1
|
Zhang X, Fu X, Chen W, Chen P, Zhu H, Yang B, Liang J, Zeng F. Amelioration of the rheumatoid arthritis microenvironment using celastrol-loaded silver-modified ceria nanoparticles for enhanced treatment. J Nanobiotechnology 2025; 23:372. [PMID: 40405278 PMCID: PMC12100825 DOI: 10.1186/s12951-025-03388-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2024] [Accepted: 04/14/2025] [Indexed: 05/24/2025] Open
Abstract
Rheumatoid arthritis (RA) is characterized by elevated levels of reactive oxygen species (ROS) and a persistent inflammatory microenvironment dominated by M1 macrophages, both of which contribute to disease progression. To address these pathological features, we developed a core-shell nanoplatform consisting of silver-modified ceria nanoparticles loaded with celastrol (Ag-CeNP@Cel). This nanoplatform significantly enhances the water solubility of celastrol and reduces its hepato-renal toxicity by enabling passive accumulation in inflamed joints. The silver-modified ceria nanoparticles synergistically combine with celastrol to scavenge excess ROS and reprogram M1 macrophages into M2 macrophages, thereby mitigating inflammatory responses and improving the rheumatoid arthritis microenvironment (RAM). Ag-CeNP@Cel exhibited robust therapeutic efficacy and safety in preclinical models, presenting an innovative approach to RA treatment by integrating ROS elimination with macrophage modulation to ameliorate inflammatory microenvironment. This study underscores the potential of Ag-CeNP@Cel as a promising therapeutic strategy for RA management.
Collapse
Affiliation(s)
- Xiaoru Zhang
- Artemisinin Research Center, Guangzhou University of Chinese Medicine, Guangzhou, 510450, China
| | - Xiaguo Fu
- Artemisinin Research Center, Guangzhou University of Chinese Medicine, Guangzhou, 510450, China
| | - Wanying Chen
- Artemisinin Research Center, Guangzhou University of Chinese Medicine, Guangzhou, 510450, China
| | - Peng Chen
- The First Affiliated Hospital, The First Clinical Medical School, Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou, 510450, China
| | - He Zhu
- The First Affiliated Hospital, The First Clinical Medical School, Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou, 510450, China
| | - Bo Yang
- Zhujiang Hospital, Southern Medical University, No.253 Industrial Avenue, Haizhu District, Guangzhou, 510280, China.
| | - Jianming Liang
- Artemisinin Research Center, Guangzhou University of Chinese Medicine, Guangzhou, 510450, China.
| | - Feng Zeng
- Artemisinin Research Center, Guangzhou University of Chinese Medicine, Guangzhou, 510450, China.
| |
Collapse
|
2
|
Ihnatsyeu-Kachan A, Saichuk A, Sharko O, Zhogla V, Abashkin V, Le Goff W, Shmanai V, Shcharbin D, Guillas I, Kim S. Biomimetic high-density lipoprotein nanoparticles for the delivery of nucleic acid-based therapeutics. Biotechnol Adv 2025:108606. [PMID: 40398645 DOI: 10.1016/j.biotechadv.2025.108606] [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: 10/31/2024] [Revised: 04/10/2025] [Accepted: 05/14/2025] [Indexed: 05/23/2025]
Abstract
The field of delivering nucleic acids (NAs) via high-density lipoprotein-mimicking nanoparticles (HDL NPs) has shown promising advancements over the past two decades. HDL NPs are designed to efficiently bind NAs, safeguard them from degradation, and help navigate through various biological barriers to deliver them into the target cell's cytosol. Some HDL NPs allow direct cytosolic delivery of NAs by a selective mechanism with the involvement of HDL's natural receptor scavenger receptor class B type I (SR-B1). In contrast, others rely on endocytic uptake of the entire NA-loaded HDL NP. Owing to their highly biocompatible nature, ability to target clinically relevant receptors, and fine tunability, NA-loaded HDL NPs are applied to treat cancer, cardiovascular diseases, and brain malignancies. They are also emerging as potent vaccines against cancers and infectious diseases. This review focuses on various architectures of NA-loaded HDL NPs, their mechanisms for NA cellular uptake, and their therapeutic efficacy in vivo. It comprehensively covers the latest nanocarriers for NA delivery that contain HDL's apolipoproteins (ApoA-I, ApoE) or their mimetic peptides, which define the unique functional and targeting capabilities of HDL NPs.
Collapse
Affiliation(s)
- Aliaksei Ihnatsyeu-Kachan
- Chemical and Biological Integrative Research Center, Biomedical Research Division, Korea Institute of Science and Technology, 5, Hwarang-ro 14-gil, Seongbuk-gu, 02792 Seoul, Republic of Korea; Division of Bio-Medical Science & Technology, KIST School, University of Science and Technology, 5, Hwarang-ro 14-gil, Seongbuk-gu, 02792 Seoul, Republic of Korea; INSERM UMR_S1166, Institute of Cardiometabolism and Nutrition (ICAN), Sorbonne University, 91 boulevard de l'Hôpital, 75013 Paris, France.
| | - Anastasiia Saichuk
- Chemical and Biological Integrative Research Center, Biomedical Research Division, Korea Institute of Science and Technology, 5, Hwarang-ro 14-gil, Seongbuk-gu, 02792 Seoul, Republic of Korea; Division of Bio-Medical Science & Technology, KIST School, University of Science and Technology, 5, Hwarang-ro 14-gil, Seongbuk-gu, 02792 Seoul, Republic of Korea
| | - Olga Sharko
- Institute of Physical Organic Chemistry of the National Academy of Sciences of Belarus, Surganova 13, 220072 Minsk, Belarus
| | - Victoriya Zhogla
- Institute of Biophysics and Cell Engineering of the National Academy of Sciences of Belarus, Akademicheskaya 27, 220072 Minsk, Belarus
| | - Viktar Abashkin
- Institute of Biophysics and Cell Engineering of the National Academy of Sciences of Belarus, Akademicheskaya 27, 220072 Minsk, Belarus
| | - Wilfried Le Goff
- INSERM UMR_S1166, Institute of Cardiometabolism and Nutrition (ICAN), Sorbonne University, 91 boulevard de l'Hôpital, 75013 Paris, France.
| | - Vadim Shmanai
- Institute of Physical Organic Chemistry of the National Academy of Sciences of Belarus, Surganova 13, 220072 Minsk, Belarus
| | - Dzmitry Shcharbin
- Institute of Biophysics and Cell Engineering of the National Academy of Sciences of Belarus, Akademicheskaya 27, 220072 Minsk, Belarus
| | - Isabelle Guillas
- INSERM UMR_S1166, Institute of Cardiometabolism and Nutrition (ICAN), Sorbonne University, 91 boulevard de l'Hôpital, 75013 Paris, France.
| | - Sehoon Kim
- Chemical and Biological Integrative Research Center, Biomedical Research Division, Korea Institute of Science and Technology, 5, Hwarang-ro 14-gil, Seongbuk-gu, 02792 Seoul, Republic of Korea; Division of Bio-Medical Science & Technology, KIST School, University of Science and Technology, 5, Hwarang-ro 14-gil, Seongbuk-gu, 02792 Seoul, Republic of Korea; KU-KIST Graduate School of Converging Science and Technology, Korea University, Anam-ro 145, Seongbuk-gu, 02841 Seoul, Republic of Korea.
| |
Collapse
|
3
|
Zhang Z, Liu Y, Liang X, Wang Q, Xu M, Yang X, Tang J, He X, He Y, Zhang D, Li C. Advances in nanodelivery systems based on apoptosis strategies for enhanced rheumatoid arthritis therapy. Acta Biomater 2025; 197:87-103. [PMID: 40154765 DOI: 10.1016/j.actbio.2025.03.043] [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: 11/26/2024] [Revised: 03/11/2025] [Accepted: 03/25/2025] [Indexed: 04/01/2025]
Abstract
Rheumatoid arthritis (RA) is a chronic systemic autoimmune disorder primarily characterized by persistent synovial inflammation and progressive bone erosion. The pathogenesis of RA involves a complex cascade of cellular and molecular events, including sustained hyperactivation of macrophages, excessive recruitment and activation of neutrophils, pathological proliferation and invasion of fibroblast-like synoviocytes (FLS), and dysregulated differentiation and function of osteoclasts (OCs). The inflammatory factors secreted by these dysregulated cells significantly disrupt the joint microenvironment through multiple pathological mechanisms, primarily by promoting synovial inflammation, cartilage matrix degradation, osteoclast-mediated bone erosion, and pathological angiogenesis. Therapeutic strategies targeting the induction of apoptosis in these malignant cells have demonstrated considerable potential in preclinical studies, offering a promising approach to enhance treatment outcomes by simultaneously reducing inflammatory cytokine production and inhibiting pathogenic cell proliferation. However, conventional therapeutic drugs are limited in clinical applications because of their high toxicity and side effects. Inflammation induces morphological and functional changes in cells within the rheumatoid arthritis microenvironment (RAM), particularly the overexpression of specific receptors on cell membranes. This phenomenon has driven the development of ligand-modified targeted nanodelivery systems (NDSs), which can specifically target and induce apoptosis in specific cell types, thereby enhancing therapeutic efficacy. This paper comprehensively reviews the research progress of targeted NDSs based on apoptosis strategies for RA therapy, with a detailed discussion of their advantages in inducing apoptosis in various disease-associated cells. Furthermore, the potential of combining apoptosis of multiple cell types for RA treatment is explored. This review is expected to improve insights into the apoptosis of malignant cells to enhance RA therapy. STATEMENT OF SIGNIFICANCE: This review highlights recent advances in nanodelivery systems (NDSs) based on apoptotic strategies for enhanced rheumatoid arthritis (RA) therapy. Unlike conventional NDSs, these optimized systems specifically induce apoptosis in malignant cells within the RA microenvironment by integrating multiple therapeutic strategies. By summarizing the latest research, our work demonstrates the potential of these NDSs to suppress inflammatory responses and prevent bone destruction through targeted elimination of malignant cells, offering a novel direction for RA treatment. This review is significant as it provides a comprehensive overview for researchers and clinicians, facilitating the development of more effective therapeutic approaches for RA and other chronic inflammatory diseases.
