana de Souza Siqueira Lima
- Laboratory of Neurosciences and Pharmacological Assays (LANEF), Department of Physiology, Federal University of Sergipe, São Cristóvão, Brazil
| | | | - Lucindo José Quintans Júnior
- Multiuser Health Center Facility (CMulti-Saúde), Federal University of Sergipe, São Cristóvão, SE, Brazil; Health Sciences Graduate Program (PPGCS), Federal University of Sergipe, São Cristóvão, SE, Brazil; Laboratory of Neurosciences and Pharmacological Assays (LANEF), Department of Physiology, Federal University of Sergipe, São Cristóvão, Brazil
| | - Bonglee Kim
- Department of Pathology, College of Korean Medicine, Kyung Hee University, Seoul 02447, Republic of Korea.
| | - Henrique D M Coutinho
- Department of Biological Chemistry, Regional University of Cariri - URCA, Crato, Brazil.
| | - Jullyana de Souza Siqueira Quintans
- Multiuser Health Center Facility (CMulti-Saúde), Federal University of Sergipe, São Cristóvão, SE, Brazil; Health Sciences Graduate Program (PPGCS), Federal University of Sergipe, São Cristóvão, SE, Brazil; Laboratory of Neurosciences and Pharmacological Assays (LANEF), Department of Physiology, Federal University of Sergipe, São Cristóvão, Brazil.
| |
Collapse
|
30
|
Yin H, Liu N, Sigdel KR, Duan L. Role of NLRP3 Inflammasome in Rheumatoid Arthritis. Front Immunol 2022; 13:931690. [PMID: 35833125 PMCID: PMC9271572 DOI: 10.3389/fimmu.2022.931690] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Accepted: 05/30/2022] [Indexed: 11/13/2022] Open
Abstract
Rheumatoid arthritis (RA) is a chronic inflammatory disease characterized by multi-articular, symmetrical and invasive arthritis resulting from immune system abnormalities involving T and B lymphocytes. Although significant progress has been made in the understanding of RA pathogenesis, the underlying mechanisms are not fully understood. Recent studies suggest that NLRP3 inflammasome, a regulator of inflammation, might play an important role in the development of RA. There have been increasing clinical and pre-clinical evidence showing the treatment of NLRP3/IL-1β in inflammatory diseases. To provide a foundation for the development of therapeutic strategies, we will briefly summarize the roles of NLRP3 inflammasome in RA and explore its potential clinical treatment.
Collapse
Affiliation(s)
- Hui Yin
- Department of Rheumatology and Clinical Immunology, Jiangxi Provincial People’s Hospital, Medical College of Nanchang University, Nanchang, China
- Department of Rheumatology and Clinical Immunology, Jiangxi Provincial People’s Hospital, The First Affiliated Hospital of Nanchang Medical College, Nanchang, China
| | - Na Liu
- Department of Rheumatology and Clinical Immunology, Jiangxi Provincial People’s Hospital, Medical College of Nanchang University, Nanchang, China
- Department of Rheumatology and Clinical Immunology, Jiangxi Provincial People’s Hospital, The First Affiliated Hospital of Nanchang Medical College, Nanchang, China
| | - Keshav Raj Sigdel
- Department of Internal Medicine, Patan Academy of Health Sciences, Kathmandu, Nepal
| | - Lihua Duan
- Department of Rheumatology and Clinical Immunology, Jiangxi Provincial People’s Hospital, Medical College of Nanchang University, Nanchang, China
- Department of Rheumatology and Clinical Immunology, Jiangxi Provincial People’s Hospital, The First Affiliated Hospital of Nanchang Medical College, Nanchang, China
- *Correspondence: Lihua Duan,
| |
Collapse
|
31
|
Tekale S, Gore V, Kendrekar P, Thore S, Kótai L, Pawar R. COVID-19 Global Pandemic Fight by Drugs: A Mini-Review on Hope and Hype. MINI-REV ORG CHEM 2022. [DOI: 10.2174/1570193x18666210629103117] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
:
Coronavirus disease 2019 (Covid-19), a serious disease caused by the Severe Acute Respiratory
Syndrome-Corona Virus-2 (SARS-CoV-2), was firstly identified in the city of Wuhan of
China in December 2019, which then spread and became a global issue due to its high transmission
rate. To date, the outbreak of COVID-19 has resulted in infection to 230,868,745 people and the death
of 4,732,669 patients. It has paralyzed the economy of all the countries worldwide. Considering the
possible mutations of SARS-CoV-2, the current medical emergency requires a longer time for drug
design and vaccine development. Drug repurposing is a promising option for potent therapeutics
against the pandemic. The present review encompasses various drugs or appropriate combinations of
already FDA-approved antimalarial, antiviral, anticancer, anti-inflammatory, and antibiotic therapeutic
candidates for use in the clinical trials as a ray of hope against COVID-19. It is expected to deliver
better clinical and laboratory outcomes of drugs as a prevention strategy for the eradication of the disease.
Collapse
Affiliation(s)
- Sunil Tekale
- Department of Chemistry, Deogiri College, Aurangabad-431005, Maharashtra, India
| | - Vishnu Gore
- Department of Chemistry, Deogiri College, Aurangabad-431005, Maharashtra, India
| | - Pravin Kendrekar
- Unit for Drug Discovery Research (UDDR), Department of Health and Environmental Sciences, Central University of Technology, Free State (CUT) Private Bag X20539, Bloemfontein, 9300, South Africa
| | - Shivaji Thore
- Department of Chemistry, Deogiri College, Aurangabad-431005, Maharashtra, India
| | - László Kótai
- Research Centre for Natural Sciences, ELKH, H-1117, Budapest, Hungary
| | - Rajendra Pawar
- Department of Chemistry, Shiv Chhatrapati College, Cidco, Aurangabad-431005, Maharashtra, India
| |
Collapse
|
32
|
Smigoc Schweiger D. Recent Advances in Immune-Based Therapies for Type 1 Diabetes. Horm Res Paediatr 2022; 96:631-645. [PMID: 35533645 DOI: 10.1159/000524866] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Accepted: 04/18/2022] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND Type 1 diabetes (T1D) is a chronic autoimmune disease characterized by progressive destruction of the pancreatic beta cells, leading to a lifelong dependence on insulin. It is associated with an increased morbidity and mortality from diabetes-related complications and a significant treatment burden. However, there has been substantial progress in therapeutic strategies that can affect the course of the disease. SUMMARY This review addresses advances in immunotherapy aimed at preserving residual beta-cell function in individuals with a recent onset of T1D and arresting the disease in pre-symptomatic stages. Recent and ongoing clinical trials have investigated the efficacy and safety of various immunotherapeutic strategies aimed at targeting several mechanisms of autoimmunity, which are thought to be important in disease pathogenesis, and therapies that also address beta-cell health. So far, T-cell-directed therapies that led to a favourable balance between T-effector cell depletion or modulation and preservation or expansion of regulatory T cells have shown the most success. Furthermore, regarding the timing of intervention, teplizumab was the first immunomodulatory agent to demonstrate a significant delay in disease progression in high-risk individuals before clinical onset. KEY MESSAGES As more targeted immune interventions with potentially fewer side effects are closer to the translation into clinical practice, some new challenges may need to be addressed. The use of combination approaches that include immunotherapeutic strategies targeting different aspects of the immune system and interventions that improve beta-cell health may be required, along with the use of individualized patient-tailored approaches, a move towards early intervention, and a focus on patient-reported outcome measures.
