1
|
Park M, Kim YS, Song H. Macrophages: a double-edged sword in female reproduction and disorders. Exp Mol Med 2025:10.1038/s12276-025-01392-6. [PMID: 39894821 DOI: 10.1038/s12276-025-01392-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Revised: 10/31/2024] [Accepted: 11/12/2024] [Indexed: 02/04/2025] Open
Abstract
Reproduction consists of sequential inflammation-like events, primarily within the endometrium, from ovulation to embryo implantation, decidualization and delivery. During the reproductive cycle, the endometrium repeatedly undergoes cyclic periods of proliferation, differentiation, tissue breakdown and repair without scarring. Owing to their phagocytic activity, macrophages, key players in innate immunity, are thought to play crucial roles in the endometrium. Endometrial macrophages actively participate in various stages of reproductive tissue remodeling, particularly during decidualization and pregnancy establishment. Traditionally considered simple bystanders that clear debris to prevent autoimmune responses in tissue homeostasis, macrophages are now recognized as main actors with broad functional plasticity that allows them to fine tune the balance between pro- and anti-inflammatory responses during tissue inflammation, remodeling and repair. Homeostatic balance is determined by the sum of various mediators produced by two distinctly polarized macrophage subpopulations. The biased polarization of tissue-resident macrophages may contribute to the pathogenesis of various diseases, such as inflammation and cancer. Thus, understanding how macrophages contribute to endometrial homeostasis is crucial for deciphering the underlying mechanisms of various reproductive disorders. Nanomedicines using extracellular vesicles, nanoparticles and noncoding RNAs have recently been applied to modulate macrophage polarization and alleviate disease phenotypes. Despite these advances, the functions of endometrial macrophages under physiological and pathophysiological conditions remain poorly understood, which complicates the development of targeted therapies. Here we update the current understanding of the homeostatic function of macrophages and the putative contribution of endometrial macrophage dysfunction to reproductive disorders in women, along with innovative molecular therapeutics to resolve this issue.
Collapse
Affiliation(s)
- Mira Park
- Department of Biomedical Science, College of Life Science, CHA University, Pocheon, Korea
| | - Yeon Sun Kim
- Department of Biomedical Science, College of Life Science, CHA University, Pocheon, Korea
| | - Haengseok Song
- Department of Biomedical Science, College of Life Science, CHA University, Pocheon, Korea.
- Division of Life Science, CHA University, Pocheon, Korea.
- Department of Life Science, Graduate School, CHA University, Pocheon, Korea.
- CHA Advanced Research Institute, Seongnam, Korea.
- KW-Bio Co., Chuncheon, Korea.
| |
Collapse
|
2
|
Wang X, Ni T, Miao J, Huang X, Feng Z. The role and mechanism of triptolide, a potential new DMARD, in the treatment of rheumatoid arthritis. Ageing Res Rev 2025; 104:102643. [PMID: 39722411 DOI: 10.1016/j.arr.2024.102643] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2024] [Revised: 12/09/2024] [Accepted: 12/16/2024] [Indexed: 12/28/2024]
Abstract
Triptolide (TP) is the primary pharmacological component of Tripterygium Glycosides (TG), which has anti-inflammatory, antiproliferative, and immunosuppressive properties, among other pharmacological actions, and has excellent potential for developing into a new DMARD. We have reviewed the effects and mechanisms of TP on immunosuppression, inhibiting synovial proliferation, and preventing articular bone destruction in the treatment of rheumatoid arthritis (RA), which is a common disease in the elderly in this paper. We have found that TP has regulatory effects on multiple vital cells in the above-mentioned pathological process of RA, such as monocytes/macrophages, dendritic cells, T cells, fibroblast-like synoviocytes, and osteoclasts. We also found that TP can regulate multiple key signaling pathways such as NF-κB, JAK/STAT, and MAPK through various molecular regulatory mechanisms, achieving regulatory effects on numerous phenotypes of the above-mentioned vital cells.
Collapse
Affiliation(s)
- Xiwen Wang
- The First Clinical Medical College, Nanjing University of Chinese Medicine, No. 138 Xianlin Avenue, Nanjing 210023, China
| | - Tianyang Ni
- The First Clinical Medical College, Nanjing University of Chinese Medicine, No. 138 Xianlin Avenue, Nanjing 210023, China
| | - Jianru Miao
- The First Clinical Medical College, Nanjing University of Chinese Medicine, No. 138 Xianlin Avenue, Nanjing 210023, China
| | - Xinyao Huang
- The First Clinical Medical College, Nanjing University of Chinese Medicine, No. 138 Xianlin Avenue, Nanjing 210023, China
| | - Zhe Feng
- The First Clinical Medical College, Nanjing University of Chinese Medicine, No. 138 Xianlin Avenue, Nanjing 210023, China.
| |
Collapse
|
3
|
Zewail MB, Doghish AS, El-Husseiny HM, Mady EA, Mohammed OA, Elbadry AMM, Elbokhomy AS, Bhnsawy A, El-Dakroury WA. Lipid-based nanocarriers: an attractive approach for rheumatoid arthritis management. Biomater Sci 2024; 12:6163-6195. [PMID: 39484700 DOI: 10.1039/d4bm01058b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2024]
Abstract
Lipid nanoparticles (LNPs) have emerged as transformative tools in modern drug delivery, offering unparalleled potential in enhancing the efficacy and safety of various therapeutics. In the context of rheumatoid arthritis (RA), a disabling autoimmune disorder characterized by chronic inflammation, joint damage, and limited patient mobility, LNPs hold significant promise for revolutionizing treatment strategies. LNPs offer several advantages over traditional drug delivery systems, including improved pharmacokinetics, enhanced tissue penetration, and reduced systemic toxicity. This article concisely summarizes the pathogenesis of RA, its associated risk factors, and therapeutic techniques and their challenges. Additionally, it highlights the noteworthy advancements made in managing RA through LNPs, including liposomes, niosomes, bilosomes, cubosomes, spanlastics, ethosomes, solid lipid nanoparticles, lipid micelles, lipid nanocapsules, nanostructured lipid carriers, etc. It also delves into the specific functional attributes of these nanocarrier systems, focusing on their role in treating and monitoring RA.
Collapse
Affiliation(s)
- Moataz B Zewail
- School of Chemical Engineering, Faculty of Sciences, Engineering and Technology, The University of Adelaide, Adelaide, SA, 5005, Australia
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Badr University in Cairo (BUC), Badr City, Cairo 11829, Egypt.
| | - Ahmed S Doghish
- Department of Biochemistry, Faculty of Pharmacy, Badr University in Cairo (BUC), Badr City, 17 Cairo, 11829, Egypt
- Biochemistry and Molecular Biology Department, Faculty of Pharmacy (Boys), Al-Azhar University, Nasr City 11231, Cairo, Egypt
| | - Hussein M El-Husseiny
- Laboratory of Veterinary Surgery, Department of Veterinary Medicine, Faculty of Agriculture, Tokyo University of Agriculture and Technology, 3-5-8 Saiwai Cho, Fuchu-shi, Tokyo 183-8509, Japan
- Department of Surgery, Anesthesiology, and Radiology, Faculty of Veterinary Medicine, Benha University, Moshtohor, Toukh, Elqaliobiya, 13736, Egypt
- Laboratory of Veterinary Physiology, Department of Veterinary Medicine, Tokyo University of Agriculture and Technology, Tokyo 183-8509, Japan
- Institute of Global Innovation Research, Tokyo University of Agriculture and Technology, 3-8-1 Harumi-cho, Fuchu-shi, Tokyo 183-8538, Japan
| | - Eman A Mady
- Laboratory of Veterinary Physiology, Department of Veterinary Medicine, Faculty of Agriculture, 10 Tokyo University of Agriculture and Technology, 3-5-8 Saiwai Cho, Fuchu-shi, Tokyo 183-8509, Japan
- Department of Animal Hygiene, Behavior, and Management, Faculty of Veterinary Medicine, Benha University, Moshtohor, Toukh, Elqaliobiya 13736, Egypt
| | - Osama A Mohammed
- Department of Pharmacology, College of Medicine, University of Bisha, Bisha 61922, Saudi Arabia
| | - Abdullah M M Elbadry
- Badr University in Cairo Research Center, Badr University in Cairo, Badr City, Cairo 11829, Egypt
| | - Amir S Elbokhomy
- Faculty of Pharmacy, Badr University in Cairo (BUC), Badr City, Cairo 11829, Egypt
| | - Abdelmenem Bhnsawy
- Faculty of Pharmacy, Badr University in Cairo (BUC), Badr City, Cairo 11829, Egypt
| | - Walaa A El-Dakroury
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Badr University in Cairo (BUC), Badr City, Cairo 11829, Egypt.
| |
Collapse
|
4
|
Bi J, Zeng J, Liu X, Mo C, Yao M, Zhang J, Yuan P, Jia B, Xu S. Drug delivery for age-related bone diseases: From therapeutic targets to common and emerging therapeutic strategies. Saudi Pharm J 2024; 32:102209. [PMID: 39697472 PMCID: PMC11653637 DOI: 10.1016/j.jsps.2024.102209] [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: 09/12/2024] [Accepted: 11/22/2024] [Indexed: 12/20/2024] Open
Abstract
With the accumulation of knowledge on aging, people have gradually realized that among the many factors that cause individual aging, the accumulation of aging cells is an essential cause of organ degeneration and, ultimately, age-related diseases. Most cells present in the bone microenvironment gradually age over time, leading to an imbalance of osteogenesis, osteoclastogenesis, adipogenesis, and chondrogenesis. This imbalance contributes to age-related bone loss and the development of age-related bone diseases, such as osteoporosis. Bone aging can prolong the lifespan and delay the development of age-related diseases. Nanoparticles have controllable and stable physical and chemical properties and can precisely target different tissues and organs. By preparing multiple easily modified and biocompatible nanoparticles as different drug delivery carriers, specifically targeting various diseased tissues for controlled-release and sustained-release administration, the delivery efficiency of drugs can be significantly improved, and the toxicity and side effects of drugs can be substantially reduced, thereby improving the therapeutic effect of age-related bone diseases. In addition, other novel anti-aging strategies (such as stem cell exosomes) also have significant scientific and practical significance in anti-aging research on age-related bone diseases. This article reviews the research progress of various nano-drug-loaded particles and emerging anti-aging methods for treating age-related bone diseases, offering new insights and directions for precise targeted clinical therapies.