Collapse
Affiliation(s)
- Zongquan Zhang
- Department of Pharmaceutical Sciences, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, 646000, China
| | - Yilin Liu
- Department of Pharmaceutical Sciences, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, 646000, China
| | - Xiaoya Liang
- Department of Pharmaceutical Sciences, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, 646000, China
| | - Qian Wang
- Classical teaching and Research Department, College of Integrated Chinese and Western medicine, Affiliated TCM Hospital of Southwest Medical University, Luzhou, 646000, China
| | - Maochang Xu
- Department of Pharmaceutical Sciences, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, 646000, China
| | - Xi Yang
- Department of Pharmaceutical Sciences, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, 646000, China
| | - Jun Tang
- Analysis and Testing Center, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, 646000, China
| | - Xinghui He
- Department of Pharmaceutical Sciences, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, 646000, China
| | - Yufeng He
- Department of Cardiology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, 646000, China
| | - Dan Zhang
- Department of Pharmaceutical Sciences, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, 646000, China; Green Pharmaceutical Technology Key Laboratory of Luzhou, School of Pharmacy, Southwest Medical University, Luzhou, 646000, China.
| | - Chunhong Li
- Department of Pharmaceutical Sciences, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, 646000, China.
| |
Collapse
|
4
|
Ihnatsyeu-Kachan A, Sharko O, Bekish A, Saichuk A, Zhogla V, Abashkin V, Ulashchik E, Shcharbin D, Le Goff W, Kontush A, Guillas I, Shmanai V, Kim S. High-density lipoprotein-like nanoparticles with cationic cholesterol derivatives for siRNA delivery. BIOMATERIALS ADVANCES 2025; 170:214202. [PMID: 39923604 DOI: 10.1016/j.bioadv.2025.214202] [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: 10/12/2024] [Revised: 01/15/2025] [Accepted: 01/24/2025] [Indexed: 02/11/2025]
Abstract
A new approach to siRNA delivery using high-density lipoprotein-like nanoparticles (HDL NPs) was investigated, incorporating oligoamine and cholesterol-derived cationic lipids (CLs) to associate siRNA with the carrier. Newly designed or commercially available compounds, including GL67 and 3-β-[N-(N',N'-dimethylaminoethane)-carbamoyl]cholesterol (DC-Cholesterol), were tested for siRNA binding, cytotoxicity, and siRNA cellular uptake. GL67 emerged as the most promising CL for siRNA delivery via HDL NPs. While it contributed to substantial siRNA uptake and cytosolic delivery in HepG2 cells, gene silencing remained limited, indicating a need for further optimization. Despite this, the study highlights the potential of positively charged cholesterol derivatives for siRNA delivery using HDL NPs. An analysis of the relationship between CL head group structure and HDL NPs' siRNA binding efficiency and cytotoxicity showed that factors such as oligoamine molecule conjugation site, linker type, amine group ethylation, and alkyl chain length between amine groups are crucial for optimizing CL design. Furthermore, the phospholipid environment surrounding CLs significantly influences HDL NPs' performance, particularly in siRNA cellular uptake. The study also revealed that intracellular siRNA trafficking varies by cell type, emphasizing the importance of customizing HDL NP formulations for specific cells. These insights are important for designing more effective HDL NPs for siRNA therapeutic delivery.
Collapse
Affiliation(s)
- Aliaksei Ihnatsyeu-Kachan
- Chemical and Biological Integrative Research Center, Biomedical Research Division, Korea Institute of Science and Technology, 5, Hwarang-ro 14-gil, Seongbuk-gu, Seoul, 02792, Republic of Korea; Division of Bio-Medical Science & Technology, KIST School, University of Science and Technology, 5, Hwarang-ro 14-gil, Seongbuk-gu, Seoul, 02841, Republic of Korea; Sorbonne Université, INSERM, Foundation for Innovation in Cardiometabolism and Nutrition (ICAN), UMR_S1166, 91 Boulevard de l'Hôpital, Paris, 75013, France
| | - Olga Sharko
- Institute of Physical Organic Chemistry of the National Academy of Sciences of Belarus, 13 Surganova Str., Minsk, 220072, Belarus
| | - Andrei Bekish
- Institute of Physical Organic Chemistry of the National Academy of Sciences of Belarus, 13 Surganova Str., Minsk, 220072, Belarus
| | - Anastasiia Saichuk
- Chemical and Biological Integrative Research Center, Biomedical Research Division, Korea Institute of Science and Technology, 5, Hwarang-ro 14-gil, Seongbuk-gu, Seoul, 02792, Republic of Korea; Division of Bio-Medical Science & Technology, KIST School, University of Science and Technology, 5, Hwarang-ro 14-gil, Seongbuk-gu, Seoul, 02841, Republic of Korea
| | - Victoriya Zhogla
- Institute of Biophysics and Cell Engineering of the National Academy of Sciences of Belarus, 27 Academicheskaya Str., Minsk, 220072, Belarus
| | - Viktar Abashkin
- Institute of Biophysics and Cell Engineering of the National Academy of Sciences of Belarus, 27 Academicheskaya Str., Minsk, 220072, Belarus
| | - Egor Ulashchik
- Institute of Physical Organic Chemistry of the National Academy of Sciences of Belarus, 13 Surganova Str., Minsk, 220072, Belarus
| | - Dzmitry Shcharbin
- Institute of Biophysics and Cell Engineering of the National Academy of Sciences of Belarus, 27 Academicheskaya Str., Minsk, 220072, Belarus
| | - Wilfried Le Goff
- Sorbonne Université, INSERM, Foundation for Innovation in Cardiometabolism and Nutrition (ICAN), UMR_S1166, 91 Boulevard de l'Hôpital, Paris, 75013, France
| | - Anatol Kontush
- Sorbonne Université, INSERM, Foundation for Innovation in Cardiometabolism and Nutrition (ICAN), UMR_S1166, 91 Boulevard de l'Hôpital, Paris, 75013, France
| | - Isabelle Guillas
- Sorbonne Université, INSERM, Foundation for Innovation in Cardiometabolism and Nutrition (ICAN), UMR_S1166, 91 Boulevard de l'Hôpital, Paris, 75013, France
| | - Vadim Shmanai
- Institute of Physical Organic Chemistry of the National Academy of Sciences of Belarus, 13 Surganova Str., Minsk, 220072, Belarus
| | - Sehoon Kim
- Chemical and Biological Integrative Research Center, Biomedical Research Division, Korea Institute of Science and Technology, 5, Hwarang-ro 14-gil, Seongbuk-gu, Seoul, 02792, Republic of Korea; Division of Bio-Medical Science & Technology, KIST School, University of Science and Technology, 5, Hwarang-ro 14-gil, Seongbuk-gu, Seoul, 02841, Republic of Korea; KU-KIST Graduate School of Converging Science and Technology, Korea University, 145 Anam-ro, Seongbuk-gu, Seoul, 02792, Republic of Korea.
| |
Collapse
|
5
|
Sawan S, Kumari A, Majie A, Ghosh A, Karmakar V, Kumari N, Ghosh S, Gorain B. siRNA-based nanotherapeutic approaches for targeted delivery in rheumatoid arthritis. BIOMATERIALS ADVANCES 2025; 168:214120. [PMID: 39577366 DOI: 10.1016/j.bioadv.2024.214120] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Revised: 11/15/2024] [Accepted: 11/15/2024] [Indexed: 11/24/2024]
Abstract
Rheumatoid arthritis (RA), characterized as a systemic autoimmune ailment, predominantly results in substantial joint and tissue damage, affecting millions of individuals globally. Modern treatment modalities are being explored as the traditional RA therapy with non-specific immunosuppressive drugs showcased potential side effects and variable responses. Research potential with small interfering RNA (siRNA) depicted potential in the treatment of RA. These siRNA-based therapies could include genes encoding pro-inflammatory cytokines like TNF-α, IL-1, and IL-6, as well as other molecular targets such as RANK, p38 MAPK, TGF-β, Wnt/Fz complex, and HIF. By downregulating the expression of these genes, siRNA-based nanoformulations can attenuate inflammation, inhibit immune system dysregulation, and prevent tissue damage associated with RA. Strategies of delivering siRNA molecules through nanocarriers could be targeted to treat RA effectively, where specific genes associated with this autoimmune disease pathology can be selectively silenced. Additionally, simultaneous targeting of multiple molecular pathways may offer synergistic therapeutic benefits, potentially leading to more effective and safer therapeutic strategies for RA patients. This review critically highlights the in-depth pathology of RA, RNA interference-mediated molecular targets, and nanocarrier-based siRNA delivery strategies, along with the challenges and opportunities to harbor future solutions.