Collapse
Affiliation(s)
- Darja Smigoc Schweiger
- Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
- Department of Endocrinology, Diabetes and Metabolic Diseases, University Children's Hospital, University Medical Centre Ljubljana, Ljubljana, Slovenia
| |
Collapse
|
33
|
Pistorius K, Ly L, Souza PR, Gomez EA, Koenis DS, Rodriguez AR, Foster J, Sosabowski J, Hopkinson M, Rajeeve V, Spur BW, Pitsillides A, Pitzalis C, Dalli J. MCTR3 reprograms arthritic monocytes to upregulate Arginase-1 and exert pro-resolving and tissue-protective functions in experimental arthritis. EBioMedicine 2022; 79:103974. [PMID: 35430453 PMCID: PMC9038546 DOI: 10.1016/j.ebiom.2022.103974] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Revised: 03/13/2022] [Accepted: 03/15/2022] [Indexed: 11/03/2022] Open
Abstract
BACKGROUND Rheumatoid arthritis (RA) is a progressive degenerative disorder that leads to joint destruction. Available treatments only target the inflammatory component with minimal impact on joint repair. We recently uncovered a previously unappreciated family of pro-resolving mediators, the maresin conjugate in tissue regeneration (MCTR), that display both immunoregulatory and tissue-protective activities. Thus, we queried whether the production of these autacoids is disrupted in RA patients and whether they can be useful in treating joint inflammation and promoting joint repair. METHODS Using a highly phenotyped RA cohort we evaluated plasma MCTR concentrations and correlated these to clinical markers of disease activity. To evaluate the immunoregulatory and tissue reparative activities we employed both in vivo models of arthritis and organ culture models. FINDINGS Herein, we observed that plasma MCTR3 concentrations were negatively correlated with joint disease activity and severity in RA patients. Evaluation of the mechanisms engaged by this mediator in arthritic mice demonstrated that MCTR3 reprograms monocytes to confer enduring joint protective properties. Single cell transcriptomic profiling and flow cytometric evaluation of macrophages from mice treated with MCTR3-reprogrammed monocytes revealed a role for Arginase-1 (Arg-1) in mediating their joint reparative and pro-resolving activities. Arg-1 inhibition reversed both the anti-arthritic and tissue reparative actions of MCTR3-reprogrammed monocytes. INTERPRETATION Our findings demonstrate that circulating MCTR3 levels are negatively correlated with disease in RA. When administered to mice in vivo, MCTR3 displayed both anti-inflammatory and joint reparative activities, protecting both cartilage and bone in murine arthritis. These activities were, at least in part, mediated via the reprogramming of mononuclear phagocyte responses. FUNDING This work was supported by funding from the European Research Council (ERC) under the European Union's Horizon 2020 research and innovation programme (grant no: 677542) and the Barts Charity (grant no: MGU0343) to J.D. J.D. is also supported by a Sir Henry Dale Fellowship jointly funded by the Wellcome Trust and the Royal Society (grant 107613/Z/15/Z).
Collapse
Affiliation(s)
- Kimberly Pistorius
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, Charterhouse Square, London, EC1M 6BQ UK
| | - Lucy Ly
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, Charterhouse Square, London, EC1M 6BQ UK
| | - Patricia R Souza
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, Charterhouse Square, London, EC1M 6BQ UK
| | - Esteban A Gomez
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, Charterhouse Square, London, EC1M 6BQ UK
| | - Duco S Koenis
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, Charterhouse Square, London, EC1M 6BQ UK
| | - Ana R Rodriguez
- Rowan University School of Osteopathic Medicine, Department of Cell Biology & Neuroscience, 2 Medical Centre Drive, Stratford NJ 08084, USA
| | - Julie Foster
- Centre for Cancer Biomarkers and Biotherapeutics, Barts Cancer Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, Charterhouse Square, London, EC1M 6BQ UK
| | - Jane Sosabowski
- Centre for Cancer Biomarkers and Biotherapeutics, Barts Cancer Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, Charterhouse Square, London, EC1M 6BQ UK
| | - Mark Hopkinson
- Department of Comparative Biomedical Sciences, Royal Veterinary College, London, UK
| | - Vinothini Rajeeve
- Mass spectrometry Laboratory, Barts Cancer Institute, Queen Mary University of London, Charterhouse Square, London, EC1M 6BQ, United Kingdom
| | - Bernd W Spur
- Rowan University School of Osteopathic Medicine, Department of Cell Biology & Neuroscience, 2 Medical Centre Drive, Stratford NJ 08084, USA
| | - Andrew Pitsillides
- Department of Comparative Biomedical Sciences, Royal Veterinary College, London, UK
| | - Costantino Pitzalis
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, Charterhouse Square, London, EC1M 6BQ UK
| | - Jesmond Dalli
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, Charterhouse Square, London, EC1M 6BQ UK; Centre for Inflammation and Therapeutic Innovation, Queen Mary University of London, London, UK.
| |
Collapse
|
34
|
Zhang W, Chen Y, Liu Q, Zhou M, Wang K, Wang Y, Nie J, Gui S, Peng D, He Z, Li Z. Emerging nanotherapeutics alleviating rheumatoid arthritis by readjusting the seeds and soils. J Control Release 2022; 345:851-879. [DOI: 10.1016/j.jconrel.2022.04.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Revised: 03/31/2022] [Accepted: 04/02/2022] [Indexed: 12/12/2022]
|
35
|
Abstract
Rheumatoid arthritis is a common chronic inflammatory disease with substantial economic, social, and personal costs. Its pathogenesis is multifactorial and complex. The ultimate goal of rheumatoid arthritis treatment is stopping or slowing down the disease progression. In the past two decades, invention of new medicines, especially biologic agents, revolutionized the management of this disease. These agents have been associated with an improved prognosis and clinical remission, especially in patients who did not respond to traditional disease-modifying anti-rheumatic drugs (DMARDs). Improvement in the understanding of the rheumatoid arthritis pathogenesis leads to the development of novel biologic therapeutic approaches. In the present paper, we summarized the current therapeutics, especially biologic agents, available for the treatment of rheumatoid arthritis.