Collapse
Affiliation(s)
- Jiaming Bi
- Department of Endodontics, Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, Guangdong, China
| | - Jiawei Zeng
- Department of Endodontics, Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, Guangdong, China
| | - Xiaohao Liu
- Department of Periodontics, Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, Guangdong, China
| | - Chuzi Mo
- Department of Endodontics, Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, Guangdong, China
| | - Mingyan Yao
- Department of Endocrinology, Baoding No.1 Central Hospital, Baoding, China
| | - Jing Zhang
- Department of Cardiology, Affiliated Hospital of Hebei University, Baoding, China
| | - Peiyan Yuan
- Department of Endodontics, Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, Guangdong, China
| | - Bo Jia
- Department of Oral and Maxillofacial Surgery, Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, Guangdong, China
| | - Shuaimei Xu
- Department of Endodontics, Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, Guangdong, China
| |
Collapse
|
5
|
Shen Z, Wang X, Lu L, Wang R, Hu D, Fan Z, Zhu L, Zhong R, Wu M, Zhou X, Cao X. Bilirubin-Modified Chondroitin Sulfate-Mediated Multifunctional Liposomes Ameliorate Acute Kidney Injury by Inducing Mitophagy and Regulating Macrophage Polarization. ACS APPLIED MATERIALS & INTERFACES 2024; 16:62693-62709. [PMID: 39492707 DOI: 10.1021/acsami.4c14169] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/05/2024]
Abstract
Acute kidney injury (AKI) is a dynamic process associated with inflammation, oxidative stress, and lipid peroxidation, in which mitochondrial mitophagy and macrophage polarization play a critical role in the pathophysiology. Based on the expression of the CD44 receptor on renal tubular epithelial cells (RTECs) and activated M1 macrophages being abnormally increased, accompanied by up-regulation of reactive oxygen species (ROS) during AKI, the conjugates of bilirubin (BR), an endogenous antioxidant which has the property of anti-inflammation, and chondroitin sulfate (CS) with CD44-targeting property could be a promising therapeutic carrier. In this study, we develop a CD44-targeted/ROS-responsive CS-BR-mediated multifunctional liposome loading celastrol (CS-BR@CLT) for the targeted therapy of AKI. CS-BR@CLT is shown to selectively accumulate in AKI mouse kidneys via targeting of CD44 receptors. Treatment with CS-BR@CLT significantly ameliorates acute kidney injury caused by ischemia-reperfusion and protects renal function. Mechanistically, CS-BR@CLT inhibits apoptosis, protects mitochondria, promotes autophagy, regulates macrophage polarization, and alleviates interstitial inflammation. Overall, our study demonstrates that CS-BR@CLT could be a promising strategy to ameliorate acute kidney injury.
Collapse
Affiliation(s)
- Ziqi Shen
- School of Pharmacy, the First Affiliated Hospital of Anhui Medical University, Anhui Medical University, Hefei, Anhui 230031, China
- Department of Pharmacy, the First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230031, China
- The Grade 3 Pharmaceutical Chemistry Laboratory of State Administration of Traditional Chinese Medicine, Hefei, Anhui 230031, China
- Sichuan Provincial Orthopedic Hospital, Chengdu 610041, China
| | - Xiaohua Wang
- Department of Pharmacy, the First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230031, China
- The Grade 3 Pharmaceutical Chemistry Laboratory of State Administration of Traditional Chinese Medicine, Hefei, Anhui 230031, China
| | - Li Lu
- School of Pharmacy, the First Affiliated Hospital of Anhui Medical University, Anhui Medical University, Hefei, Anhui 230031, China
- Department of Pharmacy, the First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230031, China
- The Grade 3 Pharmaceutical Chemistry Laboratory of State Administration of Traditional Chinese Medicine, Hefei, Anhui 230031, China
| | - Runkong Wang
- Sichuan Provincial Orthopedic Hospital, Chengdu 610041, China
| | - Danni Hu
- School of Pharmacy, the First Affiliated Hospital of Anhui Medical University, Anhui Medical University, Hefei, Anhui 230031, China
- Department of Pharmacy, the First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230031, China
- The Grade 3 Pharmaceutical Chemistry Laboratory of State Administration of Traditional Chinese Medicine, Hefei, Anhui 230031, China
| | - Ziyan Fan
- School of Pharmacy, the First Affiliated Hospital of Anhui Medical University, Anhui Medical University, Hefei, Anhui 230031, China
- Department of Pharmacy, the First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230031, China
- The Grade 3 Pharmaceutical Chemistry Laboratory of State Administration of Traditional Chinese Medicine, Hefei, Anhui 230031, China
| | - Liyang Zhu
- Sichuan Provincial Orthopedic Hospital, Chengdu 610041, China
| | - Ruixue Zhong
- Sichuan Provincial Orthopedic Hospital, Chengdu 610041, China
| | - Mingquan Wu
- Sichuan Provincial Orthopedic Hospital, Chengdu 610041, China
| | - Xu Zhou
- Sichuan Provincial Orthopedic Hospital, Chengdu 610041, China
| | - Xi Cao
- School of Pharmacy, the First Affiliated Hospital of Anhui Medical University, Anhui Medical University, Hefei, Anhui 230031, China
- Department of Pharmacy, the First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230031, China
- The Grade 3 Pharmaceutical Chemistry Laboratory of State Administration of Traditional Chinese Medicine, Hefei, Anhui 230031, China
| |
Collapse
|
6
|
Xu L, Qin J, Ma X, Wang Q, Wu W, Huang H, Cai L. Chitosan-based self-healing thermosensitive hydrogel loaded with siHMGB1 for treatment of rheumatoid arthritis via macrophage repolarization. Int J Biol Macromol 2024; 282:137102. [PMID: 39486712 DOI: 10.1016/j.ijbiomac.2024.137102] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Revised: 10/17/2024] [Accepted: 10/29/2024] [Indexed: 11/04/2024]
Abstract
Rheumatoid arthritis (RA) is a prevalent autoimmune disease marked by immune cell activation, particularly macrophages. An imbalance between pro-inflammatory M1 and anti-inflammatory M2 macrophages causes synovial inflammation and joint damage, worsening RA. This study presents a biomacromolecular hydrogel delivery system with apoferritin nanoparticles for effective delivery of small interfering high mobility group protein (siHMGB1). The system was designed to promote the polarization of M1 macrophages to the M2 phenotype by downregulating the HMGB1/TLR4/NF-κB-p65 signaling pathway, offering a potential therapeutic approach for the treatment of RA. The oxidized chondroitin sulfate - chitosan - sodium glycerol β - phosphate - Fn/siHMGB1 (OCF/siHMGB1) hydrogel system possessed temperature-sensitive and self-healing properties, enabling the sustained, stable, and efficient release of siHMGB1 at the affected joint. After effective uptake by macrophages, siHMGB1 could effectively repolarize M1-phenotype macrophages to M2-phenotype via the HMGB1/TLR4/NF-κB-p65 signaling pathway both in vitro and in vivo. Additionally, it suppressed the release of pro-inflammatory cytokines and upregulated anti-inflammatory cytokines, which significantly blocked the TLR4/p65-mediated inflammatory signaling. In conclusion, the siHMGB1-loaded hydrogel delivery system designed in this study is effective in treating RA and highlights the potential of gene therapy to induce repolarization of M1 to M2 macrophages for RA treatment.
Collapse
Affiliation(s)
- Lixing Xu
- Department of Pharmacy, Affiliated Hospital of Nantong University, Pharmacy School of Nantong University, Nantong 226001, China
| | - Jisu Qin
- Department of Pharmacy, Affiliated Hospital of Nantong University, Pharmacy School of Nantong University, Nantong 226001, China
| | - Xiaofei Ma
- Department of Pharmacy, Affiliated Hospital of Nantong University, Pharmacy School of Nantong University, Nantong 226001, China
| | - Qin Wang
- Department of Pharmacy, Affiliated Hospital of Nantong University, Pharmacy School of Nantong University, Nantong 226001, China
| | - Wenyi Wu
- Department of quality inspection, Sinopharm holding Nantong Ltd, Nantong 226001, China
| | - Haiqin Huang
- Department of Pharmacy, Affiliated Hospital of Nantong University, Pharmacy School of Nantong University, Nantong 226001, China.
| | - Liangliang Cai
- Department of Pharmacy, Affiliated Hospital of Nantong University, Pharmacy School of Nantong University, Nantong 226001, China.
| |
Collapse
|
7
|
Zhang B, Guo Y, Lu Y, Ma D, Wang X, Zhang L. Bibliometric and visualization analysis of the application of inorganic nanomaterials to autoimmune diseases. Biomater Sci 2024; 12:3981-4005. [PMID: 38979695 DOI: 10.1039/d3bm02015k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/10/2024]
Abstract
Objective: To conduct bibliometric analysis of the application of inorganic nanomaterials to autoimmune diseases to characterize current research trends and to visualize past and emerging trends in this field in the past 15 years. Methods: The evolution and thematic trends of the application of inorganic nanomaterials to autoimmune diseases from January 1, 1985, to March 15, 2024, were analyzed by bibliometric analysis of data retrieved and extracted from the Web of Science Core Collection (WoSCC) database. A total of 734 relevant reports in the literature were evaluated according to specific characteristics such as year of publication, journal, institution, country/region, references, and keywords. VOSviewer was used to build co-authorship analysis, co-occurrence analysis, co-citation analysis, and network visualization. Some important subtopics identified by bibliometric characterization are further discussed and reviewed. Result: From 2009 to 2024, annual publications worldwide increased from 11 to 95, an increase of 764%. ACS Nano published the most papers (14) with the most citations (1372). China (230 papers, 4922 citations) and the Chinese Academy of Sciences (36 papers, 718 citations) are the most productive and influential country and institution, respectively. The first 100 keywords were co-clustered to form four clusters: (1) the application of inorganic nanomaterials in drug delivery, (2) the application of inorganic nano-biosensing to autoimmune diseases, (3) the use of inorganic nanomaterials for imaging applied to autoimmune diseases, and (4) the application of inorganic nanomaterials in the treatment of autoimmune diseases. Combination therapy, microvesicles, photothermal therapy (PTT), targeting, diagnostics, transdermal, microneedling, silver nanoparticles, psoriasis, and inflammatory cytokines are the latest high-frequency keywords, marking the emerging frontier of inorganic nanomaterials in the field of autoimmune diseases. Sub-topics were further discussed to help researchers determine the scope of research topics and plan research directions. Conclusion: Over the past 39 years, the application of inorganic nanotechnology to the field of autoimmune diseases shows extensive cooperation between countries and institutions, showing a continuous increase in the number of reports in the literature, and has clinical translation prospects. Future research should further improve the safety of inorganic nanomaterials, clarify the mechanism of action of nanomaterials, establish a standardized nanomaterial preparation and performance evaluation system, and ultimately achieve the goal of early detection and precise treatment of autoimmune diseases.