Collapse
Affiliation(s)
- Sweta Sawan
- Department of Pharmaceutical Sciences and Technology, Birla Institute of Technology, Mesra, Ranchi, India
| | - Ankita Kumari
- Department of Pharmaceutical Sciences and Technology, Birla Institute of Technology, Mesra, Ranchi, India
| | - Ankit Majie
- Department of Pharmaceutical Sciences and Technology, Birla Institute of Technology, Mesra, Ranchi, India
| | - Arya Ghosh
- Department of Pharmaceutical Sciences and Technology, Birla Institute of Technology, Mesra, Ranchi, India
| | - Varnita Karmakar
- Department of Pharmaceutical Sciences and Technology, Birla Institute of Technology, Mesra, Ranchi, India
| | - Nimmy Kumari
- Department of Pharmaceutical Sciences and Technology, Birla Institute of Technology, Mesra, Ranchi, India
| | - Santanu Ghosh
- Department of Pharmaceutical Sciences and Technology, Birla Institute of Technology, Mesra, Ranchi, India.
| | - Bapi Gorain
- Department of Pharmaceutical Sciences and Technology, Birla Institute of Technology, Mesra, Ranchi, India.
| |
Collapse
|
6
|
Mirchandani AS, Sanchez-Garcia MA, Walmsley SR. How oxygenation shapes immune responses: emerging roles for physioxia and pathological hypoxia. Nat Rev Immunol 2025; 25:161-177. [PMID: 39349943 DOI: 10.1038/s41577-024-01087-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/23/2024] [Indexed: 03/04/2025]
Abstract
Most eukaryotes require oxygen for their survival and, with increasing multicellular complexity, oxygen availability and delivery rates vary across the tissues of complex organisms. In humans, healthy tissues have markedly different oxygen gradients, ranging from the hypoxic environment of the bone marrow (where our haematopoietic stem cells reside) to the lungs and their alveoli, which are among the most oxygenated areas of the body. Immune cells are therefore required to adapt to varying oxygen availability as they move from the bone marrow to peripheral organs to mediate their effector functions. These changing oxygen gradients are exaggerated during inflammation, where oxygenation is often depleted owing to alterations in tissue perfusion and increased cellular activity. As such, it is important to consider the effects of oxygenation on shaping the immune response during tissue homeostasis and disease conditions. In this Review, we address the relevance of both physiological oxygenation (physioxia) and disease-associated hypoxia (where cellular oxygen demand outstrips supply) for immune cell functions, discussing the relevance of hypoxia for immune responses in the settings of tissue homeostasis, inflammation, infection, cancer and disease immunotherapy.
Collapse
Affiliation(s)
- Ananda Shanti Mirchandani
- Centre for Inflammation Research, Institute for Regeneration and Repair, University of Edinburgh, Edinburgh, UK.
| | | | - Sarah Ruth Walmsley
- Centre for Inflammation Research, Institute for Regeneration and Repair, University of Edinburgh, Edinburgh, UK.
| |
Collapse
|
7
|
Feng C, Wang ZR, Li CY, Zhang XY, Wang XX. 3-MA attenuates collagen-induced arthritis in vivo via anti-inflammatory effect and autophagy inhibition. BMC Musculoskelet Disord 2025; 26:44. [PMID: 39806324 PMCID: PMC11727732 DOI: 10.1186/s12891-025-08274-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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/06/2024] [Accepted: 01/01/2025] [Indexed: 01/16/2025] Open
Abstract
BACKGROUND Rheumatoid arthritis (RA) is a chronic autoimmune disease which afflicts about nearly 1% of global population. RA results in synovitis and cartilage/bone damage, even disability which aggravates the health burden. Many drugs are used to relieve RA, such as glucocorticoids (GCs), non-steroidal anti-inflammatory drugs (NSAIDs), and disease-modifying anti-rheumatic drugs (DMARDs) in the clinical treatment. However, present clinical drugs have various disadvantages such as poor bioavailability and short biological half-life and drug resistance, or adverse effects. A recent study showed autophagy modulation may be a novel strategy in the treatment of RA. 3-Methylademine (3-MA), is the most widely used autophagy inhibitor, which blocks autophagy at the initiation and maturation stages. The aim of this study is to evaluate the effect of 3-MA in collagen-induced-arthritis (CIA) mice and further elucidate how 3-MA attenuated inflammation, and cartilage/bone damage in arthritis. METHODS An in-vivo mouse collagen-induced arthritis model was applied to compare differences in ankle destruction among control mice and CIA mice treated with or without 3-MA. Bone and cartilage destruction degree was evaluated by histology and micro-computed tomography (µCT). Further in-vivo assays utilized mouse serum samples to investigate inflammatory levels, oxidative levels, and bone resorption cytokines. At last, an immunofluorescence assay was applied to detect the autophagy level among the three groups. RESULTS The in-vivo mouse collagen-induced arthritis model showed that CIA mice revealed apparent hind paw and ankle swelling which was aggravated gradually along with time, while 3-MA treatment attenuated swelling gradually. µCT and histological results showed typical lesions in CIA group while 3-MA treatment alleviated arthritis-related destruction. Serum assay showed that 3-MA significantly reduced inflammatory cytokines levels, suppressed oxidative levels and bone resorption cytokines. Immunofluorescence assay revealed 3-MA significantly inhibited the abnormal autophagy level in CIA mouse ankle. CONCLUSIONS 3-MA protects bone destruction in CIA-induced mice arthritis by anti-inflammatory effect and autophagy inhibition.
Collapse
Affiliation(s)
- Chong Feng
- Tianjin Institute of Environmental and Operational Medicine, Tianjin, 300050, China
- School and Hospital of Stomatology, Tianjin Medical University, Tianjin, 300070, China
- Department of Orthodontics, Tianjin Stomatological Hospital, School of Medicine, Nankai University, Tianjin, 300041, China
- Tianjin Key Laboratory of Oral and Maxillofacial Function Reconstruction, Tianjin, 300041, China
| | - Zi-Rou Wang
- Tianjin Institute of Environmental and Operational Medicine, Tianjin, 300050, China
| | - Chen-Yu Li
- Tianjin Institute of Environmental and Operational Medicine, Tianjin, 300050, China
| | - Xiang-Yu Zhang
- School and Hospital of Stomatology, Tianjin Medical University, Tianjin, 300070, China.
| | - Xin-Xing Wang
- Tianjin Institute of Environmental and Operational Medicine, Tianjin, 300050, China.
| |
Collapse
|
8
|
Feng Y, Pan X, Li Z, Li Y, Sun Y, Yang S, He C, Dang Y, Huang L, Xiang B. Innovative Lipid Nanoparticles Co-Delivering Hydroxychloroquine and siRNA for Enhanced Rheumatoid Arthritis Therapy. Pharmaceutics 2025; 17:45. [PMID: 39861693 PMCID: PMC11769357 DOI: 10.3390/pharmaceutics17010045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2024] [Revised: 12/25/2024] [Accepted: 12/26/2024] [Indexed: 01/27/2025] Open
Abstract
Background: Rheumatoid arthritis (RA) is a debilitating autoimmune disorder characterized by chronic inflammation and joint damage. Despite advancements in treatment, complete remission remains elusive. Methods: In this study, we introduce a novel lipid nanoparticle formulation co-delivering hydroxychloroquine (HCQ) and siRNA targeting TNF-α (siTNF-α) using microfluidic technology, marking the first use of such a combination for RA therapy. Results: In LPS-stimulated RAW 264.7 cells, the nanoparticles effectively reduced inflammatory markers. When administered via an intra-articular injection in a rat model, they significantly decreased joint inflammation and demonstrated good biological safety. Conclusions: This pioneering approach highlights the potential of lipid nanoparticles as a dual-delivery platform for enhanced RA treatment through targeted intra-articular administration.
Collapse
Affiliation(s)
- Yanru Feng
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Hebei Medical University, Shijiazhuang 050017, China; (Y.F.); (X.P.); (Z.L.); (Y.L.); (Y.S.); (S.Y.); (C.H.); (Y.D.)
| | - Xintong Pan
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Hebei Medical University, Shijiazhuang 050017, China; (Y.F.); (X.P.); (Z.L.); (Y.L.); (Y.S.); (S.Y.); (C.H.); (Y.D.)
| | - Ziqian Li
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Hebei Medical University, Shijiazhuang 050017, China; (Y.F.); (X.P.); (Z.L.); (Y.L.); (Y.S.); (S.Y.); (C.H.); (Y.D.)
| | - Yue Li
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Hebei Medical University, Shijiazhuang 050017, China; (Y.F.); (X.P.); (Z.L.); (Y.L.); (Y.S.); (S.Y.); (C.H.); (Y.D.)
| | - Ya’nan Sun
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Hebei Medical University, Shijiazhuang 050017, China; (Y.F.); (X.P.); (Z.L.); (Y.L.); (Y.S.); (S.Y.); (C.H.); (Y.D.)
| | - Shaokun Yang
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Hebei Medical University, Shijiazhuang 050017, China; (Y.F.); (X.P.); (Z.L.); (Y.L.); (Y.S.); (S.Y.); (C.H.); (Y.D.)
| | - Chaoxing He
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Hebei Medical University, Shijiazhuang 050017, China; (Y.F.); (X.P.); (Z.L.); (Y.L.); (Y.S.); (S.Y.); (C.H.); (Y.D.)
| | - Yunjie Dang
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Hebei Medical University, Shijiazhuang 050017, China; (Y.F.); (X.P.); (Z.L.); (Y.L.); (Y.S.); (S.Y.); (C.H.); (Y.D.)
| | - Lu Huang
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Hebei Medical University, Shijiazhuang 050017, China; (Y.F.); (X.P.); (Z.L.); (Y.L.); (Y.S.); (S.Y.); (C.H.); (Y.D.)
| | - Bai Xiang
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Hebei Medical University, Shijiazhuang 050017, China; (Y.F.); (X.P.); (Z.L.); (Y.L.); (Y.S.); (S.Y.); (C.H.); (Y.D.)