Collapse
|
36
|
The multi-omics and analysis to reveal thermal processing enhanced anti-rheumatoid arthritis efficacy of Radix Clematidis in rats. J Pharm Biomed Anal 2022; 215:114760. [DOI: 10.1016/j.jpba.2022.114760] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Revised: 04/01/2022] [Accepted: 04/05/2022] [Indexed: 11/21/2022]
|
37
|
Yuan S, Li X, Lin A, Larsson SC. Interleukins and rheumatoid arthritis: bi-directional Mendelian randomization investigation. Semin Arthritis Rheum 2022; 53:151958. [DOI: 10.1016/j.semarthrit.2022.151958] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Revised: 01/05/2022] [Accepted: 01/12/2022] [Indexed: 10/19/2022]
|
38
|
Leng T, Wang Y, Cheng W, Wang W, Qu X, Lei B. Bioactive anti-inflammatory antibacterial metformin-contained hydrogel dressing accelerating wound healing. BIOMATERIALS ADVANCES 2022; 135:212737. [PMID: 35929210 DOI: 10.1016/j.bioadv.2022.212737] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Revised: 01/19/2022] [Accepted: 02/21/2022] [Indexed: 12/12/2022]
Abstract
Highly efficient wound healing and skin regeneration remain a challenge. Long-term inflammation and bacterial infection can inhibit the healing process and lead to the scar formation. Here, we report a hydrogel (FEM) formed by self-assembly of ε-poly-l-lysine-F127-ε-poly-l-lysine (EPL-F127-EPL) and metformin for wound repair. Especially, the role of metformin-based antibacterial hydrogel in wound healing and repair was investigated for the first time. FEM has inherent multifunctional properties, including controlled metformin release, anti-inflammatory and antibacterial activity, temperature responsiveness, injectable and self-healing capabilities. The in vivo results showed that FEM dressings accelerated the wound healing by stimulating the angiogenesis process of the wound tissue and anti-inflammation. This study shows that the multifunctional metformin-contained hydrogel scaffolds could enhance the wound repair through the anti-inflammation and accelerated angiogenesis, which could also expand the biomedical applications of metformin-based biomaterials.
Collapse
Affiliation(s)
- Tongtong Leng
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Frontier Institute of Science and Technology, Xi'an Jiaotong University, Xi'an 710054, China
| | - Yidan Wang
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Frontier Institute of Science and Technology, Xi'an Jiaotong University, Xi'an 710054, China
| | - Wei Cheng
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Frontier Institute of Science and Technology, Xi'an Jiaotong University, Xi'an 710054, China
| | - Wensi Wang
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Frontier Institute of Science and Technology, Xi'an Jiaotong University, Xi'an 710054, China.
| | - Xiaoyan Qu
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Frontier Institute of Science and Technology, Xi'an Jiaotong University, Xi'an 710054, China
| | - Bo Lei
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Frontier Institute of Science and Technology, Xi'an Jiaotong University, Xi'an 710054, China; Department of Orthopedics, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, China; State Key Laboratory for Mechanical Behavior of Materials, Instrument Analysis Center, Xi'an Jiaotong University, Xi'an 710054, China; State Key Laboratory for Manufacturing Systems Engineering, Xi'an Jiaotong University, Xi'an 710054, China.
| |
Collapse
|
39
|
Jiang H, Xu F, Zeng L, Li C, Chen Y, Wang L, Li Z, Liu R. Saponins from Nigella glandulifera seeds attenuate collagen-induced rheumatoid arthritis in rats via the OPG/RANKL/NF-κB and Ang/Tie-2 pathways. JOURNAL OF ETHNOPHARMACOLOGY 2022; 283:114714. [PMID: 34624452 DOI: 10.1016/j.jep.2021.114714] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Revised: 09/25/2021] [Accepted: 10/04/2021] [Indexed: 06/13/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Nigella glandulifera Freyn et Sint. (N. glandulifera) seeds are widely used in traditional Uyghur medicine for a variety of immuno-inflammatory diseases. The total saponins from N. glandulifera seeds (TSNGS) have been shown to have analgesic, antioxidant, and anti-inflammatory effects that can alleviate joint pain and swelling. AIM OF THE STUDY Rheumatoid arthritis (RA) is a chronic and progressive, debilitating autoimmune disease for which current treatments are not sufficiently effective and result in unsatisfactory side effects. This study aimed to mechanistically investigate the therapeutic effects of TSNGS on RA. MATERIALS AND METHODS Qualitative analysis of TSNGS was performed using ultra-high-performance liquid chromatography-Q-Orbitrap-high-resolution mass spectrometry. Rats with collagen-induced arthritis (CIA), IL-1β-induced HFLS-RAs, and VEGF-induced HUVECs were analyzed to determine the efficacy and mechanism of TSNGS on RA. RESULTS Twenty-one compounds were identified in TSNGS. TSNGS (10, 50, or 250 mg/kg) reduced the severity of arthritis, indicated by a lower arthritis score, reduced paw swelling, and body weight in rats with CIA. TSNGS ameliorated histopathological changes involving inflammatory infiltration, bone degeneration, and angiogenesis in knee and ankle joints. TSNGS improved the immuno-inflammatory response by restoring the levels of the cytokines IFN-γ, TNF-α, IL-1β, IL-6, IL-17A, IL-4, and IL-10, and increasing the number of CD4+CD25+ Tregs in the peripheral circulation and Foxp3 levels in knee joints in rats with CIA. Furthermore, TSNGS increased the OPG/RANKL ratio and downregulated p-p65 in serum and joint synovia. Inhibition of angiogenesis by TSNGS was associated with recovery of the angiogenesis-related Ang/Tie-2 signaling pathway. CONCLUSIONS It was established that TSNGS provides a therapeutic effect on RA by alleviating synovitis, bone degeneration, and angiogenesis via the OPG/RANKL/NF-κB and Ang/Tie-2 pathways and may be used for the treatment of RA.