Collapse
Affiliation(s)
- Baiyan Zhang
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Taiyuan, 030032, China
- School of Pharmacy, Shanxi Medical University, Jinzhong 030619, Shanxi, China
| | - Yuanyuan Guo
- School of Pharmacy, Shanxi Medical University, Jinzhong 030619, Shanxi, China
| | - Yu Lu
- The First Clinical Medical College of Shanxi Medical University, Jinzhong 030619, Shanxi, China
| | - Dan Ma
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Taiyuan, 030032, China
| | - Xiahui Wang
- School of Pharmacy, Shanxi Medical University, Jinzhong 030619, Shanxi, China
| | - Liyun Zhang
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Taiyuan, 030032, China
| |
Collapse
|
8
|
Gan J, Zhang X, Chen G, Hao X, Zhao Y, Sun L. CXCR4-Expressing Mesenchymal Stem Cells Derived Nanovesicles for Rheumatoid Arthritis Treatment. Adv Healthc Mater 2024; 13:e2303300. [PMID: 38145406 DOI: 10.1002/adhm.202303300] [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: 09/28/2023] [Revised: 11/21/2023] [Indexed: 12/26/2023]
Abstract
Cell membrane camouflage technology, which a demonstrated value for the bionic replication of natural cell membrane properties, is an active area of ongoing research readily applicable to nanomedicine. How to realize immune evasion, slow down the clearance from the body, and improve targeting are still worth great efforts for this technology. Herein, novel cell membrane-mimicked nanovesicles from genetically engineered mesenchymal stem cells (MSCs) are presented as a potential anti-inflammatory platform for rheumatoid arthritis (RA) management. Utilizing the synthetic biology approach, the biomimetic nanoparticles are constructed by fusing C-X-C motif chemokine receptor4 (CXCR4)-anchored MSC membranes onto drug-loaded polymeric cores (MCPNs), which make them ideal decoys of stromal cell-derived factor-1 (SDF-1)-targeted arthritis. These resulting nanocomplexes function to escape from the immune system and enhance accumulation in the established inflamed joints via the CXCR4/SDF-1 chemotactic signal axis, thereby achieving an affinity to activated macrophages and synovial fibroblasts. It is further demonstrated that the MCPNs can significantly suppress synovial inflammation and relieve pathological conditions with favorable safety properties in collagen-induced arthritis mice. These findings indicate the clinical value of MCPNs as biomimetic nanodrugs for RA therapy and related diseases.
Collapse
Affiliation(s)
- Jingjing Gan
- Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, Medical School, Nanjing University, Nanjing, 210002, China
| | - Xiaoxuan Zhang
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096, China
| | - Guangcai Chen
- Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, Medical School, Nanjing University, Nanjing, 210002, China
| | - Xubin Hao
- Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, Medical School, Nanjing University, Nanjing, 210002, China
| | - Yuanjin Zhao
- Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, Medical School, Nanjing University, Nanjing, 210002, China
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096, China
| | - Lingyun Sun
- Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, Medical School, Nanjing University, Nanjing, 210002, China
- Department of Rheumatology and Immunology, The First Affliated Hospital of Anhui Medical University, 218 Jixi Road, Hefei, 230022, China
| |
Collapse
|
9
|
Huang X, Zhang W. Macrophage membrane-camouflaged biomimetic nanovesicles for targeted treatment of arthritis. Ageing Res Rev 2024; 95:102241. [PMID: 38387516 DOI: 10.1016/j.arr.2024.102241] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2023] [Revised: 02/03/2024] [Accepted: 02/19/2024] [Indexed: 02/24/2024]
Abstract
Arthritis has become the most common joint disease globally. Current attention has shifted towards preventing the disease and exploring pharmaceutical and surgical treatments for early-stage arthritis. M2 macrophages are known for their anti-inflammatory properties and their ability to support cartilage repair, offering relief from arthritis. Whereas, it remains a great challenge to promote the beneficial secretion of M2 macrophages to prevent the progression of arthritis. Therefore, it is warranted to investigate new strategies that could use the functions of M2 macrophages and enhance its therapeutic effects. This review aims to explore the macrophage cell membrane-coated biomimetic nanovesicles for targeted treatment of arthritis such as osteoarthritis (OA), rheumatoid arthritis (RA), and gouty arthritis (GA). Cell membrane-camouflaged biomimetic nanovesicle has attracted increasing attention, which successfully combine the advantages and properties of both cell membrane and delivered drug. We discuss the roles of macrophages in the pathophysiology and therapeutic targets of arthritis. Then, the common preparation strategies of macrophage membrane-coated nanovesicles are concluded. Moreover, we investigate the applications of macrophage cell membrane-camouflaged nanovesicles for arthritis, such as OA, RA, and GA. Taken together, macrophage cell membrane-camouflaged nanovesicles hold the tremendous prospect for biomedical applications in the targeted treatment of arthritis.
Collapse
Affiliation(s)
- Xin Huang
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Weiyue Zhang
- Department of Endocrinology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China.
| |
Collapse
|
10
|
Zerrouk N, Alcraft R, Hall BA, Augé F, Niarakis A. Large-scale computational modelling of the M1 and M2 synovial macrophages in rheumatoid arthritis. NPJ Syst Biol Appl 2024; 10:10. [PMID: 38272919 PMCID: PMC10811231 DOI: 10.1038/s41540-024-00337-5] [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: 09/18/2023] [Accepted: 01/11/2024] [Indexed: 01/27/2024] Open
Abstract
Macrophages play an essential role in rheumatoid arthritis. Depending on their phenotype (M1 or M2), they can play a role in the initiation or resolution of inflammation. The M1/M2 ratio in rheumatoid arthritis is higher than in healthy controls. Despite this, no treatment targeting specifically macrophages is currently used in clinics. Thus, devising strategies to selectively deplete proinflammatory macrophages and promote anti-inflammatory macrophages could be a promising therapeutic approach. State-of-the-art molecular interaction maps of M1 and M2 macrophages in rheumatoid arthritis are available and represent a dense source of knowledge; however, these maps remain limited by their static nature. Discrete dynamic modelling can be employed to study the emergent behaviours of these systems. Nevertheless, handling such large-scale models is challenging. Due to their massive size, it is computationally demanding to identify biologically relevant states in a cell- and disease-specific context. In this work, we developed an efficient computational framework that converts molecular interaction maps into Boolean models using the CaSQ tool. Next, we used a newly developed version of the BMA tool deployed to a high-performance computing cluster to identify the models' steady states. The identified attractors are then validated using gene expression data sets and prior knowledge. We successfully applied our framework to generate and calibrate the M1 and M2 macrophage Boolean models for rheumatoid arthritis. Using KO simulations, we identified NFkB, JAK1/JAK2, and ERK1/Notch1 as potential targets that could selectively suppress proinflammatory macrophages and GSK3B as a promising target that could promote anti-inflammatory macrophages in rheumatoid arthritis.
Collapse
Affiliation(s)
- Naouel Zerrouk
- GenHotel, Laboratoire Européen de Recherche Pour La Polyarthrite Rhumatoïde, University Paris-Saclay, University Evry, Evry, France
- Sanofi R&D Data and Data Science, Artificial Intelligence & Deep Analytics, Omics Data Science, 1, Av Pierre Brossolette, 91385, Chilly-Mazarin, France
| | - Rachel Alcraft
- Advanced Research Computing Centre, University College London, London, UK
| | - Benjamin A Hall
- Department of Medical Physics and Biomedical Engineering, University College London, London, UK
| | - Franck Augé
- Sanofi R&D Data and Data Science, Artificial Intelligence & Deep Analytics, Omics Data Science, 1, Av Pierre Brossolette, 91385, Chilly-Mazarin, France
| | - Anna Niarakis
- GenHotel, Laboratoire Européen de Recherche Pour La Polyarthrite Rhumatoïde, University Paris-Saclay, University Evry, Evry, France.
- Lifeware Group, Inria Saclay, Palaiseau, France.
| |
Collapse
|
11
|
Manchanda N, Vishkarma H, Goyal M, Shah S, Famta P, Talegaonkar S, Srivastava S. Surface Functionalized Lipid Nanoparticles in Promoting Therapeutic Outcomes: An Insight View of the Dynamic Drug Delivery System. Curr Drug Targets 2024; 25:278-300. [PMID: 38409709 DOI: 10.2174/0113894501285598240216065627] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Revised: 01/27/2024] [Accepted: 02/01/2024] [Indexed: 02/28/2024]
Abstract
Compared to the conventional approach, nanoparticles (NPs) facilitate a non-hazardous, non-toxic, non-interactive, and biocompatible system, rendering them incredibly promising for improving drug delivery to target cells. When that comes to accomplishing specific therapeutic agents like drugs, peptides, nucleotides, etc., lipidic nanoparticulate systems have emerged as even more robust. They have asserted impressive ability in bypassing physiological and cellular barriers, evading lysosomal capture and the proton sponge effect, optimizing bioavailability, and compliance, lowering doses, and boosting therapeutic efficacy. However, the lack of selectivity at the cellular level hinders its ability to accomplish its potential to the fullest. The inclusion of surface functionalization to the lipidic NPs might certainly assist them in adapting to the basic biological demands of a specific pathological condition. Several ligands, including peptides, enzymes, polymers, saccharides, antibodies, etc., can be functionalized onto the surface of lipidic NPs to achieve cellular selectivity and avoid bioactivity challenges. This review provides a comprehensive outline for functionalizing lipid-based NPs systems in prominence over target selectivity. Emphasis has been put upon the strategies for reinforcing the therapeutic performance of lipidic nano carriers' using a variety of ligands alongside instances of relevant commercial formulations.