- Hebei Key Laboratory of Innovative Drug Research and Evaluation, Hebei Medical University, Shijiazhuang 050017, China
| |
Collapse
|
9
|
Guo D, Liu H, Zhao S, Lu X, Wan H, Zhao Y, Liang X, Zhang A, Wu M, Xiao Z, Hu N, Li Z, Xie D. Synergistic rheumatoid arthritis therapy by interrupting the detrimental feedback loop to orchestrate hypoxia M1 macrophage polarization using an enzyme-catalyzed nanoplatform. Bioact Mater 2024; 41:221-238. [PMID: 39149592 PMCID: PMC11324459 DOI: 10.1016/j.bioactmat.2024.07.026] [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: 05/25/2024] [Revised: 07/14/2024] [Accepted: 07/17/2024] [Indexed: 08/17/2024] Open
Abstract
A detrimental feedback loop between hypoxia and oxidative stress consistently drives macrophage polarization toward a pro-inflammatory M1 phenotype, thus persistently aggravating rheumatoid arthritis (RA) progression. Herein, an enzyme-catalyzed nanoplatform with synergistic hypoxia-relieving and reactive oxygen species (ROS)-scavenging properties was developed using bovine serum albumin-bilirubin-platinum nanoparticles (BSA-BR-Pt NPs). Bilirubin was employed to eliminate ROS, while platinum exhibited a synergistic effect in scavenging ROS and simultaneously generated oxygen. In mice RA model, BSA-BR-Pt NPs treatment exhibited superior effects, resulting in significant improvements in joint inflammation, cartilage damage, and bone erosion, compared to methotrexate, the most widely used antirheumatic drug. Mechanistically, RNA-sequencing data and experimental results elucidated that BSA-BR-Pt NPs induced a re-polarization of hypoxic M1 macrophages to M2 macrophages via switching glycolysis to oxidative phosphorylation through the inhibition of HIF-1α pathway. Collectively, this research for the first time elaborated the underlying mechanism of enzyme-catalyzed nanoplatform in orchestrating macrophage polarization, and identified a novel therapeutic strategy for RA and other inflammatory disorders.
Collapse
Affiliation(s)
- Dong Guo
- Department of Orthopedic Surgery, Center for Orthopedic Surgery, The Third Affiliated Hospital of Southern Medical University, Guangzhou, 510630, PR China
- Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, Guangzhou, 510630, PR China
| | - Hui Liu
- Department of Orthopedic Surgery, Center for Orthopedic Surgery, The Third Affiliated Hospital of Southern Medical University, Guangzhou, 510630, PR China
- Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, Guangzhou, 510630, PR China
- Key Laboratory of Luminescence Analysis and Molecular Sensing (Southwest University), Ministry of Education, School of Materials and Energy, Southwest University, Chongqing, 400715, PR China
| | - Sheng Zhao
- Key Laboratory of Luminescence Analysis and Molecular Sensing (Southwest University), Ministry of Education, School of Materials and Energy, Southwest University, Chongqing, 400715, PR China
| | - Xinya Lu
- Department of Orthopedic Surgery, Center for Orthopedic Surgery, The Third Affiliated Hospital of Southern Medical University, Guangzhou, 510630, PR China
- Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, Guangzhou, 510630, PR China
| | - Haoyu Wan
- Department of Orthopedic Surgery, Center for Orthopedic Surgery, The Third Affiliated Hospital of Southern Medical University, Guangzhou, 510630, PR China
- Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, Guangzhou, 510630, PR China
| | - Yitao Zhao
- Department of Orthopedic Surgery, Center for Orthopedic Surgery, The Third Affiliated Hospital of Southern Medical University, Guangzhou, 510630, PR China
- Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, Guangzhou, 510630, PR China
| | - Xinzhi Liang
- Department of Orthopedic Surgery, Center for Orthopedic Surgery, The Third Affiliated Hospital of Southern Medical University, Guangzhou, 510630, PR China
- Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, Guangzhou, 510630, PR China
| | - Anbiao Zhang
- Department of Orthopedic Surgery, Center for Orthopedic Surgery, The Third Affiliated Hospital of Southern Medical University, Guangzhou, 510630, PR China
- Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, Guangzhou, 510630, PR China
| | - Mengyuan Wu
- Key Laboratory of Luminescence Analysis and Molecular Sensing (Southwest University), Ministry of Education, School of Materials and Energy, Southwest University, Chongqing, 400715, PR China
| | - Zhisheng Xiao
- Department of Orthopedic Surgery, Center for Orthopedic Surgery, The Third Affiliated Hospital of Southern Medical University, Guangzhou, 510630, PR China
- Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, Guangzhou, 510630, PR China
| | - Ning Hu
- Department of Orthopaedics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, PR China
| | - Zhong Li
- Department of Biomedical Engineering, The Chinese University of Hong Kong, NT, Hong Kong, PR China
| | - Denghui Xie
- Department of Orthopedic Surgery, Center for Orthopedic Surgery, The Third Affiliated Hospital of Southern Medical University, Guangzhou, 510630, PR China
- Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, Guangzhou, 510630, PR China
| |
Collapse
|
10
|
Cai L, Xiong PF, Li T, Li C, Wu ZX, Hong YL, Wang JT, Zhang MY, Yang XQ, Xu QQ, Shi H, Luo QC, Li R, Liu MM. Discovery of novel diaryl substituted isoquinolin-1(2H)-one derivatives as hypoxia-inducible factor-1 signaling inhibitors for the treatment of rheumatoid arthritis. Eur J Med Chem 2024; 271:116417. [PMID: 38688063 DOI: 10.1016/j.ejmech.2024.116417] [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: 01/22/2024] [Revised: 04/10/2024] [Accepted: 04/11/2024] [Indexed: 05/02/2024]
Abstract
Since synovial hypoxic microenvironment significantly promotes the pathological progress of rheumatoid arthritis (RA), hypoxia-inducible factor 1 (HIF-1) has been emerged as a promising target for the development of novel therapeutic agents for RA treatment. In this study, we designed and synthesized a series of diaryl substituted isoquinolin-1(2H)-one derivatives as HIF-1 signaling inhibitors using scaffold-hopping strategy. By modifying the substituents on N-atom and 6-position of isoquinolin-1-one, we discovered compound 17q with the most potent activities against HIF-1 (IC50 = 0.55 μM) in a hypoxia-reactive element (HRE) luciferase reporter assay. Further pharmacological studies revealed that 17q concentration-dependently blocked hypoxia-induced HIF-1α protein accumulation, reduced inflammation response, inhibited cellular invasiveness and promoted VHL-dependent HIF-1α degradation in human RA synovial cell line. Moreover, 17q improved the pathological injury of ankle joints, decreased angiogenesis and attenuated inflammation response in the adjuvant-induced arthritis (AIA) rat model, indicating the promising therapeutic potential of compound 17q as an effective HIF-1 inhibitor for RA therapy.
Collapse
Affiliation(s)
- Li Cai
- School of Basic Medicine, Anhui Medical University, Hefei, 230032, Anhui Province, PR China; Department of Pathology, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, Anhui Province, PR China
| | - Peng-Fei Xiong
- Anhui Province Key Laboratory of Inflammation and Immune Diseases, School of Pharmacy, Anhui Medical University, Hefei, 230032, Anhui Province, PR China
| | - Tao Li
- Anhui Province Key Laboratory of Inflammation and Immune Diseases, School of Pharmacy, Anhui Medical University, Hefei, 230032, Anhui Province, PR China
| | - Chong Li
- Anhui Province Key Laboratory of Inflammation and Immune Diseases, School of Pharmacy, Anhui Medical University, Hefei, 230032, Anhui Province, PR China
| | - Zheng-Xing Wu
- Anhui Province Key Laboratory of Inflammation and Immune Diseases, School of Pharmacy, Anhui Medical University, Hefei, 230032, Anhui Province, PR China
| | - Ya-Ling Hong
- Anhui Province Key Laboratory of Inflammation and Immune Diseases, School of Pharmacy, Anhui Medical University, Hefei, 230032, Anhui Province, PR China
| | - Jin-Ting Wang
- The First Clinical Medical College, Anhui Medical University, Hefei, 230032, Anhui Province, PR China
| | - Meng-Yue Zhang
- The Second Clinical Medical College, Anhui Medical University, Hefei, 230032, Anhui Province, PR China
| | - Xi-Qin Yang
- The Second Clinical Medical College, Anhui Medical University, Hefei, 230032, Anhui Province, PR China
| | - Qian-Qian Xu
- Anhui Province Key Laboratory of Inflammation and Immune Diseases, School of Pharmacy, Anhui Medical University, Hefei, 230032, Anhui Province, PR China
| | - Huan Shi
- Anhui Province Key Laboratory of Inflammation and Immune Diseases, School of Pharmacy, Anhui Medical University, Hefei, 230032, Anhui Province, PR China
| | - Qi-Chao Luo
- School of Basic Medicine, Anhui Medical University, Hefei, 230032, Anhui Province, PR China.