Collapse
Affiliation(s)
- Hailun Jiang
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, PR China
| | - Fang Xu
- Key Laboratory of Uighur Medicine of Xinjiang Uygur Autonomous Region, Xinjiang Institute of Materia Medica, Urumqi, 830004, PR China
| | - Li Zeng
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, PR China
| | - Chenyang Li
- Key Laboratory of Uighur Medicine of Xinjiang Uygur Autonomous Region, Xinjiang Institute of Materia Medica, Urumqi, 830004, PR China
| | - Yan Chen
- Key Laboratory of Uighur Medicine of Xinjiang Uygur Autonomous Region, Xinjiang Institute of Materia Medica, Urumqi, 830004, PR China
| | - Linlin Wang
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, PR China
| | - Zhuorong Li
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, PR China.
| | - Rui Liu
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, PR China.
| |
Collapse
|
40
|
Gao T, Liu T, Ko CJ, Zhang L, Joo D, Xie X, Zhu L, Li Y, Cheng X, Sun SC. Myeloid cell TBK1 restricts inflammatory responses. Proc Natl Acad Sci U S A 2022; 119:e2107742119. [PMID: 35074921 PMCID: PMC8794809 DOI: 10.1073/pnas.2107742119] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Accepted: 11/23/2021] [Indexed: 12/21/2022] Open
Abstract
Proinflammatory cytokine production by innate immune cells plays a crucial role in inflammatory diseases, but the molecular mechanisms controlling the inflammatory responses are poorly understood. Here, we show that TANK-binding kinase 1 (TBK1) serves as a vital regulator of proinflammatory macrophage function and protects against tissue inflammation. Myeloid cell-conditional Tbk1 knockout (MKO) mice spontaneously developed adipose hypertrophy and metabolic disorders at old ages, associated with increased adipose tissue M1 macrophage infiltration and proinflammatory cytokine expression. When fed with a high-fat diet, the Tbk1-MKO mice also displayed exacerbated hepatic inflammation and insulin resistance, developing symptoms of nonalcoholic steatohepatitis. Furthermore, myeloid cell-specific TBK1 ablation exacerbates inflammation in experimental colitis. Mechanistically, TBK1 functions in macrophages to suppress the NF-κB and MAP kinase signaling pathways and thus attenuate induction of proinflammatory cytokines, particularly IL-1β. Ablation of IL-1 receptor 1 (IL-1R1) eliminates the inflammatory symptoms of Tbk1-MKO mice. These results establish TBK1 as a pivotal anti-inflammatory mediator that restricts inflammation in different disease models.
Collapse
Affiliation(s)
- Tianxiao Gao
- Department of Immunology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030
| | - Ting Liu
- Department of Immunology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030
| | - Chun-Jung Ko
- Department of Immunology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030;
- Graduate Institute of Immunology, College of Medicine, National Taiwan University, Taipei, Taiwan 100233
| | - Lingyun Zhang
- Department of Immunology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030
| | - Donghyun Joo
- Department of Immunology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030
| | - Xiaoping Xie
- Department of Immunology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030
| | - Lele Zhu
- Department of Immunology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030
| | - Yanchuan Li
- Department of Immunology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030
| | - Xuhong Cheng
- Department of Immunology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030
| | - Shao-Cong Sun
- Department of Immunology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030;
- MD Anderson Cancer Center UT Health Graduate School of Biomedical Sciences, Houston, TX 77030
| |
Collapse
|
41
|
Docherty S, Harley R, McAuley JJ, Crowe LAN, Pedret C, Kirwan PD, Siebert S, Millar NL. The effect of exercise on cytokines: implications for musculoskeletal health: a narrative review. BMC Sports Sci Med Rehabil 2022; 14:5. [PMID: 34991697 PMCID: PMC8740100 DOI: 10.1186/s13102-022-00397-2] [Citation(s) in RCA: 110] [Impact Index Per Article: 36.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Accepted: 12/31/2021] [Indexed: 02/07/2023]
Abstract
The physiological effects of physical exercise are ubiquitously reported as beneficial to the cardiovascular and musculoskeletal systems. Exercise is widely promoted by medical professionals to aid both physical and emotional wellbeing; however, mechanisms through which this is achieved are less well understood. Despite numerous beneficial attributes, certain types of exercise can inflict significant significant physiological stress. Several studies document a key relationship between exercise and immune activation. Activation of the innate immune system occurs in response to exercise and it is proposed this is largely mediated by cytokine signalling. Cytokines are typically classified according to their inflammatory properties and evidence has shown that cytokines expressed in response to exercise are diverse and may act to propagate, modulate or mitigate inflammation in musculoskeletal health. The review summarizes the existing literature on the relationship between exercise and the immune system with emphasis on how exercise-induced cytokine expression modulates inflammation and the immune response.
Collapse
Affiliation(s)
- Sophie Docherty
- Institute of Infection, Immunity and Inflammation, College of Medicine, Veterinary and Life Sciences, University of Glasgow, 120 University Avenue, Glasgow, G12 8TA, Scotland, UK
| | - Rachael Harley
- Institute of Infection, Immunity and Inflammation, College of Medicine, Veterinary and Life Sciences, University of Glasgow, 120 University Avenue, Glasgow, G12 8TA, Scotland, UK
| | - Joseph J McAuley
- Institute of Infection, Immunity and Inflammation, College of Medicine, Veterinary and Life Sciences, University of Glasgow, 120 University Avenue, Glasgow, G12 8TA, Scotland, UK
| | - Lindsay A N Crowe
- Institute of Infection, Immunity and Inflammation, College of Medicine, Veterinary and Life Sciences, University of Glasgow, 120 University Avenue, Glasgow, G12 8TA, Scotland, UK
| | - Carles Pedret
- Sports Medicine and Imaging Department, Clinica Diagonal, C/Sant Mateu 24-26, 08950, Esplugues de Llobregat, Spain
| | - Paul D Kirwan
- School of Physiotherapy, Royal College of Surgeons in Ireland, Dublin, Ireland
- Physiotherapy Department, Connolly Hospital, Dublin, Ireland
| | - Stefan Siebert
- Institute of Infection, Immunity and Inflammation, College of Medicine, Veterinary and Life Sciences, University of Glasgow, 120 University Avenue, Glasgow, G12 8TA, Scotland, UK
| | - Neal L Millar
- Institute of Infection, Immunity and Inflammation, College of Medicine, Veterinary and Life Sciences, University of Glasgow, 120 University Avenue, Glasgow, G12 8TA, Scotland, UK.