Collapse
Affiliation(s)
- Namish Manchanda
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Delhi Pharmaceutical Sciences and Research University (DPSRU), Government of NCT of Delhi, Mehrauli-Badarpur Road, Pushp Vihar Sector-3, New Delhi-110017, Delhi (NCT), India
- Centre of Pharmaceutical Nanotechnology, Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), S.A.S Nagar, India
- Department of Pharmaceuticals, Ministry of Chemicals & Fertilizers, Government of India, Sector-67, S.A.S Nagar, Mohali-160062, Punjab, India
| | - Harish Vishkarma
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Delhi Pharmaceutical Sciences and Research University (DPSRU), Government of NCT of Delhi, Mehrauli-Badarpur Road, Pushp Vihar Sector-3, New Delhi-110017, Delhi (NCT), India
| | - Muskan Goyal
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Delhi Pharmaceutical Sciences and Research University (DPSRU), Government of NCT of Delhi, Mehrauli-Badarpur Road, Pushp Vihar Sector-3, New Delhi-110017, Delhi (NCT), India
| | - Saurabh Shah
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, India
- Department of Pharmaceuticals, Ministry of Chemicals & Fertilizers, Government of India, Balanagar, Hyderabad-500037, Telangana, India
| | - Paras Famta
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, India
- Department of Pharmaceuticals, Ministry of Chemicals & Fertilizers, Government of India, Balanagar, Hyderabad-500037, Telangana, India
| | - Sushama Talegaonkar
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Delhi Pharmaceutical Sciences and Research University (DPSRU), Government of NCT of Delhi, Mehrauli-Badarpur Road, Pushp Vihar Sector-3, New Delhi-110017, Delhi (NCT), India
| | - Saurabh Srivastava
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, India
- Department of Pharmaceuticals, Ministry of Chemicals & Fertilizers, Government of India, Balanagar, Hyderabad-500037, Telangana, India
| |
Collapse
|
12
|
Zhang W, Zhang Y, Zhang J, Deng C, Zhang C. Naringenin ameliorates collagen-induced arthritis through activating AMPK-mediated autophagy in macrophages. Immun Inflamm Dis 2023; 11:e983. [PMID: 37904715 PMCID: PMC10588338 DOI: 10.1002/iid3.983] [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: 01/31/2023] [Revised: 08/01/2023] [Accepted: 08/03/2023] [Indexed: 11/01/2023] Open
Abstract
BACKGROUND Naringenin is widely recognized for its notable attributes, including anti-inflammatory, anti-cancer, and immunomodulatory activities. However, its specific implications for rheumatoid arthritis (RA) and the underlying mechanisms remain to be explored. This study aimed to investigate the therapeutic efficacy and pharmacological mechanism of Naringenin in the treatment of collagen-induced arthritis (CIA). METHODS A CIA model was established in DBA/1 mice, and various doses of Naringenin were administered orally to assess its impact on RA. The study also involved lipopolysaccharides (LPS)-induced RAW264.7 cells to further evaluate the effects of Naringenin. Mechanistic studies were conducted to elucidate the signaling pathways involved in Naringenin's actions. RESULTS Naringenin significantly alleviated foot inflammation in DBA/1 CIA mice and attenuated the levels of pro-inflammatory cytokines in serum. It also enhanced antioxidant capacity in the CIA model. In vitro studies with LPS-induced RAW264.7 cells demonstrated that Naringenin attenuated pro-inflammatory cytokines and reactive oxygen species (ROS) levels. Mechanistic studies confirmed that Naringenin activated autophagy and increased autophagic flux. Blocking autophagy, either by silencing Atg5 or inhibiting autophagolysosome using chloroquine, effectively counteracted the impact of Naringenin on pro-inflammatory cytokines. Further exploration revealed that Naringenin activated the AMPK/ULK1 signaling pathway, and inhibition of AMPK reversed the initiation of autophagy and reduced pro-inflammatory cytokine secretion induced by Naringenin. CONCLUSIONS This study unveils a novel mechanism by which Naringenin may be used to treat RA. It demonstrates the therapeutic efficacy of Naringenin in a CIA model by reducing inflammation, modulating cytokine levels, and enhancing antioxidant capacity. Moreover, the activation of autophagy through the AMPK/ULK1 signaling pathway appears to play a critical role in Naringenin's anti-inflammatory effects. These findings suggest potential strategies for the development of anti-rheumatic medications based on Naringenin.
Collapse
Affiliation(s)
- Wei Zhang
- Department of OrthopedicAffiliated Hospital of North Sichuan Medical CollegeNanchongChina
| | - Yuan Zhang
- Department of OrthopedicAffiliated Hospital of North Sichuan Medical CollegeNanchongChina
| | - Jianguang Zhang
- Department of OrthopedicAffiliated Hospital of North Sichuan Medical CollegeNanchongChina
| | - Chunbiao Deng
- Department of OrthopedicAffiliated Hospital of North Sichuan Medical CollegeNanchongChina
| | - Chao Zhang
- Department of OrthopedicAffiliated Hospital of North Sichuan Medical CollegeNanchongChina
| |
Collapse
|
13
|
Mohapatra A, Mohanty A, Sathiyamoorthy P, Chahal S, Vijayan V, Rajendrakumar SK, Park IK. Targeted treatment of gouty arthritis by biomineralized metallic nanozyme-mediated oxidative stress-mitigating nanotherapy. J Mater Chem B 2023; 11:7684-7695. [PMID: 37464890 DOI: 10.1039/d3tb00669g] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/20/2023]
Abstract
Gouty arthritis is characterized by chronic deposition of monosodium urate (MSU) crystals in the joints and other tissues, resulting in the production of excess reactive oxygen species (ROS) and proinflammatory cytokines that intensify synovial inflammation. This condition is mainly associated with inflammatory M1 macrophage activation and oxidative stress production. Hence, gout symptoms can often be resolved by eliminating M1 macrophage activation and scavenging oxidative stress in the inflamed areas. Herein, we developed M1-macrophage-targeting biomineralized metallic nanozymes (FALNZs) that deplete oxidative stress and reduce the M1 macrophage levels to mitigate gouty arthritis. Intra-articular injection of the FALNZs targets inflammatory macrophages and suppresses ROS levels in joints with MSU-crystal-induced arthritis. In addition, the FALNZs alleviate joint swelling, inflammatory cytokine production, and pathological features of the joints. Overall, the proposed therapeutic approach is biocompatible and is an effective ROS scavenger for the treatment of gouty pathogenesis.
Collapse
Affiliation(s)
- Adityanarayan Mohapatra
- Department of Biomedical Science, BK21 PLUS Center for Creative Biomedical Scientists, Chonnam National University Medical School, Gwangju 61469, South Korea.
| | - Ayeskanta Mohanty
- Department of Biomedical Science, BK21 PLUS Center for Creative Biomedical Scientists, Chonnam National University Medical School, Gwangju 61469, South Korea.
| | - Padmanaban Sathiyamoorthy
- Department of Biomedical Science, BK21 PLUS Center for Creative Biomedical Scientists, Chonnam National University Medical School, Gwangju 61469, South Korea.
| | - Sahil Chahal
- Department of Biomedical Science, BK21 PLUS Center for Creative Biomedical Scientists, Chonnam National University Medical School, Gwangju 61469, South Korea.
| | - Veena Vijayan
- Department of Biomedical Science, BK21 PLUS Center for Creative Biomedical Scientists, Chonnam National University Medical School, Gwangju 61469, South Korea.
| | | | - In-Kyu Park
- Department of Biomedical Science, BK21 PLUS Center for Creative Biomedical Scientists, Chonnam National University Medical School, Gwangju 61469, South Korea.
| |
Collapse
|
14
|
Yang X, Qian H, Meng J, Jiang H, Yuan T, Yang S, Luo Y, Bao N, Zhao J, Wang D. Lonicerin alleviates the progression of experimental rheumatoid arthritis by downregulating M1 macrophages through the NF-κB signaling pathway. Phytother Res 2023; 37:3939-3950. [PMID: 37114508 DOI: 10.1002/ptr.7853] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2022] [Revised: 03/20/2023] [Accepted: 04/13/2023] [Indexed: 04/29/2023]
Abstract
The present study aimed to evaluate anti-rheumatoid arthritis (RA) effect of Lonicerin (LON), a safe compound with anti-inflammatory and immunomodulatory properties. Nevertheless, the exact role of LON in RA remains elusive. In this test, the anti-RA effect of LON was evaluated in collagen-induced arthritis (CIA) mouse model. Relevant parameters were measured during the experiment; ankle tissue and serum were collected at the end of the experiment for radiology, histopathology, and inflammation analysis. ELISA, qRT-PCR, immunofluorescence, and western blot were used to explore the effect of LON on the polarization of macrophages and related signal pathways. It was discovered that LON treatment attenuated the disease progression of CIA mice with lower paw swelling, clinical score, mobility, and inflammatory response. LON treatment significantly decreased M1 marker levels in CIA mice and LPS/IFN-γ-induced RAW264.7 cells, while slightly increasing M2 marker levels in CIA mice and IL-4-induced RAW264.7 cells. Mechanistically, LON attenuated the activation of the NF-κB signaling pathway, which contributes to M1 macrophage polarization and inflammasome activation. In addition, LON inhibited NLRP3 inflammasome activation in M1 macrophages, thereby reducing inflammation by inhibiting IL-1β and IL-18 release. These results indicated that LON might exert anti-RA effects by regulating the polarization of M1/M2 macrophage, especially by inhibiting macrophage polarization toward M1.