| | - Rong Li
- Anhui Province Key Laboratory of Inflammation and Immune Diseases, School of Pharmacy, Anhui Medical University, Hefei, 230032, Anhui Province, PR China; Institute of Health and Medicine, Hefei Comprehensive National Science Center, Hefei, 230026, Anhui Province, PR China.
| | - Ming-Ming Liu
- Anhui Province Key Laboratory of Inflammation and Immune Diseases, School of Pharmacy, Anhui Medical University, Hefei, 230032, Anhui Province, PR China.
| |
Collapse
|
11
|
Chernikov IV, Bachkova IK, Sen’kova AV, Meschaninova MI, Savin IA, Vlassov VV, Zenkova MA, Chernolovskaya EL. Cholesterol-Modified Anti-Il6 siRNA Reduces the Severity of Acute Lung Injury in Mice. Cells 2024; 13:767. [PMID: 38727303 PMCID: PMC11083178 DOI: 10.3390/cells13090767] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Revised: 04/27/2024] [Accepted: 04/29/2024] [Indexed: 05/13/2024] Open
Abstract
Small interfering RNA (siRNA) holds significant therapeutic potential by silencing target genes through RNA interference. Current clinical applications of siRNA have been primarily limited to liver diseases, while achievements in delivery methods are expanding their applications to various organs, including the lungs. Cholesterol-conjugated siRNA emerges as a promising delivery approach due to its low toxicity and high efficiency. This study focuses on developing a cholesterol-conjugated anti-Il6 siRNA and the evaluation of its potency for the potential treatment of inflammatory diseases using the example of acute lung injury (ALI). The biological activities of different Il6-targeted siRNAs containing chemical modifications were evaluated in J774 cells in vitro. The lead cholesterol-conjugated anti-Il6 siRNA after intranasal instillation demonstrated dose-dependent therapeutic effects in a mouse model of ALI induced by lipopolysaccharide (LPS). The treatment significantly reduced Il6 mRNA levels, inflammatory cell infiltration, and the severity of lung inflammation. IL6 silencing by cholesterol-conjugated siRNA proves to be a promising strategy for treating inflammatory diseases, with potential applications beyond the lungs.
Collapse
Affiliation(s)
- Ivan V. Chernikov
- Institute of Chemical Biology and Fundamental Medicine, Siberian Branch of the Russian Academy of Sciences, Acad. Lavrentiev Ave. 8, 630090 Novosibirsk, Russia; (I.V.C.); (I.K.B.); (A.V.S.); (M.I.M.); (I.A.S.); (M.A.Z.)
| | - Irina K. Bachkova
- Institute of Chemical Biology and Fundamental Medicine, Siberian Branch of the Russian Academy of Sciences, Acad. Lavrentiev Ave. 8, 630090 Novosibirsk, Russia; (I.V.C.); (I.K.B.); (A.V.S.); (M.I.M.); (I.A.S.); (M.A.Z.)
- Faculty of Natural Sciences, Novosibirsk State University, Pirogova Str., 1, 630090 Novosibirsk, Russia
| | - Aleksandra V. Sen’kova
- Institute of Chemical Biology and Fundamental Medicine, Siberian Branch of the Russian Academy of Sciences, Acad. Lavrentiev Ave. 8, 630090 Novosibirsk, Russia; (I.V.C.); (I.K.B.); (A.V.S.); (M.I.M.); (I.A.S.); (M.A.Z.)
| | - Mariya I. Meschaninova
- Institute of Chemical Biology and Fundamental Medicine, Siberian Branch of the Russian Academy of Sciences, Acad. Lavrentiev Ave. 8, 630090 Novosibirsk, Russia; (I.V.C.); (I.K.B.); (A.V.S.); (M.I.M.); (I.A.S.); (M.A.Z.)
| | - Innokenty A. Savin
- Institute of Chemical Biology and Fundamental Medicine, Siberian Branch of the Russian Academy of Sciences, Acad. Lavrentiev Ave. 8, 630090 Novosibirsk, Russia; (I.V.C.); (I.K.B.); (A.V.S.); (M.I.M.); (I.A.S.); (M.A.Z.)
| | - Valentin V. Vlassov
- Institute of Chemical Biology and Fundamental Medicine, Siberian Branch of the Russian Academy of Sciences, Acad. Lavrentiev Ave. 8, 630090 Novosibirsk, Russia; (I.V.C.); (I.K.B.); (A.V.S.); (M.I.M.); (I.A.S.); (M.A.Z.)
| | - Marina A. Zenkova
- Institute of Chemical Biology and Fundamental Medicine, Siberian Branch of the Russian Academy of Sciences, Acad. Lavrentiev Ave. 8, 630090 Novosibirsk, Russia; (I.V.C.); (I.K.B.); (A.V.S.); (M.I.M.); (I.A.S.); (M.A.Z.)
| | - Elena L. Chernolovskaya
- Institute of Chemical Biology and Fundamental Medicine, Siberian Branch of the Russian Academy of Sciences, Acad. Lavrentiev Ave. 8, 630090 Novosibirsk, Russia; (I.V.C.); (I.K.B.); (A.V.S.); (M.I.M.); (I.A.S.); (M.A.Z.)
| |
Collapse
|
12
|
Huang Y, Guo X, Wu Y, Chen X, Feng L, Xie N, Shen G. Nanotechnology's frontier in combatting infectious and inflammatory diseases: prevention and treatment. Signal Transduct Target Ther 2024; 9:34. [PMID: 38378653 PMCID: PMC10879169 DOI: 10.1038/s41392-024-01745-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Revised: 12/27/2023] [Accepted: 01/11/2024] [Indexed: 02/22/2024] Open
Abstract
Inflammation-associated diseases encompass a range of infectious diseases and non-infectious inflammatory diseases, which continuously pose one of the most serious threats to human health, attributed to factors such as the emergence of new pathogens, increasing drug resistance, changes in living environments and lifestyles, and the aging population. Despite rapid advancements in mechanistic research and drug development for these diseases, current treatments often have limited efficacy and notable side effects, necessitating the development of more effective and targeted anti-inflammatory therapies. In recent years, the rapid development of nanotechnology has provided crucial technological support for the prevention, treatment, and detection of inflammation-associated diseases. Various types of nanoparticles (NPs) play significant roles, serving as vaccine vehicles to enhance immunogenicity and as drug carriers to improve targeting and bioavailability. NPs can also directly combat pathogens and inflammation. In addition, nanotechnology has facilitated the development of biosensors for pathogen detection and imaging techniques for inflammatory diseases. This review categorizes and characterizes different types of NPs, summarizes their applications in the prevention, treatment, and detection of infectious and inflammatory diseases. It also discusses the challenges associated with clinical translation in this field and explores the latest developments and prospects. In conclusion, nanotechnology opens up new possibilities for the comprehensive management of infectious and inflammatory diseases.
Collapse
Affiliation(s)
- Yujing Huang
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, and West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China
| | - Xiaohan Guo
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, and West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China
| | - Yi Wu
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, and West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China
| | - Xingyu Chen
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, and West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China
| | - Lixiang Feng
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, and West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China
| | - Na Xie
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, and West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China.
| | - Guobo Shen
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, and West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China.
| |
Collapse
|
13
|
Fu X, Song Y, Feng X, Liu Z, Gao W, Song H, Zhang Q. Synergistic chemotherapy/PTT/oxygen enrichment by multifunctional liposomal polydopamine nanoparticles for rheumatoid arthritis treatment. Asian J Pharm Sci 2024; 19:100885. [PMID: 38434718 PMCID: PMC10906176 DOI: 10.1016/j.ajps.2024.100885] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Revised: 12/20/2023] [Accepted: 12/25/2023] [Indexed: 03/05/2024] Open
Abstract
Amultifunctional liposomal polydopamine nanoparticle (MPM@Lipo) was designed in this study, to combine chemotherapy, photothermal therapy (PTT) and oxygen enrichment to clear hyperproliferating inflammatory cells and improve the hypoxic microenvironment for rheumatoid arthritis (RA) treatment. MPM@Lipo significantly scavenged intracellular reactive oxygen species and relieved joint hypoxia, thus contributing to the repolarization of M1 macrophages into M2 phenotype. Furthermore, MPM@Lipo could accumulate at inflammatory joints, inhibit the production of inflammatory factors, and protect cartilage in vivo, effectively alleviating RA progression in a rat adjuvant-induced arthritis model. Moreover, upon laser irradiation, MPM@Lipo can elevate the temperature to not only significantly obliterate excessively proliferating inflammatory cells but also accelerate the production of methotrexate and oxygen, resulting in excellent RA treatment effects. Overall, the use of synergistic chemotherapy/PTT/oxygen enrichment therapy to treat RA is a powerful potential strategy.