| |
Collapse
|
42
|
Bhattacharya HS, Srivastava R, Gummaluri SS, Agarwal MC, Bhattacharya P, Astekar MS. Comparison of blood parameters between periodontitis patients and healthy participants: A cross-sectional hematological study. J Oral Maxillofac Pathol 2022; 26:77-81. [PMID: 35571313 PMCID: PMC9106243 DOI: 10.4103/jomfp.jomfp_349_21] [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: 09/24/2021] [Revised: 01/06/2022] [Accepted: 01/21/2022] [Indexed: 11/04/2022] Open
Abstract
Background Bacteria and their products involved in periodontitis evoke an immunoinflammatory response in the host tissue. Inflammatory diseases, such as periodontitis, are often not just a local event, but may have systemic ramifications, including elevations in the numbers of circulating leukocytes, acute-phase proteins and oxidative stress markers. It is now emerging that also erythrocytes are affected by chronic inflammatory diseases. This phenomenon, named "anemia of inflammation," is not caused by marrow deficiencies or other diseases. The present study aimed to assess whether there was any relation between chronic periodontitis and hematological parameters. Materials and Methods A total of 80 patients were included in the study and were divided into the healthy and periodontitis groups. Blood sample was obtained from each participant for hematological analysis of leukocytes, erythrocytes, platelets, red blood cell (RBC) distribution width (RDW), mean corpuscular volume (MCV), platelet count and neutrophil-leukocyte ratio (NLR). Further, the values were gathered and subjected to statistical analysis. Unpaired t-test was performed to assess the statistical significance between the groups and P < 0.05 and < 0.001 were considered to be statistically significant. Results Results show statistically significant difference seen in leukocytes, lymphocytes, RDW, MCV, platelet count and NLR which was higher in patients with periodontitis, all other parameters are nonsignificant. Conclusion Thus, within limitations, it can be concluded that increased levels of leukocytes, lymphocytes, RDW, MCV, platelet count and NLR depict the inflammatory state and destructive nature of periodontitis.
Collapse
Affiliation(s)
- Hirak S Bhattacharya
- Department of Periodontology and Implantology, Institute of Dental Sciences, Bareilly, Uttar Pradesh, India
| | - Rishabh Srivastava
- Department of Periodontology and Implantology, Kothiwal Dental College and Research Centre, Moradabad, Uttar Pradesh, India
| | - Shiva Shankar Gummaluri
- Department of Periodontology and Implantology, Sree Sai Dental College and Research Institute, Srikakulam, Andhra Pradesh, India
| | - Manvi Chandra Agarwal
- Department of Periodontology and Implantology, Institute of Dental Sciences, Bareilly, Uttar Pradesh, India
| | - Preeti Bhattacharya
- Department of Orthodontics and Dentofacial Orthopaedics, Institute of Dental Sciences, Bareilly, Uttar Pradesh, India
| | - Madhusudan S Astekar
- Department of Oral and Maxillofacial Pathology and Microbiology, Institute of Dental Sciences, Bareilly, Uttar Pradesh, India
| |
Collapse
|
43
|
van Beers JJBC, Damoiseaux JGMC. Treatment of Autoimmune Diseases with Therapeutic Antibodies: Lessons Learned from PID Patients Allow for Stratification of the Infection Risk. Methods Mol Biol 2022; 2313:27-44. [PMID: 34478130 DOI: 10.1007/978-1-0716-1450-1_2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Over the years, a wide variety of therapeutic antibodies has been successfully introduced in the autoimmunology clinic and many more are on the edge to follow. Many of these treatments address either a pathogenic circulating molecule or a cell-bound molecule. Whereas the former target results in neutralization of the soluble factor, the latter target either inhibits cellular function or induces selective cell death. If this targeted molecule or cell is part of the immune system, this therapy evokes a state of immunodeficiency. Knowing the exact function of the respective components enables the risk stratification for possible infectious complications in patients treated with biologics. Much of the understanding of the function of immune cells and their associated molecules, in relation to redundancy in the immune system, is derived from studies in knockout mice. However, as mice are not men in terms of their life-expectancy, their infection exposure, or the composition of their immune system, the most useful knowledge for estimating the consequence of therapeutic intervention on immune competence comes from monitoring patients. In the current chapter, we focus on patients with a primary immunodeficiency (PID) because they provide us with a unique perspective to estimate the redundancy of a certain genetic defect for overall immune competence. These patients have inborn errors of the immune system that, in general, are due to single gene defects. Depending on the immunological pathway that is defective, patients can present with different types of (opportunistic) infectious diseases, as well as other clinical manifestations. Based on selected examples, we focus in this chapter on finding parallels in the infectious risk of autoimmune patients treated with biologics and PID patients with a defect in the immunological pathway that is affected by the respective biologic. The goal is to learn from the (dis)similarities between both patient populations in terms of safety profiles of biologic treatments.
Collapse
Affiliation(s)
- Joyce J B C van Beers
- Central Diagnostic Laboratory, Maastricht University Medical Center, Maastricht, The Netherlands
| | - Jan G M C Damoiseaux
- Central Diagnostic Laboratory, Maastricht University Medical Center, Maastricht, The Netherlands.