Collapse
Affiliation(s)
- Xiaojiang Yang
- Department of Orthopedics, Jinling Hospital, School of Medicine, Nanjing University, Nanjing, China
| | - Hong Qian
- Department of Orthopedics, Jinling Hospital, The First School of Clinical Medicine, Southern Medical University, Nanjing, China
| | - Jia Meng
- Department of Orthopedics, Jinling Hospital, School of Medicine, Nanjing University, Nanjing, China
| | - Hui Jiang
- Department of Orthopedics, Jinling Hospital, School of Medicine, Nanjing University, Nanjing, China
| | - Tao Yuan
- Department of Orthopedics, Jinling Hospital, School of Medicine, Nanjing University, Nanjing, China
| | - Shaoqiang Yang
- Department of Orthopedics, Jinling Hospital, School of Medicine, Nanjing University, Nanjing, China
| | - Yibin Luo
- Department of Orthopedics, Jinling Hospital, School of Medicine, Nanjing University, Nanjing, China
| | - Ninrong Bao
- Department of Orthopedics, Jinling Hospital, School of Medicine, Nanjing University, Nanjing, China
| | - Jianning Zhao
- Department of Orthopedics, Jinling Hospital, School of Medicine, Nanjing University, Nanjing, China
- Department of Orthopedics, Jinling Hospital, The First School of Clinical Medicine, Southern Medical University, Nanjing, China
| | - Dongsheng Wang
- Department of Orthopedics, Jinling Hospital, School of Medicine, Nanjing University, Nanjing, China
| |
Collapse
|
15
|
He S, Deng H, Li P, Hu J, Yang Y, Xu Z, Liu S, Guo W, Guo Q. Arthritic Microenvironment-Dictated Fate Decisions for Stem Cells in Cartilage Repair. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2207715. [PMID: 37518822 PMCID: PMC10520688 DOI: 10.1002/advs.202207715] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Revised: 06/05/2023] [Indexed: 08/01/2023]
Abstract
The microenvironment and stem cell fate guidance of post-traumatic articular cartilage regeneration is primarily the focus of cartilage tissue engineering. In articular cartilage, stem cells are characterized by overlapping lineages and uneven effectiveness. Within the first 12 weeks after trauma, the articular inflammatory microenvironment (AIME) plays a decisive role in determining the fate of stem cells and cartilage. The development of fibrocartilage and osteophyte hyperplasia is an adverse outcome of chronic inflammation, which results from an imbalance in the AIME during the cartilage tissue repair process. In this review, the sources for the different types of stem cells and their fate are summarized. The main pathophysiological events that occur within the AIME as well as their protagonists are also discussed. Additionally, regulatory strategies that may guide the fate of stem cells within the AIME are proposed. Finally, strategies that provide insight into AIME pathophysiology are discussed and the design of new materials that match the post-traumatic progress of AIME pathophysiology in a spatial and temporal manner is guided. Thus, by regulating an appropriately modified inflammatory microenvironment, efficient stem cell-mediated tissue repair may be achieved.
Collapse
Affiliation(s)
- Songlin He
- School of MedicineNankai UniversityTianjin300071China
- Institute of Orthopedicsthe First Medical CenterChinese PLA General HospitalBeijing Key Lab of Regenerative Medicine in OrthopedicsKey Laboratory of Musculoskeletal Trauma & War Injuries PLABeijing100853China
| | - Haotian Deng
- School of MedicineNankai UniversityTianjin300071China
- Institute of Orthopedicsthe First Medical CenterChinese PLA General HospitalBeijing Key Lab of Regenerative Medicine in OrthopedicsKey Laboratory of Musculoskeletal Trauma & War Injuries PLABeijing100853China
| | - Peiqi Li
- School of MedicineNankai UniversityTianjin300071China
- Institute of Orthopedicsthe First Medical CenterChinese PLA General HospitalBeijing Key Lab of Regenerative Medicine in OrthopedicsKey Laboratory of Musculoskeletal Trauma & War Injuries PLABeijing100853China
| | - Jingjing Hu
- Department of GastroenterologyInstitute of GeriatricsChinese PLA General HospitalBeijing100853China
| | - Yongkang Yang
- Institute of Orthopedicsthe First Medical CenterChinese PLA General HospitalBeijing Key Lab of Regenerative Medicine in OrthopedicsKey Laboratory of Musculoskeletal Trauma & War Injuries PLABeijing100853China
| | - Ziheng Xu
- Institute of Orthopedicsthe First Medical CenterChinese PLA General HospitalBeijing Key Lab of Regenerative Medicine in OrthopedicsKey Laboratory of Musculoskeletal Trauma & War Injuries PLABeijing100853China
| | - Shuyun Liu
- School of MedicineNankai UniversityTianjin300071China
- Institute of Orthopedicsthe First Medical CenterChinese PLA General HospitalBeijing Key Lab of Regenerative Medicine in OrthopedicsKey Laboratory of Musculoskeletal Trauma & War Injuries PLABeijing100853China
| | - Weimin Guo
- Department of Orthopaedic SurgeryGuangdong Provincial Key Laboratory of Orthopedics and TraumatologyFirst Affiliated HospitalSun Yat‐Sen UniversityGuangzhouGuangdong510080China
| | - Quanyi Guo
- School of MedicineNankai UniversityTianjin300071China
- Institute of Orthopedicsthe First Medical CenterChinese PLA General HospitalBeijing Key Lab of Regenerative Medicine in OrthopedicsKey Laboratory of Musculoskeletal Trauma & War Injuries PLABeijing100853China
| |
Collapse
|
16
|
Luo X, Xiong H, Jiang Y, Fan Y, Zuo C, Chen D, Chen L, Lin H, Gao J. Macrophage Reprogramming via Targeted ROS Scavenging and COX-2 Downregulation for Alleviating Inflammation. Bioconjug Chem 2023. [PMID: 37330989 DOI: 10.1021/acs.bioconjchem.3c00239] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/20/2023]
Abstract
Inflammation-related diseases affect large populations of people in the world and cause substantial healthcare burdens, which results in significant costs in time, material, and labor. Preventing or relieving uncontrolled inflammation is critical for the treatment of these diseases. Herein, we report a new strategy for alleviating inflammation by macrophage reprogramming via targeted reactive oxygen species (ROS) scavenging and cyclooxygenase-2 (COX-2) downregulation. As a proof of concept, we synthesize a multifunctional compound named MCI containing a mannose-based macrophage targeting moiety, an indomethacin (IMC)-based segment for inhibiting COX-2, and a caffeic acid (CAF)-based section for ROS clearance. As revealed by a series of in vitro experiments, MCI could significantly attenuate the expression of COX-2 and the level of ROS, leading to M1 to M2 macrophage reprogramming, as evidenced by the reduction and the elevation in the levels of pro-inflammatory M1 markers and anti-inflammatory M2 markers, respectively. Furthermore, in vivo experiments show MCI's promising therapeutic effects on rheumatoid arthritis (RA). Our work illustrates the success of targeted macrophage reprogramming for inflammation alleviation, which sheds light on the development of new anti-inflammatory drugs.
Collapse
Affiliation(s)
- Xiangjie Luo
- Fujian Provincial Key Laboratory of Chemical Biology, The MOE Key Laboratory of Spectrochemical Analysis & Instrumentation, and Department of Chemical Biology, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen 361005, China
| | - Hui Xiong
- Fujian Provincial Key Laboratory of Chemical Biology, The MOE Key Laboratory of Spectrochemical Analysis & Instrumentation, and Department of Chemical Biology, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen 361005, China
| | - Yuhang Jiang
- Fujian Provincial Key Laboratory of Chemical Biology, The MOE Key Laboratory of Spectrochemical Analysis & Instrumentation, and Department of Chemical Biology, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen 361005, China
| | - Yifan Fan
- Fujian Provincial Key Laboratory of Chemical Biology, The MOE Key Laboratory of Spectrochemical Analysis & Instrumentation, and Department of Chemical Biology, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen 361005, China
| | - Cuicui Zuo
- Fujian Provincial Key Laboratory of Chemical Biology, The MOE Key Laboratory of Spectrochemical Analysis & Instrumentation, and Department of Chemical Biology, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen 361005, China
| | - Dongxia Chen
- Fujian Provincial Key Laboratory of Chemical Biology, The MOE Key Laboratory of Spectrochemical Analysis & Instrumentation, and Department of Chemical Biology, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen 361005, China
| | - Limin Chen
- Fujian Provincial Key Laboratory of Chemical Biology, The MOE Key Laboratory of Spectrochemical Analysis & Instrumentation, and Department of Chemical Biology, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen 361005, China
| | - Hongyu Lin
- Fujian Provincial Key Laboratory of Chemical Biology, The MOE Key Laboratory of Spectrochemical Analysis & Instrumentation, and Department of Chemical Biology, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen 361005, China
| | - Jinhao Gao
- Fujian Provincial Key Laboratory of Chemical Biology, The MOE Key Laboratory of Spectrochemical Analysis & Instrumentation, and Department of Chemical Biology, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen 361005, China
| |
Collapse
|
17
|
Lu Y, Zhou J, Wang Q, Cai J, Yu B, Dai Q, Bao Y, Chen R, Zhang Z, Zhang D, Hou T. Glucocorticoid-loaded pH/ROS Dual-Responsive Nanoparticles Alleviate Joint Destruction by Downregulating the NF-κB Signaling Pathway. Acta Biomater 2023; 164:458-473. [PMID: 37072065 DOI: 10.1016/j.actbio.2023.04.012] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Revised: 04/06/2023] [Accepted: 04/07/2023] [Indexed: 04/20/2023]
Abstract
Rheumatoid arthritis (RA) is an autoimmune disease causing severe symptoms that are difficult to treat. Nano-drug delivery system is recognized as a promising strategy for management of RA. However, how to thoroughly release payloads from nanoformulations and synergistic therapy of RA needs to be further investigated. To address this issue, a pH and reactive oxygen species (ROS) dual-responsive, methylprednisolone (MPS)-loaded and arginine-glycine-aspartic acid (RGD)-modified nanoparticles (NPs) were fabricated using phytochemical and ROS-responsive moiety co-modified α-cyclodextrin (α-CD) as a carrier. In vitro and in vivo experiments verified that the pH/ROS dual-responsive nanomedicine could be efficiently internalized by activated macrophages and synovial cells, and the released MPS could promote transformation of M1-type macrophages into M2 phenotype, thereby down-regulating pro-inflammatory cytokines. In vivo experiments demonstrated that the pH/ROS dual-responsive nanomedicine was remarkably accumulated in the inflamed joints of mice with collagen-induced arthritis (CIA). The accumulated nanomedicine could obviously relieve joint swelling and cartilage destruction without obvious adverse effects. Importantly, the expression of interleukin-6 and tumor necrosis factor-α in the joints of CIA mice were significantly inhibited by the pH/ROS dual-responsive nanomedicine in comparison with free drug and non-targeted counterparts. In addition, the expression of the NF-κB signaling pathway molecules P65 was also significantly decreased by nanomedicine-treatment. Our results reveal that MPS-loaded pH/ROS dual-responsive NPs can effectively alleviate joint destruction via down-regulation of the NF-κB signaling pathway. STATEMENT OF SIGNIFICANCE: Nanomedicine is recognized as an attractive method for the targeting treatment of rheumatoid arthritis (RA). To thorough release of payloads from nanoformulations and synergistic therapy of RA, herein, a phytochemical and ROS-responsive moiety co-modified α-cyclodextrin was used as a pH/ROS dual-responsive carrier to encapsulate methylprednisolone to manage RA. The fabricated nanomedicine can effectively release its payloads under pH and/or ROS microenvironment, and the released drugs dramatically promote transformation of M1-type macrophages into M2 phenotype to reduce the release of pro-inflammatory cytokines. The prepared nanomedicine also obviously decreased the NF-κB signaling pathway molecule P65 expression in the joints, thereby down-regulating pro-inflammatory cytokines expression to alleviate joint swelling and cartilage destruction. We provided a candidate for the targeting treatment of RA.