Collapse
Affiliation(s)
- Xiaoling Fu
- School of Pharmacy, Fujian Medical University, Fuzhou 350108, China
- Fuzong Clinical Medical College of Fujian Medical University, Fuzhou 350025, China
| | - Yutong Song
- First school of clinical medicine, Nanjing Medical University, Nanjing 211166, China
| | - Xianquan Feng
- Fuzong Clinical Medical College of Fujian Medical University, Fuzhou 350025, China
| | - Zhihong Liu
- Fuzong Clinical Medical College of Fujian Medical University, Fuzhou 350025, China
| | - Wenhao Gao
- School of Pharmacy, Fujian Medical University, Fuzhou 350108, China
- Fuzong Clinical Medical College of Fujian Medical University, Fuzhou 350025, China
| | - Hongtao Song
- School of Pharmacy, Fujian Medical University, Fuzhou 350108, China
- Fuzong Clinical Medical College of Fujian Medical University, Fuzhou 350025, China
| | - Qian Zhang
- School of Pharmacy, Fujian Medical University, Fuzhou 350108, China
| |
Collapse
|
14
|
Zhang S, Zhang M, Zhang J, Li G, Lu X, Sun F, Liu W. Photoresponsive metal-organic framework with combined photodynamic therapy and hypoxia-activated chemotherapy for the targeted treatment of rheumatoid arthritis. Colloids Surf B Biointerfaces 2024; 234:113707. [PMID: 38181689 DOI: 10.1016/j.colsurfb.2023.113707] [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: 09/09/2023] [Revised: 11/20/2023] [Accepted: 12/12/2023] [Indexed: 01/07/2024]
Abstract
Activated M1-type macrophages, which produce inflammatory factors that exacerbate rheumatoid arthritis (RA), represent crucial target cells for inhibiting the disease process. In this study, we developed a novel photoresponsive targeted drug delivery system (TPNPs-HA) that can effectively deliver the hypoxia-activated prodrug tirapazamine (TPZ) specifically to activated macrophages. After administration, this metal-organic framework, PCN-224, constructed uing the photosensitizer porphyrin, exhibits the ability to generate excessive toxic reactive oxygen species (ROS) when exposed to near-infrared light. Additionally, the oxygen-consumed hypoxic environment further activates the chemotherapeutic effect of TPZ, thus creating a synergistic combination of photodynamic therapy (PDT) and hypoxia-activated chemotherapy (HaCT) to promote the elimination of activated M1-type macrophages. The results highlight the significantly potential of this photoresponsive nano-delivery system in providing substantial relief for RA. Furthermore, these findings support its effectiveness in inhibiting the disease process of RA, thereby offering new possibilities for the development of precise and accurate strategies for RA.
Collapse
Affiliation(s)
- Shixin Zhang
- Department of Anesthesiology, The Second Hospital of Jilin University, Changchun 130022, China; School of Life Sciences, Jilin University, Changchun 130012, China
| | - Miaomiao Zhang
- School of Life Sciences, Jilin University, Changchun 130012, China
| | - Jingbo Zhang
- School of Life Sciences, Jilin University, Changchun 130012, China
| | - Ge Li
- School of Life Sciences, Jilin University, Changchun 130012, China
| | - Xinyue Lu
- School of Life Sciences, Jilin University, Changchun 130012, China
| | - Fengying Sun
- School of Life Sciences, Jilin University, Changchun 130012, China
| | - Wenhua Liu
- Department of Anesthesiology, The Second Hospital of Jilin University, Changchun 130022, China.
| |
Collapse
|
15
|
Zhang B, Li J, Jiang J, Lin X, Sun X, Wang Q. Overcoming delivery barriers for RNA therapeutics in the treatment of rheumatoid arthritis. Eur J Pharm Biopharm 2023; 192:147-160. [PMID: 37844708 DOI: 10.1016/j.ejpb.2023.10.010] [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: 08/28/2023] [Revised: 10/06/2023] [Accepted: 10/13/2023] [Indexed: 10/18/2023]
Abstract
RNA therapeutics can manipulate gene expression or protein production, making them suitable for treating a wide range of diseases. Theoretically, any disease that has a definite biological target would probably find feasible therapeutic approach from RNA-based therapeutics. Numerous clinical trials using RNA therapeutics fighting against cancer, infectious diseases or inherited diseases have been reported and achieved desirable therapeutic efficacy. So far, encouraging findings from various animal experimental studies have also confirmed the great potential of RNA-based therapies in the treatment of rheumatic arthritis (RA). However, the in vivo multiple physiological barriers still seriously compromise the therapeutic efficacy of RNA drugs. Thus, safe and effective delivery strategies for RNA therapeutics are quite essential for their further and wide application in RA therapy. In this review, we will discuss the recent progress achieved using RNA-based therapeutics and focus on delivery strategies that can overcome the in vivo delivery barriers in RA treatment. Furthermore, discussion about the existing problems in current RNA delivery systems for RA therapy has been also included here.
Collapse
Affiliation(s)
- Bin Zhang
- Key Laboratory of Advanced Technologies of Materials, Ministry of Education and School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu 610031, China
| | - Jiao Li
- Key Laboratory of Advanced Technologies of Materials, Ministry of Education and School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu 610031, China
| | - Jiayu Jiang
- Patent Examination Cooperation Sichuan Center of the Patent office, Chengdu 610213, China
| | - Xin Lin
- Key Laboratory of Advanced Technologies of Materials, Ministry of Education and School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu 610031, China
| | - Xun Sun
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, Chengdu 610041, China
| | - Qin Wang
- Institute of Biomedical Engineering, College of Medicine, Southwest Jiaotong University, Chengdu 610031, China.
| |
Collapse
|
16
|
Rani R, Raina N, Sharma A, Kumar P, Tulli HS, Gupta M. Advancement in nanotechnology for treatment of rheumatoid arthritis: scope and potential applications. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2023; 396:2287-2310. [PMID: 37166463 DOI: 10.1007/s00210-023-02514-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Accepted: 04/25/2023] [Indexed: 05/12/2023]
Abstract
Rheumatoid arthritis is a hyperactive immune disorder that results in severe inflammation in synovial joints, cartilage, and bone deterioration, resulting in immobilization of joints. Traditional approaches for the treatment of rheumatoid arthritis are associated with some limiting factors such as suboptimal patient compliance, inability to control the progression of disorder, and safety concerns. Therefore, innovative drug delivery carriers for efficient therapeutic delivery at inflamed synovial sites with better safety assessment are urgently needed to address these issues. From this perspective, nanotechnology is an outstanding alternative to traditional drug delivery approaches, and it has shown great promise in developing novel carriers to treat rheumatoid arthritis. Considering the current research and future application of nanocarriers, it is believed that nanocarriers can be a crucial element in rheumatoid arthritis treatment. This paper covers all currently available pathophysiological aspects of rheumatoid arthritis and treatment options. Future research for the reduction of synovial inflammation should focus on developing multifunction nanoparticles capable of delivering therapeutic agents with improved safety, efficacy, and cost-effectiveness to be commercialized.
Collapse
Affiliation(s)
- Radha Rani
- Department of Pharmaceutics, Delhi Pharmaceutical Sciences and Research University, New Delhi, India
| | - Neha Raina
- Department of Pharmaceutics, Delhi Pharmaceutical Sciences and Research University, New Delhi, India
| | - Ajay Sharma
- Institute of Nuclear Medicine & Allied Sciences (INMAS-DRDO), Ministry of Defence, Brig. SK Mazumdar Marg, Lucknow Road, Timarpur, Delhi-110054, India
| | - Pramod Kumar
- Institute of Lung Health and Immunity, Helmholtz Zentrum München, 85764, Neuherberg, Germany
| | - Hardeep Singh Tulli
- Department of Biotechnology, Maharishi Markandeshwar Engineering College, Maharishi Markandeshwar (Deemed to Be University), Mullana-Ambala, 133207, India
| | - Madhu Gupta
- Department of Pharmaceutics, Delhi Pharmaceutical Sciences and Research University, New Delhi, India.
| |
Collapse
|
17
|
Yadav K, Sahu KK, Sucheta, Gnanakani SPE, Sure P, Vijayalakshmi R, Sundar VD, Sharma V, Antil R, Jha M, Minz S, Bagchi A, Pradhan M. Biomedical applications of nanomaterials in the advancement of nucleic acid therapy: Mechanistic challenges, delivery strategies, and therapeutic applications. Int J Biol Macromol 2023; 241:124582. [PMID: 37116843 DOI: 10.1016/j.ijbiomac.2023.124582] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Revised: 04/19/2023] [Accepted: 04/20/2023] [Indexed: 04/30/2023]
Abstract
In the past few decades, substantial advancement has been made in nucleic acid (NA)-based therapies. Promising treatments include mRNA, siRNA, miRNA, and anti-sense DNA for treating various clinical disorders by modifying the expression of DNA or RNA. However, their effectiveness is limited due to their concentrated negative charge, instability, large size, and host barriers, which make widespread application difficult. The effective delivery of these medicines requires safe vectors that are efficient & selective while having non-pathogenic qualities; thus, nanomaterials have become an attractive option with promising possibilities despite some potential setbacks. Nanomaterials possess ideal characteristics, allowing them to be tuned into functional bio-entity capable of targeted delivery. In this review, current breakthroughs in the non-viral strategy of delivering NAs are discussed with the goal of overcoming challenges that would otherwise be experienced by therapeutics. It offers insight into a wide variety of existing NA-based therapeutic modalities and techniques. In addition to this, it provides a rationale for the use of non-viral vectors and a variety of nanomaterials to accomplish efficient gene therapy. Further, it discusses the potential for biomedical application of nanomaterials-based gene therapy in various conditions, such as cancer therapy, tissue engineering, neurological disorders, and infections.