| |
Collapse
|
44
|
Zhang H, Liu J, Zhang P, Li D, Feng G, Huandike M, Sun S, Chai L, Zhou J. Herbal Formula Longteng Decoction Promotes the Regression of Synovial Inflammation in Collagen-Induced Arthritis Mice by Regulating Type 2 Innate Lymphocytes. Front Pharmacol 2021; 12:778845. [PMID: 35002715 PMCID: PMC8735860 DOI: 10.3389/fphar.2021.778845] [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: 09/17/2021] [Accepted: 11/29/2021] [Indexed: 11/13/2022] Open
Abstract
The etiology and pathogenesis of rheumatoid arthritis (RA) have not yet been fully elucidated, with greater adverse drug effects in traditional treatment of RA. It is particularly necessary to develop and study Chinese herbal formula as a supplement and alternative drug for the treatment of RA. The traditional Chinese medicine compound Longteng Decoction (LTD), as an empirical prescription in the treatment of RA in Dongzhimen Hospital of Beijing University of Chinese Medicine, has been widely used in clinic. Type 2 innate lymphocytes (ILC2s) have specific transcription factors and signature cytokines that are very similar to Th cells, which have been proved to be necessary in addressing RA inflammation, and are potential targets for RA prevention and treatment. Our previous studies have confirmed that LTD can intervene in the differentiation of peripheral blood Th17 and Treg cells, reduce joint pain index and swelling degree, shorten the time of morning stiffness, reduce ESR, and inhibit joint inflammation. However, it is unclear whether LTD can promote the regression of RA synovial inflammation by regulating the immune response mechanism of ILC2s.Therefore, our team established a collagen-induced arthritis mouse model and conducted an experimental study with LTD as the intervention object. The results showed that joint swelling, synovial inflammatory infiltration, and articular cartilage destruction were alleviated in CIA mice after intervention with LTD. The proliferation and differentiation of Th17 inflammatory cells and the secretion of proinflammatory cytokines (IL-17 and IFN-γ) were inhibited. In addition, LTD can also activate ILC2s to secrete the anti-inflammatory cytokine IL-4, activate the STAT6 signaling pathway, and act synergistic with Treg cells to inhibit the infiltration of type M1 macrophages in synovial tissue and promote its transformation to M2 phenotype. Taken together, these results confirm that LTD can be used as an adjunct or alternative to RA therapy by modulating the ILC2s immune response network and slowing down the inflammatory process of synovial tissue.
Collapse
Affiliation(s)
- Huijie Zhang
- Key Laboratory of Chinese Internal Medicine of Ministry of Education and Beijing, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Juan Liu
- Key Laboratory of Chinese Internal Medicine of Ministry of Education and Beijing, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Pingxin Zhang
- Key Laboratory of Chinese Internal Medicine of Ministry of Education and Beijing, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Dongyang Li
- Key Laboratory of Chinese Internal Medicine of Ministry of Education and Beijing, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Guiyu Feng
- Key Laboratory of Chinese Internal Medicine of Ministry of Education and Beijing, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Meiyier Huandike
- Key Laboratory of Chinese Internal Medicine of Ministry of Education and Beijing, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Song Sun
- Key Laboratory of Chinese Internal Medicine of Ministry of Education and Beijing, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Limin Chai
- Key Laboratory of Chinese Internal Medicine of Ministry of Education and Beijing, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Jingwei Zhou
- Key Laboratory of Chinese Internal Medicine of Ministry of Education and Beijing, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
- Department of Rheumatology, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| |
Collapse
|
45
|
Wang L, Liu L, Hong X, Liu D, Cheng Z. Delanzomib, a Novel Proteasome Inhibitor, Combined With Adalimumab Drastically Ameliorates Collagen-Induced Arthritis in Rats by Improving and Prolonging the Anti-TNF-α Effect of Adalimumab. Front Pharmacol 2021; 12:782385. [PMID: 34880764 PMCID: PMC8645831 DOI: 10.3389/fphar.2021.782385] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Accepted: 11/08/2021] [Indexed: 12/16/2022] Open
Abstract
Delanzomib is a novel proteasome inhibitor initially developed for treating multiple myeloma. It was found to inhibit the expression of tumor necrosis factor alpha (TNF-α). This study aimed to investigate the ameliorating effect of delanzomib on collagen-induced arthritis (CIA) and to explore the pharmacodynamics and pharmacokinetics (PK) interactions between delanzomib and adalimumab. Rats with CIA were randomly assigned to receive the treatment with delanzomib, adalimumab, delanzomib combined with adalimumab, or placebo. Visual inspection and biochemical examinations including TNF-α, interleukin 6, and C-reactive protein were performed to assess arthritis severity during the treatment. The adalimumab concentration in rats was determined to evaluate the PK interaction between delanzomib and adalimumab. Also, the levels of neonatal Fc receptor (FcRn) and FcRn mRNA were measured to explore the role of FcRn in the PK interaction between delanzomib and adalimumab. As a result, delanzomib combined with adalimumab exhibited stronger anti-arthritis activity than a single drug because both drugs synergistically reduced TNF-α level in vivo. Delanzomib also decreased adalimumab elimination in rats by increasing the level of FcRn. The slower elimination of adalimumab in rats further prolonged the anti-TNF-α effect of adalimumab. Moreover, FcRn level was increased by delanzomib via suppressing FcRn degradation rather than promoting FcRn production. In conclusion, delanzomib combined with adalimumab may be a potential therapeutic approach for treating rheumatoid arthritis. The initial finding that the PK interaction occurred between delanzomib and adalimumab may have clinical relevance for patients who simultaneously take proteasome inhibitors and anti-TNF-α therapeutic proteins.