Collapse
Affiliation(s)
- Yanzhu Lu
- Department of Orthopaedics, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing 400038, China; Department of Chemistry, College of Basic Medicine, Army Medical University (Third Military Medical University), Chongqing 400038, China; Department of Orthopaedics, 958th Hospital of Chinese People's Liberation Army (Third Military Medical University), Chongqing 400038, China
| | - Jiangling Zhou
- Department of Orthopaedics, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing 400038, China; National & Regional United Engineering Lab of Tissue Engineering, Department of Orthopaedics, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing 400038, China
| | - Qianmei Wang
- Department of Pharmacy, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing 400038, China
| | - Juan Cai
- Department of Orthopaedics, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing 400038, China; National & Regional United Engineering Lab of Tissue Engineering, Department of Orthopaedics, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing 400038, China
| | - Bo Yu
- Department of Orthopaedics, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing 400038, China; National & Regional United Engineering Lab of Tissue Engineering, Department of Orthopaedics, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing 400038, China
| | - Qijie Dai
- Department of Orthopaedics, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing 400038, China; National & Regional United Engineering Lab of Tissue Engineering, Department of Orthopaedics, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing 400038, China
| | - Ying Bao
- Department of Chemistry, College of Basic Medicine, Army Medical University (Third Military Medical University), Chongqing 400038, China
| | - Rui Chen
- Department of Chemistry, College of Basic Medicine, Army Medical University (Third Military Medical University), Chongqing 400038, China
| | - Zhongrong Zhang
- Department of Orthopaedics, 958th Hospital of Chinese People's Liberation Army (Third Military Medical University), Chongqing 400038, China.
| | - Dinglin Zhang
- Department of Chemistry, College of Basic Medicine, Army Medical University (Third Military Medical University), Chongqing 400038, China.
| | - Tianyong Hou
- Department of Orthopaedics, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing 400038, China; National & Regional United Engineering Lab of Tissue Engineering, Department of Orthopaedics, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing 400038, China.
| |
Collapse
|
18
|
Yang L, Sha Y, Wei Y, Fang H, Jiang J, Yin L, Zhong Z, Meng F. Mannose-mediated nanodelivery of methotrexate to macrophages augments rheumatoid arthritis therapy. Biomater Sci 2023; 11:2211-2220. [PMID: 36748266 DOI: 10.1039/d2bm02072f] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Rheumatoid arthritis (RA) is a chronic autoimmune disease that gravely jeopardizes the quality of life of numerous people. Methotrexate (MTX) is a disease-modifying anti-rheumatic drug commonly used in clinics; however, it suffers from slow onset, moderate efficacy, and adverse reactions such as renal dysfunction, myelosuppression, and bone erosion after long-term treatment. Here, we explored macrophage targeted delivery of MTX using mannose-installed chimaeric polymersomes (Man-PMTX) as an advanced treatment for RA. Man-PMTX exhibited high (∼18 wt%) and robust loading of MTX, uniform size of 51-55 nm, minimal hemolytic activity, and glutathione-actuated drug release property. Man-PMTX showed better uptake by activated macrophages than PMTX, and more repolarization of bone marrow-derived macrophages (BMDMs) to anti-inflammatory M2 type macrophages and less secretion of TNF-α and IL-1β compared with free MTX and PMTX. In vivo studies revealed that Man-PMTX showed significantly higher accumulation in inflammatory joints than in healthy joints and effectively treated RA by relieving inflammation, repolarizing macrophages from M1 type to M2 type, and mitigating proinflammatory cytokines. Accordingly, Man-PMTX effectively protected the synovium and bone from damage. Mannose-mediated nanodelivery of methotrexate to macrophages appears to be an attractive strategy to augment rheumatoid arthritis therapy.
Collapse
Affiliation(s)
- Liang Yang
- Biomedical Polymers Laboratory, College of Chemistry, Chemical Engineering and Materials Science, and State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou, 215123, P. R. China. .,College of Pharmaceutical Sciences, Soochow University, Suzhou, 215123, P. R. China
| | - Yongjie Sha
- Biomedical Polymers Laboratory, College of Chemistry, Chemical Engineering and Materials Science, and State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou, 215123, P. R. China. .,College of Pharmaceutical Sciences, Soochow University, Suzhou, 215123, P. R. China
| | - Yuansong Wei
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Collaborative Innovation Center of Suzhou Nano Science & Technology, Soochow University, Suzhou 215123, China.
| | - Hanghang Fang
- Biomedical Polymers Laboratory, College of Chemistry, Chemical Engineering and Materials Science, and State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou, 215123, P. R. China.
| | - Jingjing Jiang
- Biomedical Polymers Laboratory, College of Chemistry, Chemical Engineering and Materials Science, and State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou, 215123, P. R. China.
| | - Lichen Yin
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Collaborative Innovation Center of Suzhou Nano Science & Technology, Soochow University, Suzhou 215123, China.
| | - Zhiyuan Zhong
- Biomedical Polymers Laboratory, College of Chemistry, Chemical Engineering and Materials Science, and State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou, 215123, P. R. China. .,College of Pharmaceutical Sciences, Soochow University, Suzhou, 215123, P. R. China
| | - Fenghua Meng
- Biomedical Polymers Laboratory, College of Chemistry, Chemical Engineering and Materials Science, and State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou, 215123, P. R. China.
| |
Collapse
|
19
|
Baruah M, Kwon HY, Cho H, Chang YT, Samanta A. A Photoinduced Electron Transfer-Based Hypochlorite-Specific Fluorescent Probe for Selective Imaging of Proinflammatory M1 in a Rheumatoid Arthritis Model. Anal Chem 2023; 95:4147-4154. [PMID: 36800528 DOI: 10.1021/acs.analchem.2c05218] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/19/2023]
Abstract
The differentiation of the distinct phenotypes of macrophages is essential for monitoring the stage of inflammatory diseases for accurate diagnosis and treatment. Recent studies revealed that the level of hypochlorite (OCl-) varies from activated M1 macrophages (killing pathogens) to M2 (resolution of inflammation) during inflammation. Thus, we developed a simple and efficient fluorescent probe for discriminating M1 from M0 and M2. Herein, fluorescent-based imaging is applied as an alternative to immunohistochemistry, which is challenging due to the tedious process and high cost. We developed a hypochlorite-specific probe PMS-T to differentiate M1 and M2, employing a metabolism-oriented live-cell distinction. This probe enables the detection of inflammatory rheumatoid arthritis in an ex vivo mouse model. Thus, it can be a potential chemical tool for monitoring inflammatory diseases, including rheumatoid arthritis, that may overcome the existing barriers of immunohistochemistry.