Collapse
Affiliation(s)
- Krishna Yadav
- Raipur Institute of Pharmaceutical Education and Research, Sarona, Raipur, Chhattisgarh 492010, India
| | - Kantrol Kumar Sahu
- Institute of Pharmaceutical Research, GLA University, Mathura, Uttar Pradesh 281406, India
| | - Sucheta
- School of Medical and Allied Sciences, K. R. Mangalam University, Gurugram, Haryana 122103, India
| | | | - Pavani Sure
- Department of Pharmaceutics, Vignan Institute of Pharmaceutical Sciences, Hyderabad, Telangana, India
| | - R Vijayalakshmi
- Department of Pharmaceutical Analysis, GIET School of Pharmacy, Chaitanya Knowledge City, Rajahmundry, AP 533296, India
| | - V D Sundar
- Department of Pharmaceutical Technology, GIET School of Pharmacy, Chaitanya Knowledge City, Rajahmundry, AP 533296, India
| | - Versha Sharma
- Department of Biotechnology, School of Biological Sciences, Dr. Harisingh Gour Central University, Sagar, M.P. 470003, India
| | - Ruchita Antil
- Addenbrookes Hospital, Cambridge University Hospitals NHS Foundation Trust, England, United Kingdom of Great Britain and Northern Ireland
| | - Megha Jha
- Department of Biotechnology, School of Biological Sciences, Dr. Harisingh Gour Central University, Sagar, M.P. 470003, India
| | - Sunita Minz
- Department of Pharmacy, Indira Gandhi National Tribal University, Amarkantak, M.P., 484887, India
| | - Anindya Bagchi
- Tumor Initiation & Maintenance Program, Sanford Burnham Prebys Medical Discovery Institute, 10901 North Torrey Pines Road La Jolla, CA 92037, USA
| | | |
Collapse
|
18
|
Liu Y, Chen L, Chen Z, Liu M, Li X, Kou Y, Hou M, Wang H, Li X, Tian B, Dong J. Multifunctional Janus Nanoplatform for Efficiently Synergistic Theranostics of Rheumatoid Arthritis. ACS NANO 2023; 17:8167-8182. [PMID: 37083341 DOI: 10.1021/acsnano.2c11777] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/03/2023]
Abstract
Progress has been made in the application of nanomedicine in rheumatoid arthritis (RA) treatment. However, the whole process of monitoring and treatment of RA remains a formidable challenge due to the complexity of the chronic autoimmune disease. In this study, we develop a Janus nanoplatform (denoted as Janus-CPS) composed of CeO2-Pt nanozyme subunit on one side and periodic mesoporous organosilica (PMO) subunit on another side for simultaneous early diagnosis and synergistic therapy of RA. The Janus nanostructure, which enables more active sites to be exposed, enhances the reactive oxygen species scavenging capability of CeO2-Pt nanozyme subunit as compared to their core-shell counterpart. Furthermore, micheliolide (MCL), an extracted compound from natural plants with anti-osteoclastogenesis effects, is loaded into the mesopores of PMO subunit to synergize with the anti-inflammation effect of nanozymes for efficient RA treatment, which has been demonstrated by in vitro cellular experiments and in vivo collagen-induced arthritis (CIA) model. In addition, by taking advantage of the second near-infrared window (NIR-II) fluorescent imaging, indocyanine green (ICG)-loaded Janus-CPS exhibits desirable effectiveness in detecting RA lesions at a very early stage. It is anticipated that such a Janus nanoplatform may offer an alternative strategy of functional integration for versatile theranostics.
Collapse
Affiliation(s)
- Yuyi Liu
- Department of Orthopaedic Surgery, Zhongshan Hospital, Fudan University, Shanghai 200032, P. R. China
| | - Liang Chen
- Materdicine Lab, School of Life Sciences, Shanghai University, Shanghai 200444, P. R. China
| | - Zhiyang Chen
- Department of Orthopaedic Surgery, Zhongshan Hospital, Fudan University, Shanghai 200032, P. R. China
| | - Minchao Liu
- Department of Chemistry, State Key Laboratory of Molecular Engineering of Polymers, iChem, Shanghai Key Laboratory of Molecular Catalysis and Innovative Materials, Fudan University, Shanghai 200433, P. R. China
| | - Xilei Li
- Department of Orthopaedic Surgery, Zhongshan Hospital, Fudan University, Shanghai 200032, P. R. China
| | - Yufang Kou
- Department of Chemistry, State Key Laboratory of Molecular Engineering of Polymers, iChem, Shanghai Key Laboratory of Molecular Catalysis and Innovative Materials, Fudan University, Shanghai 200433, P. R. China
| | - MengMeng Hou
- Department of Chemistry, State Key Laboratory of Molecular Engineering of Polymers, iChem, Shanghai Key Laboratory of Molecular Catalysis and Innovative Materials, Fudan University, Shanghai 200433, P. R. China
| | - Huiren Wang
- Department of Orthopaedic Surgery, Zhongshan Hospital, Fudan University, Shanghai 200032, P. R. China
| | - Xiaomin Li
- Department of Chemistry, State Key Laboratory of Molecular Engineering of Polymers, iChem, Shanghai Key Laboratory of Molecular Catalysis and Innovative Materials, Fudan University, Shanghai 200433, P. R. China
| | - Bo Tian
- Department of Orthopaedic Surgery, Zhongshan Hospital, Fudan University, Shanghai 200032, P. R. China
| | - Jian Dong
- Department of Orthopaedic Surgery, Zhongshan Hospital, Fudan University, Shanghai 200032, P. R. China
- Department of Orthopaedic Surgery, Shanghai Baoshan District Wusong Center Hospital, Zhongshan Hospital Wusong Branch, Fudan University, Shanghai 200940, P. R. China
| |
Collapse
|
19
|
Zhang X, Liu Y, Xiao C, Guan Y, Gao Z, Huang W. Research Advances in Nucleic Acid Delivery System for Rheumatoid Arthritis Therapy. Pharmaceutics 2023; 15:1237. [PMID: 37111722 PMCID: PMC10145518 DOI: 10.3390/pharmaceutics15041237] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Revised: 03/31/2023] [Accepted: 04/05/2023] [Indexed: 04/29/2023] Open
Abstract
Rheumatoid arthritis (RA) is a chronic inflammatory autoimmune disease that affects the lives of nearly 1% of the total population worldwide. With the understanding of RA, more and more therapeutic drugs have been developed. However, lots of them possess severe side effects, and gene therapy may be a potential method for RA treatment. A nanoparticle delivery system is vital for gene therapy, as it can keep the nucleic acids stable and enhance the efficiency of transfection in vivo. With the development of materials science, pharmaceutics and pathology, more novel nanomaterials and intelligent strategies are applied to better and safer gene therapy for RA. In this review, we first summarized the existing nanomaterials and active targeting ligands used for RA gene therapy. Then, we introduced various gene delivery systems for RA treatment, which may enlighten the relevant research in the future.
Collapse
Affiliation(s)
- Xintong 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; (X.Z.); (Y.L.); (C.X.)
- Beijing Key Laboratory of Drug Delivery Technology and Novel Formulations, Department of Pharmaceutics, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Yanhong Liu
- 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; (X.Z.); (Y.L.); (C.X.)
- Beijing Key Laboratory of Drug Delivery Technology and Novel Formulations, Department of Pharmaceutics, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Congcong Xiao
- 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; (X.Z.); (Y.L.); (C.X.)
- Beijing Key Laboratory of Drug Delivery Technology and Novel Formulations, Department of Pharmaceutics, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Youyan Guan
- Department of Urology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China;
| | - Zhonggao Gao
- 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; (X.Z.); (Y.L.); (C.X.)
- Beijing Key Laboratory of Drug Delivery Technology and Novel Formulations, Department of Pharmaceutics, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Wei Huang
- 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; (X.Z.); (Y.L.); (C.X.)
- Beijing Key Laboratory of Drug Delivery Technology and Novel Formulations, Department of Pharmaceutics, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| |
Collapse
|
20
|
Yang N, Li M, Wu L, Song Y, Yu S, Wan Y, Cheng W, Yang B, Mou X, Yu H, Zheng J, Li X, Yu X. Peptide-anchored neutrophil membrane-coated biomimetic nanodrug for targeted treatment of rheumatoid arthritis. J Nanobiotechnology 2023; 21:13. [PMID: 36639772 PMCID: PMC9837964 DOI: 10.1186/s12951-023-01773-x] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Accepted: 01/08/2023] [Indexed: 01/15/2023] Open
Abstract
Macrophage polarization determines the production of cytokines that fuel the initiation and evolution of rheumatoid arthritis (RA). Thus, modulation of macrophage polarization might represent a potential therapeutic strategy for RA. However, coordinated modulation of macrophages in the synovium and synovial fluid has not been achieved thus far. Herein, we develop a biomimetic ApoA-I mimetic peptide-modified neutrophil membrane-wrapped F127 polymer (R4F-NM@F127) for targeted drug delivery during RA treatment. Due to the high expression of adhesion molecules and chemokine receptors on neutrophils, the neutrophil membrane coating can endow the nanocarrier with synovitis-targeting ability, with subsequent recruitment to the synovial fluid under the chemotactic effects of IL-8. Moreover, R4F peptide modification further endows the nanocarrier with the ability to target the SR-B1 receptor, which is highly expressed on macrophages in the synovium and synovial fluid. Long-term in vivo imaging shows that R4F-NM@F127 preferentially accumulates in inflamed joints and is engulfed by macrophages. After loading of the anti-inflammatory drug celastrol (Cel), R4F-NM@F127-Cel shows a significant reduction in hepatotoxicity, and effectively inhibits synovial inflammation and alleviates joint damage by reprogramming macrophage polarization. Thus, our results highlight the potential of the coordinated targeted modulation of macrophages as a promising therapeutic option for the treatment of RA.