Collapse
Affiliation(s)
- Lei Wang
- Department of Rheumatology and Immunology, The Second Clinical Medical College, Jinan University (Shenzhen People’s Hospital), Shenzhen, China
- Integrated Chinese and Western Medicine Postdoctoral Research Station, Jinan University, Guangzhou, China
| | - Lixiong Liu
- Department of Rheumatology and Immunology, The Second Clinical Medical College, Jinan University (Shenzhen People’s Hospital), Shenzhen, China
| | - Xiaoping Hong
- Department of Rheumatology and Immunology, The Second Clinical Medical College, Jinan University (Shenzhen People’s Hospital), Shenzhen, China
| | - Dongzhou Liu
- Department of Rheumatology and Immunology, The Second Clinical Medical College, Jinan University (Shenzhen People’s Hospital), Shenzhen, China
| | - Zeneng Cheng
- Research Institute of Drug Metabolism and Pharmacokinetics, School of Xiangya Pharmaceutical Sciences, Central South University, Changsha, China
| |
Collapse
|
46
|
Yang T, Sun K, Wang C, Swarnkar G, Quan S, Kress D, Xiao J, Alippe Y, Zheng H, Brophy RH, Hao D, McAlinden A, Abu-Amer Y, Shen J, Mbalaviele G. Gasdermin D deficiency attenuates arthritis induced by traumatic injury but not autoantibody-assembled immune complexes. Arthritis Res Ther 2021; 23:286. [PMID: 34784954 PMCID: PMC8594229 DOI: 10.1186/s13075-021-02668-8] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Accepted: 10/31/2021] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Gasdermin D (GSDMD) is cleaved by several proteases including by caspase-1, a component of intracellular protein complexes called inflammasomes. Caspase-1 also converts pro-interleukin-1β (pro-IL-1β) and pro-IL-18 into bioactive IL-1β and IL-18, respectively. GSDMD amino-terminal fragments form plasma membrane pores, which mediate the secretion of IL-1β and IL-18 and cause the inflammatory form of cell death pyroptosis. Here, we tested the hypothesis that GSDMD contributes to joint degeneration in the K/BxN serum transfer-induced arthritis (STIA) model in which autoantibodies against glucose-6-phosphate isomerase promote the formation of pathogenic immune complexes on the surface of myeloid cells, which highly express the inflammasomes. The unexpected outcomes with the STIA model prompted us to determine the role of GSDMD in the post-traumatic osteoarthritis (PTOA) model caused by meniscus ligamentous injury (MLI) based on the hypothesis that this pore-forming protein is activated by signals released from damaged joint tissues. METHODS Gsdmd +/+ and Gsdmd-/- mice were injected with K/BxN mouse serum or subjected to MLI to cause STIA or PTOA, respectively. Paw and ankle swelling and DXA scanning were used to assess the outcomes in the STIA model whereas histopathology and micro-computed tomography (μCT) were utilized to monitor joints in the PTOA model. Murine and human joint tissues were also examined for GSDMD, IL-1β, and IL-18 expression by qPCR, immunohistochemistry, or immunoblotting. RESULTS GSDMD levels were higher in serum-inoculated paws compared to PBS-injected paws. Unexpectedly, ablation of GSDMD failed to reduce joint swelling and osteolysis, suggesting that GSDMD was dispensable for the pathogenesis of STIA. GSDMD levels were also higher in MLI compared to sham-operated joints. Importantly, ablation of GSDMD attenuated MLI-associated cartilage degradation (p = 0.0097), synovitis (p = 0.014), subchondral bone sclerosis (p = 0.0006), and subchondral bone plate thickness (p = 0.0174) based on histopathological and μCT analyses. CONCLUSION GSDMD plays a key role in the pathogenesis of PTOA, but not STIA, suggesting that its actions in experimental arthropathy are tissue context-specific.
Collapse
Affiliation(s)
- Tong Yang
- Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, China
- Division of Bone and Mineral Diseases, Washington University School of Medicine, 660 South Euclid Avenue, Campus Box 8301, St. Louis, MO, 63110, USA
| | - Kai Sun
- Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, China
- Division of Bone and Mineral Diseases, Washington University School of Medicine, 660 South Euclid Avenue, Campus Box 8301, St. Louis, MO, 63110, USA
| | - Chun Wang
- Division of Bone and Mineral Diseases, Washington University School of Medicine, 660 South Euclid Avenue, Campus Box 8301, St. Louis, MO, 63110, USA
| | - Gaurav Swarnkar
- Department of Orthopaedic Surgery, Washington University School of Medicine, St. Louis, MO, USA
| | - Songtao Quan
- Luoyang Orthopedic - Traumatological Hospital of Henan Province, Luoyang, Henan, China
| | - Dustin Kress
- Division of Bone and Mineral Diseases, Washington University School of Medicine, 660 South Euclid Avenue, Campus Box 8301, St. Louis, MO, 63110, USA
| | - Jianqiu Xiao
- Division of Bone and Mineral Diseases, Washington University School of Medicine, 660 South Euclid Avenue, Campus Box 8301, St. Louis, MO, 63110, USA
| | - Yael Alippe
- Division of Bone and Mineral Diseases, Washington University School of Medicine, 660 South Euclid Avenue, Campus Box 8301, St. Louis, MO, 63110, USA
| | - Hongjun Zheng
- Department of Orthopaedic Surgery, Washington University School of Medicine, St. Louis, MO, USA
| | - Robert H Brophy
- Department of Orthopaedic Surgery, Washington University School of Medicine, St. Louis, MO, USA
| | - Dingjun Hao
- Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, China
| | - Audrey McAlinden
- Department of Orthopaedic Surgery, Washington University School of Medicine, St. Louis, MO, USA
- Shriners Hospital for Children, St. Louis, MO, USA
| | - Yousef Abu-Amer
- Department of Orthopaedic Surgery, Washington University School of Medicine, St. Louis, MO, USA
- Shriners Hospital for Children, St. Louis, MO, USA
| | - Jie Shen
- Department of Orthopaedic Surgery, Washington University School of Medicine, St. Louis, MO, USA
| | - Gabriel Mbalaviele
- Division of Bone and Mineral Diseases, Washington University School of Medicine, 660 South Euclid Avenue, Campus Box 8301, St. Louis, MO, 63110, USA.
| |
Collapse
|
47
|
Cheng Y, Zhao Y, Zheng Y. Therapeutic potential of triptolide in autoimmune diseases and strategies to reduce its toxicity. Chin Med 2021; 16:114. [PMID: 34743749 PMCID: PMC8572577 DOI: 10.1186/s13020-021-00525-z] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Accepted: 10/27/2021] [Indexed: 12/18/2022] Open
Abstract
With the increasing epidemiology of autoimmune disease worldwide, there is an urgent need for effective drugs with low cost in clinical treatment. Triptolide, the most potent bioactive compound from traditional Chinese herb Tripterygium Wilfordii Hook F, possesses immunosuppression and anti-inflammatory activity. It is a potential drug for the treatment of various autoimmune diseases, but its clinical application is still restricted due to severe toxicity. In this review, the pharmacodynamic effects and pharmacological mechanisms of triptolide in autoimmune diseases are summarized. Triptolide exerts therapeutic effect by regulating the function of immune cells and the expression of cytokines through inflammatory signaling pathways, as well as maintaining redox balance and gut microbiota homeostasis. Meanwhile, the research progress on toxicity of triptolide to liver, kidney, reproductive system, heart, spleen, lung and gastrointestinal tract has been systematically reviewed. In vivo experiments on different animals and clinical trials demonstrate the dose- and time- dependent toxicity of triptolide through different administration routes. Furthermore, we focus on the strategies to reduce toxicity of triptolide, including chemical structural modification, novel drug delivery systems, and combination pharmacotherapy. This review aims to reveal the potential therapeutic prospect and limitations of triptolide in treating autoimmune diseases, thus providing guiding suggestions for further study and promoting its clinical translation.