Collapse
Affiliation(s)
- Mousumi Baruah
- Molecular Sensors and Therapeutics (MST) Research Laboratory, Department of Chemistry, School of Natural Sciences, Shiv Nadar University, Delhi NCR, NH 91, Tehsil Dadri, Greater Noida, Uttar Pradesh 201314, India
| | - Haw-Young Kwon
- Center for Self-Assembly and Complexity, Institute for Basic Science (IBS), Pohang 37673, Republic of Korea
| | - Heewon Cho
- School of Interdisciplinary Bioscience and Bioengineering, Pohang University of Science and Technology (POSTECH), Pohang 37673, Republic of Korea
| | - Young-Tae Chang
- Center for Self-Assembly and Complexity, Institute for Basic Science (IBS), Pohang 37673, Republic of Korea.,Department of Chemistry, Pohang University of Science and Technology (POSTECH), Pohang 37673, Republic of Korea
| | - Animesh Samanta
- Molecular Sensors and Therapeutics (MST) Research Laboratory, Department of Chemistry, School of Natural Sciences, Shiv Nadar University, Delhi NCR, NH 91, Tehsil Dadri, Greater Noida, Uttar Pradesh 201314, India
| |
Collapse
|
20
|
The Role of Macrophages in the Pathogenesis of Atherosclerosis. Cells 2023; 12:cells12040522. [PMID: 36831189 PMCID: PMC9954519 DOI: 10.3390/cells12040522] [Citation(s) in RCA: 36] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2022] [Revised: 01/27/2023] [Accepted: 02/02/2023] [Indexed: 02/08/2023] Open
Abstract
A wide variety of cell populations, including both immune and endothelial cells, participate in the pathogenesis of atherosclerosis. Among these groups, macrophages deserve special attention because different populations of them can have completely different effects on atherogenesis and inflammation in atherosclerosis. In the current review, the significance of different phenotypes of macrophages in the progression or regression of atherosclerosis will be considered, including their ability to become the foam cells and the consequences of this event, as well as their ability to create a pro-inflammatory or anti-inflammatory medium at the site of atherosclerotic lesions as a result of cytokine production. In addition, several therapeutic strategies directed to the modulation of macrophage activity, which can serve as useful ideas for future drug developments, will be considered.
Collapse
|
21
|
Li M, Cui H, Cao Y, Lin Y, Yang Y, Gao M, Zhang W, Wang C. Deep eutectic solvents-Hydrogels for the topical management of rheumatoid arthritis. J Control Release 2023; 354:664-679. [PMID: 36682725 DOI: 10.1016/j.jconrel.2023.01.050] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Revised: 01/09/2023] [Accepted: 01/17/2023] [Indexed: 01/24/2023]
Abstract
Deep eutectic solvents (DES) have demonstrated their ability to facilitate skin penetrability of rigid nanoparticles (NPs). Here, we reported a feasible and simple transdermal delivery strategy using mesoporous silica nanoparticles impregnated in DES hydrogels for topical management of rheumatoid arthritis (RA). To achieve this goal, nanoceria was immobilized within a silica nanoparticle matrix (MSN) and encapsulated with methotrexate (MTX). The functionalized nanoparticles were first engineered in an Arginine (Arg)-citric acid (CA) DES and then transferred to the carbomer hydrogel matrix. Due to the strong affinity of DES hydrogels to the skin, combined with solvent-driven "Drag" effects, the prepared DES-MSNs hydrogels produced dynamic mobility of MSNs through skin layers, resulting in high skin penetrability. After application to the skin, the hydrogel solvent drove the rigid NPs across the skin barrier in a nonintrusive manner, resulting in sustained penetration and accumulation of MSNs at subcutaneous inflammation sites. Subsequently, the MTX payload exerted a direct therapeutic effect, while nanoceria moderated the inflammatory microenvironment by initiating reactive oxygen species (ROS) scavenging and transformation of the macrophage phenotype. In this way, the synergistic action of the combination of immuno- and chemotherapy of the drug and its carrier on RA was achieved. Our work provides a novel strategy for multisite regulation and controlled management of RA in a noninvasive way.
Collapse
Affiliation(s)
- Mingjian Li
- School of Life Science and Technology, Kunming University of Science and Technology, Kunming 650500, PR China
| | - Hao Cui
- School of Life Science and Technology, Kunming University of Science and Technology, Kunming 650500, PR China
| | - Yubiao Cao
- School of Life Science and Technology, Kunming University of Science and Technology, Kunming 650500, PR China
| | - Yameng Lin
- School of Life Science and Technology, Kunming University of Science and Technology, Kunming 650500, PR China
| | - Ye Yang
- School of Life Science and Technology, Kunming University of Science and Technology, Kunming 650500, PR China
| | - Mingju Gao
- College of Notoginseng Medicine and Pharmacy, Wenshan University, Wenshan 663000, Yunnan, PR China
| | - Wen Zhang
- State Key Laboratory of Geological Processes and Mineral Resources, China University of Geosciences, Wuhan 430074, PR China.
| | - Chengxiao Wang
- School of Life Science and Technology, Kunming University of Science and Technology, Kunming 650500, PR China.
| |
Collapse
|
22
|
Wang S, Ji T, Wang L, Qu Y, Wang X, Wang W, Lv M, Wang Y, Li X, Jiang P. Exploration of the mechanism by which Huangqi Guizhi Wuwu decoction inhibits Lps-induced inflammation by regulating macrophage polarization based on network pharmacology. BMC Complement Med Ther 2023; 23:8. [PMID: 36624435 PMCID: PMC9830836 DOI: 10.1186/s12906-022-03826-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Accepted: 12/22/2022] [Indexed: 01/11/2023] Open
Abstract
BACKGROUND Huangqi Guizhi Wuwu decoction (HQGZWWD) is a traditional Chinese herbal medicine formulation with significant anti-inflammatory activity. However, its underlying mechanism remains unknown. Through network pharmacology and experimental validation, this study aimed to examine the potential mechanism of HQGZWWD in regulating macrophage polarization and inflammation. METHODS The active components were obtained from the Traditional Chinese Medicine Systems Pharmacology database and Analysis Platform (TCMSP), whereas the corresponding targets were obtained from the TCMSP and Swiss Target Prediction database. The GeneCards database identified targets associated with macrophage polarization and inflammation. Multiple networks were developed to identify the key compounds, principal biological processes, and pathways of HQGZWWD that regulate macrophage polarization and inflammation. Autodock Vina is utilized to assess the binding ability between targets and active compounds. Finally, confirm the experiment's central hypothesis. Human histiocytic lymphoma (U-937) cells were transformed into M1 macrophages following stimulation with Lipopolysaccharide (LPS) to evaluate the effect of HQGZWWD drug-containing mouse serum (HQGZWWD serum) on regulating macrophage polarization and inflammation. RESULTS A total of 54 active components and 859 HQGZWWD targets were obtained. There were 9972 targets associated with macrophage polarization and 11,109 targets associated with inflammation. After screening, 34 overlapping targets were identified, of which 5 were identified as central targets confirmed by experiments, including the α7 nicotinic acetylcholine receptor (α7 nAchR), interleukin 6 (IL-6), Interleukin-1 beta (IL-1β), interleukin 10 (IL-10) and growth factor beta (TGF-β1). Pathway enrichment analysis revealed that 34 overlapping targets were enriched in multiple pathways associated with macrophage polarization and inflammation, including the TGF beta signaling pathway, NF-kappa B signaling pathway, JAK-STAT signaling pathway, and TNF signaling pathway. Molecular docking confirmed that the majority of HQGZWWD's compounds can bind to the target. In vitro experiments, HQGZWWD serum was shown to up-regulate the expression of α7 nAchR, reduce the number of M1 macrophages, stimulate the production of M2 macrophages, inhibit the expression of pro-inflammatory cytokines IL-6 and IL1-β, and increase the expression of anti-inflammatory cytokines IL-10 and TGF-β1. CONCLUSION HQGZWWD can regulate the number of M1/M2 macrophages and the level of inflammatory cytokines, and the underlying mechanism may be related to the up-regulation of α7 nAchR expression.
Collapse
Affiliation(s)
- Sutong Wang
- grid.464402.00000 0000 9459 9325Shandong University of Traditional Chinese Medicine, Jinan, 250014 Shandong China
| | - Tianshu Ji
- grid.464402.00000 0000 9459 9325Shandong University of Traditional Chinese Medicine, Jinan, 250014 Shandong China
| | - Lin Wang
- grid.464402.00000 0000 9459 9325Shandong University of Traditional Chinese Medicine, Jinan, 250014 Shandong China
| | - Yiwei Qu
- grid.464402.00000 0000 9459 9325Shandong University of Traditional Chinese Medicine, Jinan, 250014 Shandong China
| | - Xinhui Wang
- grid.464402.00000 0000 9459 9325Shandong University of Traditional Chinese Medicine, Jinan, 250014 Shandong China
| | - Wenting Wang
- grid.464481.b0000 0004 4687 044XNational Clincial Research Center for Cardiovascular Diseases of Traditional Chinese Medicine, Xiyuan Hospital of China Academy of Chinese Medical Sciences, Beijing, 100091 China
| | - Mujie Lv
- grid.464402.00000 0000 9459 9325Shandong University of Traditional Chinese Medicine, Jinan, 250014 Shandong China
| | - Yongcheng Wang
- grid.479672.9Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, 250011 China
| | - Xiao Li
- grid.479672.9Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, 250011 China
| | - Ping Jiang
- grid.479672.9Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, 250011 China
| |
Collapse
|
23
|
Macrophage-Targeted Dextran Sulfate-Dexamethasone Conjugate Micelles for Effective Treatment of Rheumatoid Arthritis. MOLECULES (BASEL, SWITZERLAND) 2023; 28:molecules28020591. [PMID: 36677648 PMCID: PMC9863669 DOI: 10.3390/molecules28020591] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Revised: 12/28/2022] [Accepted: 12/29/2022] [Indexed: 01/11/2023]
Abstract
Rheumatoid arthritis (RA) is a chronic, systemic immune disease that causes joint affection and even disability. Activated macrophages play an important role in the pathogenesis and progression of RA by producing pro-inflammatory factors. The use of dexamethasone (DXM) is effective in relieving the intractable pain and inflammatory progression of RA. However, long-term use of DXM is strongly associated with increased rates of diabetes, osteoporosis, bone fractures, and mortality, which hinders its clinical use. In this study, the dextran sulfate-cisaconitic anhydride-dexamethasone (DXM@DS-cad-DXM) micelles were prepared to treat RA by selectively recognizing scavenger receptor (SR) on the activated macrophages. The potent targeting property of DXM@DS-cad-DXM micelles to SR was by fluorescence microscope. Additionally, the effective accumulation and powerful anti-inflammatory activity of DXM@DS-cad-DXM micelles were observed in the inflamed joints of adjuvant-induced arthritis (AIA) rats after intravenous administration. Overall, DXM@DS-cad-DXM micelles are a potentially effective nanomedicine for targeted therapy of RA.