Collapse
Affiliation(s)
- Ni Yang
- grid.254148.e0000 0001 0033 6389Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy, China Three Gorges University, Yichang, 443002 China ,grid.254148.e0000 0001 0033 6389Institute of Infection and Inflammation, China Three Gorges University, Yichang, 443002 China ,grid.254148.e0000 0001 0033 6389College of Basic Medical Science, China Three Gorges University, Yichang, 443002 China
| | - Miaomiao Li
- grid.254148.e0000 0001 0033 6389Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy, China Three Gorges University, Yichang, 443002 China ,grid.254148.e0000 0001 0033 6389Institute of Infection and Inflammation, China Three Gorges University, Yichang, 443002 China ,grid.254148.e0000 0001 0033 6389College of Basic Medical Science, China Three Gorges University, Yichang, 443002 China
| | - Ling Wu
- grid.254148.e0000 0001 0033 6389The People’s Hospital of China Three Gorges University, Yichang, 443099 China
| | - Yinhong Song
- grid.254148.e0000 0001 0033 6389Institute of Infection and Inflammation, China Three Gorges University, Yichang, 443002 China ,grid.254148.e0000 0001 0033 6389College of Basic Medical Science, China Three Gorges University, Yichang, 443002 China
| | - Shi Yu
- grid.254148.e0000 0001 0033 6389College of Basic Medical Science, China Three Gorges University, Yichang, 443002 China
| | - Yingying Wan
- grid.254148.e0000 0001 0033 6389Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy, China Three Gorges University, Yichang, 443002 China ,grid.254148.e0000 0001 0033 6389Institute of Infection and Inflammation, China Three Gorges University, Yichang, 443002 China ,grid.254148.e0000 0001 0033 6389College of Basic Medical Science, China Three Gorges University, Yichang, 443002 China
| | - Wenjing Cheng
- grid.254148.e0000 0001 0033 6389Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy, China Three Gorges University, Yichang, 443002 China ,grid.254148.e0000 0001 0033 6389Institute of Infection and Inflammation, China Three Gorges University, Yichang, 443002 China ,grid.254148.e0000 0001 0033 6389College of Basic Medical Science, China Three Gorges University, Yichang, 443002 China
| | - Baoye Yang
- grid.254148.e0000 0001 0033 6389Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy, China Three Gorges University, Yichang, 443002 China ,grid.254148.e0000 0001 0033 6389Institute of Infection and Inflammation, China Three Gorges University, Yichang, 443002 China ,grid.254148.e0000 0001 0033 6389College of Basic Medical Science, China Three Gorges University, Yichang, 443002 China
| | - Xiaoqin Mou
- grid.254148.e0000 0001 0033 6389Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy, China Three Gorges University, Yichang, 443002 China ,grid.254148.e0000 0001 0033 6389Institute of Infection and Inflammation, China Three Gorges University, Yichang, 443002 China ,grid.254148.e0000 0001 0033 6389College of Basic Medical Science, China Three Gorges University, Yichang, 443002 China
| | - Hong Yu
- grid.254148.e0000 0001 0033 6389Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy, China Three Gorges University, Yichang, 443002 China ,grid.254148.e0000 0001 0033 6389Institute of Infection and Inflammation, China Three Gorges University, Yichang, 443002 China ,grid.254148.e0000 0001 0033 6389College of Basic Medical Science, China Three Gorges University, Yichang, 443002 China
| | - Jing Zheng
- grid.254148.e0000 0001 0033 6389The People’s Hospital of China Three Gorges University, Yichang, 443099 China
| | - Xinzhi Li
- grid.254148.e0000 0001 0033 6389College of Basic Medical Science, China Three Gorges University, Yichang, 443002 China
| | - Xiang Yu
- grid.254148.e0000 0001 0033 6389Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy, China Three Gorges University, Yichang, 443002 China ,grid.254148.e0000 0001 0033 6389Institute of Infection and Inflammation, China Three Gorges University, Yichang, 443002 China ,grid.254148.e0000 0001 0033 6389College of Basic Medical Science, China Three Gorges University, Yichang, 443002 China
| |
Collapse
|
21
|
Chen Y, Zhu M, Huang B, Jiang Y, Su J. Advances in cell membrane-coated nanoparticles and their applications for bone therapy. BIOMATERIALS ADVANCES 2023; 144:213232. [PMID: 36502750 DOI: 10.1016/j.bioadv.2022.213232] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Revised: 11/24/2022] [Accepted: 11/29/2022] [Indexed: 06/17/2023]
Abstract
Due to the specific structure of natural bone, most of the therapeutics are incapable to be delivered into the targeted site with effective concentrations. Nanotechnology has provided a good way to improve this issue, cell membrane mimetic nanoparticles (NPs) have been emerging as an ideal nanomaterial which integrates the advantages of natural cell membranes with synthetic NPs to significantly improve the biocompatibility as well as achieving long-lasting circulation and targeted delivery. In addition, functionalized modifications of the cell membrane facilitate more precise targeting and therapy. Here, an overview of the preparation of cell membrane-coated NPs and the properties of cell membranes from different cell sources has been given to expatiate their function and potential applications. Strategies for functionalized modification of cell membranes are also briefly described. The application of cell membrane-coated NPs for bone therapy is then presented according to the function of cell membranes. Moreover, the prospects and challenges of cell membrane-coated NPs for translational medicine have also been discussed.
Collapse
Affiliation(s)
- Yutong Chen
- Organoid Research Centre, Institute of Translational Medicine, Shanghai University, Shanghai 200444, PR China; School of Medicine, Shanghai University, Shanghai 200444, PR China; School of Life Sciences, Shanghai University, Shanghai 200444, PR China
| | - Mengru Zhu
- Organoid Research Centre, Institute of Translational Medicine, Shanghai University, Shanghai 200444, PR China; School of Medicine, Shanghai University, Shanghai 200444, PR China
| | - Biaotong Huang
- Organoid Research Centre, Institute of Translational Medicine, Shanghai University, Shanghai 200444, PR China; Wenzhou Institute of Shanghai University, Wenzhou 325000, PR China.
| | - Yingying Jiang
- Organoid Research Centre, Institute of Translational Medicine, Shanghai University, Shanghai 200444, PR China.
| | - Jiacan Su
- Organoid Research Centre, Institute of Translational Medicine, Shanghai University, Shanghai 200444, PR China.
| |
Collapse
|
22
|
Sun H, Zhan M, Mignani S, Shcharbin D, Majoral JP, Rodrigues J, Shi X, Shen M. Modulation of Macrophages Using Nanoformulations with Curcumin to Treat Inflammatory Diseases: A Concise Review. Pharmaceutics 2022; 14:2239. [PMID: 36297677 PMCID: PMC9611033 DOI: 10.3390/pharmaceutics14102239] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2022] [Revised: 10/14/2022] [Accepted: 10/15/2022] [Indexed: 11/16/2022] Open
Abstract
Curcumin (Cur), a traditional Chinese medicine extracted from natural plant rhizomes, has become a candidate drug for the treatment of diseases due to its anti-inflammatory, anticancer, antioxidant, and antibacterial activities. However, the poor water solubility and low bioavailability of Cur limit its therapeutic effects for clinical applications. A variety of nanocarriers have been successfully developed to improve the water solubility, in vivo distribution, and pharmacokinetics of Cur, as well as to enhance the ability of Cur to polarize macrophages and relieve macrophage oxidative stress or anti-apoptosis, thus accelerating the therapeutic effects of Cur on inflammatory diseases. Herein, we review the design and development of diverse Cur nanoformulations in recent years and introduce the biomedical applications and potential therapeutic mechanisms of Cur nanoformulations in common inflammatory diseases, such as arthritis, neurodegenerative diseases, respiratory diseases, and ulcerative colitis, by regulating macrophage behaviors. Finally, the perspectives of the design and preparation of future nanocarriers aimed at efficiently exerting the biological activity of Cur are briefly discussed.
Collapse
Affiliation(s)
- Huxiao Sun
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, College of Biological Science and Medical Engineering, Donghua University, Shanghai 201620, China
| | - Mengsi Zhan
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, College of Biological Science and Medical Engineering, Donghua University, Shanghai 201620, China
| | - Serge Mignani
- CQM—Centro de Química da Madeira, MMRG, Universidade da Madeira, Campus Universitário da Penteada, 9020-105 Funchal, Portugal
- Laboratoire de Chimie et de Biochimie Pharmacologiques et Toxicologique, Université Paris Descartes, PRES Sorbonne Paris Cité, CNRS UMR 860, 45, rue des Saints Peres, 75006 Paris, France
| | - Dzmitry Shcharbin
- Institute of Biophysics and Cell Engineering of NASB, Akademicheskaya 27, 220072 Minsk, Belarus
| | - Jean-Pierre Majoral
- Laboratoire de Chimie de Coordination du CNRS, 205 Route de Narbonne, CEDEX 4, 31077 Toulouse, France
- Laboratoire de Chimie de Coordination du CNRS, Université Toulouse, 31077 Toulouse, France
| | - João Rodrigues
- CQM—Centro de Química da Madeira, MMRG, Universidade da Madeira, Campus Universitário da Penteada, 9020-105 Funchal, Portugal
| | - Xiangyang Shi
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, College of Biological Science and Medical Engineering, Donghua University, Shanghai 201620, China
- CQM—Centro de Química da Madeira, MMRG, Universidade da Madeira, Campus Universitário da Penteada, 9020-105 Funchal, Portugal
| | - Mingwu Shen
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, College of Biological Science and Medical Engineering, Donghua University, Shanghai 201620, China
| |
Collapse
|
23
|
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]
|