Collapse
Affiliation(s)
- Yaxin Cheng
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau, China
| | - Yonghua Zhao
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau, China. .,Guangdong-Hong Kong-Macau Joint Lab on Chinese Medicine and Immune Disease Research, University of Macau, Macau, China.
| | - Ying Zheng
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau, China.
| |
Collapse
|
48
|
Targeting of Janus Kinases Limits Pro-Inflammatory but Also Immunosuppressive Circuits in the Crosstalk between Synovial Fibroblasts and Lymphocytes. Biomedicines 2021; 9:biomedicines9101413. [PMID: 34680530 PMCID: PMC8533088 DOI: 10.3390/biomedicines9101413] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Revised: 10/01/2021] [Accepted: 10/04/2021] [Indexed: 12/29/2022] Open
Abstract
Crosstalk between synovial fibroblasts (SF) and immune cells plays a central role in the development of rheumatoid arthritis (RA). Janus kinase inhibitors (JAKi) have proven efficacy in the treatment of RA, although clinical responses are heterogeneous. Currently, little is known regarding how JAKi affect pro- and anti-inflammatory circuits in the bidirectional interplay between SF and immune cells. Here, we examined the effects of tofacitinib, baricitinib and upadacitinib on crosstalk between SF and T or B lymphocytes in vitro and compared them with those of biologic disease modifying anti-rheumatic drugs (bDMARDs). JAKi dose-dependently suppressed cytokine secretion of T helper (Th) cells and decreased interleukin (IL)-6 and matrix metalloproteinase (MMP)3 secretion of SF stimulated by Th cells. Importantly, JAK inhibition attenuated the enhanced memory response of chronically stimulated SF. Vice versa, JAKi reduced the indoleamine-2,3-dioxygenase (IDO)1-mediated suppression of T cell-proliferation by SF. Remarkably, certain bDMARDs were as efficient as JAKi in suppressing the IL-6 and MMP3 secretion of SF stimulated by Th (adalimumab, secukinumab) or B cells (canakinumab) and combining bDMARDs with JAKi had synergistic effects. In conclusion, JAKi limit pro-inflammatory circuits in the crosstalk between SF and lymphocytes; however, they also weaken the immunosuppressive functions of SF. Both effects were dose-dependent and may contribute to heterogeneity in clinical response to treatment.
Collapse
|
49
|
Makkar R, Behl T, Bungau S, Kumar A, Arora S. Understanding the Role of Inflammasomes in Rheumatoid Arthritis. Inflammation 2021; 43:2033-2047. [PMID: 32712858 DOI: 10.1007/s10753-020-01301-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Inflammasomes are the molecular pathways that activate upon conditions of infection or stress and trigger the activation and maturation of inflammatory cytokines. Immune reactions in conjugation with inflammatory processes play a pivotal role in developing innumerable diseases. An over reactive immune system fabricates many allergic and hypersensitive reactions in response to autoantibodies activated against modified self-epitopes and similar molecules. Rheumatoid arthritis (RA) is a complex autoimmune inflammatory disorder commencing with inflammation in small joints like hands, knees, and wrist eventually entrapping larger joints such as spine. The formation of autoantibodies called rheumatoid factor (RF) and citrullinated proteins against immunoglobulin G symbolizes autoimmune nature of the disease. The presence of autoantibodies embarks principal diagnostic hallmark of the disease. With the advancement of technology, the therapeutic approach is also advancing. A new era of molecules, namely inflammasomes, are activated upon infection or in response to stress and trigger the activation of various proinflammatory cytokines such interleukins which engage in the defense mechanism of the innate immunity. Robust linking among the activity of dysregulated inflammasomes and the heritable acquired inflammatory diseases and disorders emphasizes the significance of this pathway in altering the immune responses. The current review highlights the functioning of inflammasomes and their possible role in disease dysregulation.
Collapse
Affiliation(s)
- Rashita Makkar
- Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, India
| | - Tapan Behl
- Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, India.
| | - Simona Bungau
- Department of Pharmacy, Faculty of Medicine and Pharmacy, University of Oradea, Oradea, Romania
| | - Arun Kumar
- Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, India
| | - Sandeep Arora
- Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, India
| |
Collapse
|
50
|
Elzokm SS, Fouda MA, Abdel Moneim RA, El-Mas MM. Distinct effects of calcineurin dependent and independent immunosuppressants on endotoxaemia-induced nephrotoxicity in rats: Role of androgens. Clin Exp Pharmacol Physiol 2021; 48:1261-1270. [PMID: 34042216 DOI: 10.1111/1440-1681.13526] [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: 01/19/2021] [Revised: 05/05/2021] [Accepted: 05/24/2021] [Indexed: 11/26/2022]
Abstract
Evidence suggests that immunosuppressant therapies protect against harmful effects of endotoxaemia. In this study, we tested whether calcineurin-dependent (cyclosporine/tacrolimus) and -independent (sirolimus) immunosuppressants variably influence nephrotoxicity induced by endotoxaemia and whether this interaction is modulated by testosterone. We investigated the effects of immunosuppressants on renal histopathological, biochemical and inflammatory profiles in endotoxic male rats and the role of androgenic state in the interaction. Six-hour treatment of rats with lipopolysaccharide (LPS, 3 mg/kg) increased (i) serum urea/creatinine, (ii) width of proximal/distal tubules, (iii) tubular degeneration and vacuolation, (iv) Western protein expressions of renal toll-like receptor 4, monocyte chemoattractant protein-1, and NADPH oxidase-2, and (v) serum tumour necrosis factor-α and myeloperoxidase. These endotoxic manifestations were intensified and eliminated upon concurrent exposure to cyclosporine and sirolimus, respectively. The cyclosporine actions appear to be a class rather than a drug effect because similar exacerbation of LPS nephrotoxicity was observed in rats treated with tacrolimus, another calcineurin inhibitor (CNI). Moreover, the deteriorated renal outcomes in LPS/tacrolimus-treated rats were reduced after castration or androgen receptor blockade by flutamide. The data suggest opposite effects for calcineurin-dependent (exaggeration) and -independent immunosuppressants (amelioration) on renal defects of endotoxaemia and implicate androgenic pathways in the worsened endotoxic renal profile induced by CNIs.
Collapse
Affiliation(s)
- Shrouk S Elzokm
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Pharos University in Alexandria, Alexandria, Egypt
| | - Mohamed A Fouda
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Alexandria University, Alexandria, Egypt
| | - Rahab A Abdel Moneim
- Department of Histology, Faculty of Medicine, Alexandria University, Alexandria, Egypt
| | - Mahmoud M El-Mas
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Alexandria University, Alexandria, Egypt
- Department of Pharmacology and Toxicology, Faculty of Medicine, Kuwait University, Kuwait City, Kuwait
| |
Collapse
|