Collapse
|
24
|
Gan PR, Wang RH, Deng R, Wu H, Bu YH, Chen FY, Dong XT, Ke JT. Geniposide inhibits SphK1 membrane targeting to restore macrophage polarization balance in collagen-induced arthritis mice. Eur J Pharmacol 2022; 933:175271. [PMID: 36108735 DOI: 10.1016/j.ejphar.2022.175271] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Revised: 08/23/2022] [Accepted: 09/08/2022] [Indexed: 11/03/2022]
Abstract
Imbalance of macrophage polarization plays a critical role in the progression of rheumatoid arthritis (RA). Geniposide (GE) has been shown to exert anti-inflammatory effects. However, the effect of GE on macrophage polarization remains unclear. Here, we investigated the regulation of GE on the imbalance of macrophage polarization in RA and how it functions. We established a mouse model of collagen-induced arthritis (CIA) and isolated bone marrow-derived macrophages (BMDMs). The results confirmed that pro-inflammatory M1 macrophages were dominant in CIA mice, but the polarization imbalance of macrophages was restored to a certain extent after GE treatment. Furthermore, the membrane targeting of sphingosine kinase 1 (SphK1) was increased in BMDMs of CIA mice, as manifested by increased membrane and cytoplasmic expression of p-SphK1 and high secretion level of sphingosine-1-phosphate (S1P). RAW264.7 cells were stimulated with lipopolysaccharide (LPS)-interferon (IFN)-γ or interleukin (IL)-4 to induce M1 or M2 phenotype, respectively, to revalidate the results obtained in BMDMs. The results again observed SphK1 membrane targeting in LPS-IFN-γ-stimulated RAW264.7 cells. Selective inhibition of SphK1 by PF543 or inhibition of the S1P receptors by FTY720 both restored the proportion of M1 and M2 macrophages in LPS-IFN-γ-stimulated RAW264.7 cells, confirming that SphK1 membrane targeting mediated a proportional imbalance in M1 and M2 macrophage polarization. In addition, GE inhibited SphK1 membrane targeting and kinase activity. Taken together, results confirmed that the inhibition of SphK1 membrane targeting by GE was responsible for restoring the polarization balance of macrophages in CIA mice.
Collapse
Affiliation(s)
- Pei-Rong Gan
- College of Pharmacy, Anhui University of Chinese Medicine, Qian Jiang Road 1, Hefei, 230012, China; Key Laboratory of Xin'an Medicine, Ministry of Education, Hefei, 230012, China; Anhui Province Key Laboratory of Research & Development of Chinese Medicine, Hefei, 230012, China
| | - Rong-Hui Wang
- College of Pharmacy, Anhui University of Chinese Medicine, Qian Jiang Road 1, Hefei, 230012, China; Key Laboratory of Xin'an Medicine, Ministry of Education, Hefei, 230012, China; Anhui Province Key Laboratory of Research & Development of Chinese Medicine, Hefei, 230012, China
| | - Ran Deng
- Key Laboratory of Xin'an Medicine, Ministry of Education, Hefei, 230012, China; Anhui Province Key Laboratory of Research & Development of Chinese Medicine, Hefei, 230012, China; School of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, 230012, China.
| | - Hong Wu
- College of Pharmacy, Anhui University of Chinese Medicine, Qian Jiang Road 1, Hefei, 230012, China; Key Laboratory of Xin'an Medicine, Ministry of Education, Hefei, 230012, China; Anhui Province Key Laboratory of Research & Development of Chinese Medicine, Hefei, 230012, China.
| | - Yan-Hong Bu
- College of Pharmacy, Anhui University of Chinese Medicine, Qian Jiang Road 1, Hefei, 230012, China; Key Laboratory of Xin'an Medicine, Ministry of Education, Hefei, 230012, China; Anhui Province Key Laboratory of Research & Development of Chinese Medicine, Hefei, 230012, China
| | - Fang-Yuan Chen
- College of Pharmacy, Anhui University of Chinese Medicine, Qian Jiang Road 1, Hefei, 230012, China; Key Laboratory of Xin'an Medicine, Ministry of Education, Hefei, 230012, China; Anhui Province Key Laboratory of Research & Development of Chinese Medicine, Hefei, 230012, China
| | - Xin-Tong Dong
- College of Pharmacy, Anhui University of Chinese Medicine, Qian Jiang Road 1, Hefei, 230012, China; Key Laboratory of Xin'an Medicine, Ministry of Education, Hefei, 230012, China; Anhui Province Key Laboratory of Research & Development of Chinese Medicine, Hefei, 230012, China
| | - Jiang-Tao Ke
- College of Pharmacy, Anhui University of Chinese Medicine, Qian Jiang Road 1, Hefei, 230012, China; Key Laboratory of Xin'an Medicine, Ministry of Education, Hefei, 230012, China; Anhui Province Key Laboratory of Research & Development of Chinese Medicine, Hefei, 230012, China
| |
Collapse
|
25
|
Xanthones from Securidaca inappendiculata Hassk. attenuate collagen-induced arthritis in rats by inhibiting the nicotinamide phosphoribosyltransferase/glycolysis pathway and macrophage polarization. Int Immunopharmacol 2022; 111:109137. [PMID: 36001918 DOI: 10.1016/j.intimp.2022.109137] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Revised: 08/02/2022] [Accepted: 08/05/2022] [Indexed: 11/24/2022]
Abstract
Securidaca inappendiculata (SI) Hassk. is a traditional medicine used to treat rheumatoid arthritis. Recent studies have reported that macrophages are the primary regulators of joint homeostasis and their polarization is closely related to their metabolic mode. Here, we aimed to investigate the relationship between the joint protective effect of SI's xanthone-rich fraction (XRF) on collagen-induced arthritis (CIA) in rats and the nicotinamide phosphoribosyltransferase (NAMPT)-glycolysis-polarization axis of macrophages. CIA model rats were treated with oral XRF and therapeutic efficacy was assessed based on arthritis score, degree of paw swelling, histological examination, and immunohistochemical analysis. Serum levels of cytokines, cellular metabolite concentrations, and protein and mRNA expression were determined by enzyme-linked immunosorbent assay (ELISA), western blotting (WB), and quantitative real-time PCR (RT-qPCR), respectively. The effects of dihydroxy-3,4-dimethoxyxanthone (XAN), a representative SI-derived compound, on RAW264.7 macrophages was analyzed in vitro using confocal laser scanning and flow cytometry. We found that XRF treatment significantly alleviated disease severity in CIA model rats. Levels of pro-inflammatory cytokines in the serum and M1 markers in synovium were reduced after XRF treatment, accompanied by an increase in the levels of anti-inflammatory cytokines and M2 markers. Further, XRF significantly suppressed synovial glycolysis by regulating NAMPT. In vitro studies further showed that XAN induced repolarization of lipopolysaccharide (LPS)-induced RAW264.7 macrophages with M1-M2 phenotype. Moreover, XAN negatively regulated glycolysis in the LPS-induced RAW264.7 macrophages in correlation with changes in NAMPT expression. Overall, the findings of this study suggest that the joint protective effects of XRF are achieved by inhibiting the NAMPT/glycolysis pathway and thereby regulating macrophage polarization.
Collapse
|
26
|
Kang Y, Zhou H, Jin W. Rothia nasimurium as a Cause of Disease: First Isolation from Farmed Geese. Vet Sci 2022; 9:vetsci9050197. [PMID: 35622725 PMCID: PMC9145032 DOI: 10.3390/vetsci9050197] [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: 03/18/2022] [Revised: 04/13/2022] [Accepted: 04/15/2022] [Indexed: 11/29/2022] Open
Abstract
Rothia nasimurium was known previously as an opportunistic pathogen of animals. However, there are few reports regarding the pathogenicity of Rothia nasimurium. In September 2020, geese contracted a disease of unknown cause which brought economic losses to a farm in Jiangsu Province, China, prompting a series of investigations. The bacterium was isolated, cultured, and purified, and then identified using Gram staining, biochemical tests, matrix-assisted laser desorption/ionization time of flight mass spectrometry, and 16S rRNA sequence analysis. After determining the obtained bacteria species, antibiotic susceptibility tests and animal regression experiments were carried out. A strain of bacterium was successfully isolated from the livers of the diseased geese, which was identified as a strain of the Gram-positive bacterium Rothia nasimurium according to the 16S rRNA sequencing results. By indexing references, no goose was reported to have been infected with Rothia nasimurium. The antibiotic susceptibility testing showed that only four antibiotics (amikacin, cefazolin, fosfomycin, and ampicillin/sulbactam) could effectively inhibit the growth of the Rothia nasimurium strain. The animal regression experiments showed that the novel isolated strain could infect goslings, and it also causes serious depilation of goslings. The results of the manuscript expanded the range of pathogenic microorganisms in geese, which is helpful to develop methods for avian endemic control.
Collapse
Affiliation(s)
- Yuhui Kang
- College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, China; (Y.K.); (H.Z.)
| | - Hongshan Zhou
- College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, China; (Y.K.); (H.Z.)
| | - Wenjie Jin
- College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, China; (Y.K.); (H.Z.)
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonosis, Yangzhou 225009, China
- Ministry of Education Key Laboratory of Poultry Preventive Medicine, Yangzhou 225009, China
- Correspondence:
| |
Collapse
|
27
|
Zhao C, Song W, Ma J, Wang N. Macrophage-derived hybrid exosome-mimic nanovesicles loaded with black phosphorus for multimodal rheumatoid arthritis therapy. Biomater Sci 2022; 10:6731-6739. [DOI: 10.1039/d2bm01274j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Multimodal anti-inflammatory activity for Rheumatoid arthritis (RA) management.
Collapse
Affiliation(s)
- Chengwu Zhao
- Department of Sports Medicine, The First Hospital of Jilin University, Changchun, China
| | - Wenxia Song
- Department of Pathology, The First Hospital of Jilin University, Changchun, China
| | - Jie Ma
- Department of Clinical Pharmacy, The First Hospital of Jilin University, Changchun, China
| | - Nan Wang
- Department of Hand and Foot Surgery, The First Hospital of Jilin University, Changchun, China
| |
Collapse
|