1
|
Zhao X, Wu S, Ni S, Zhong Y, Qin X, Zhang K, Qu K, Zhu L, Wu W. Tannic Acid-Based Biomimetic Nanomedicine with Pathological Reactive Oxygen Species-Responsive Cargo Release for Relieving Inflammation in Rheumatoid Arthritis. ACS APPLIED MATERIALS & INTERFACES 2024. [PMID: 39448903 DOI: 10.1021/acsami.4c11494] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/26/2024]
Abstract
Rheumatoid arthritis (RA) is a chronic disease characterized by immune cell infiltration and cartilage damage. The local lesion of RA shows severe oxidative stress and proinflammatory cytokine secretion. For drug therapy, the efficacy of agents, such as methotrexate (MTX), may be greatly limited, resulting from the low bioavailability, immune clearance, and toxic side effects. A nanocarrier (TA-PBA NPs) was developed with anti-inflammatory and antioxidant activities, combined with MTX to prepare nanomedicine (MTX NPs) for synergistic treatment of RA. Moreover, inspired by the biological functions homing to inflammation lesion of macrophages, the biomimetic nanomedicine camouflaged with macrophage membrane (MM@MTX NPs) was constructed. TA-PBA NPs could timely promote MTX release in response to the overaccumulated ROS to exhibit high anti-inflammatory and antioxidant activities for alleviating RA progression. The experimental results confirmed that MM@MTX NPs could significantly reduce the secretion of proinflammatory cytokines (TNF-α) while significantly increasing the typical anti-inflammatory cytokines (IL-10), promote the phenotype transformation of macrophages from M1 to M2, and up-regulate the Nrf2-keap1 pathway-related proteins (HO-1 and NRF2) to positively regulate the local inflammation for effectively inhibiting RA development. Thus, MM@MTX NPs represent a possible candidate as a safe and efficient nanotherapy platform for RA management.
Collapse
Affiliation(s)
- Xiong Zhao
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing 400044, China
| | - Shuai Wu
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing 400044, China
| | - Sheng Ni
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing 400044, China
| | - Yuan Zhong
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing 400044, China
| | - Xian Qin
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing 400044, China
- Chongqing Municipality Clinical Research Center for Endocrinology and Metabolic Diseases, Chongqing University Three Gorges Hospital, Chongqing 404000, China
| | - Kun Zhang
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing 400044, China
- Chongqing Municipality Clinical Research Center for Endocrinology and Metabolic Diseases, Chongqing University Three Gorges Hospital, Chongqing 404000, China
| | - Kai Qu
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing 400044, China
- Chongqing Municipality Clinical Research Center for Endocrinology and Metabolic Diseases, Chongqing University Three Gorges Hospital, Chongqing 404000, China
| | - Li Zhu
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing 400044, China
| | - Wei Wu
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing 400044, China
- JinFeng Laboratory, Chongqing 401329, China
| |
Collapse
|
2
|
Zhu H, Huang D, Wang J, Zhao Y, Sun L. Viral Mimicking Polyplexes as Hierarchical Unpacking Vectors for Rheumatoid Arthritis Treatment. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2402888. [PMID: 38923874 PMCID: PMC11348054 DOI: 10.1002/advs.202402888] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/01/2024] [Revised: 05/08/2024] [Indexed: 06/28/2024]
Abstract
Nano-delivery systems hold great promise for the treatment of rheumatoid arthritis (RA). Current research efforts are primarily focused on enhancing their targeting capabilities and efficacy. Here, this study proposes a novel viral-mimicking ternary polyplexes system for the controlled delivery of the anti-inflammatory drug Cyclosporin A (CsA) to effectively treat RA. The ternary polyplexes consist of a nanogel core loaded with CsA and a hyaluronic acid shell, which facilitates CD44-mediated targeting. By mimicking the Trojan Horse strategy employed by viruses, these polyplexes undergo a stepwise process of deshielding and disintegration within the inflamed joints. This process leads to the release of CsA within the cells and the scavenging of pathogenic factors. This study demonstrates that these viral-mimicking ternary polyplexes exhibit rapid targeting, high accumulation, and prolonged persistence in the joints of RA mice. As a result, they effectively reduce inflammation and alleviate symptoms. These results highlight the potential of viral-mimicking ternary polyplexes as a promising therapeutic approach for the targeted and programmed delivery of drugs to treat not only RA but also other autoimmune diseases.
Collapse
Affiliation(s)
- Haofang Zhu
- Department of Rheumatology and ImmunologyThe First Affiliated Hospital of Anhui Medical University218 Jixi RoadHefei230022P.R. China
- Department of Rheumatology and ImmunologyInstitute of Translational MedicineThe Affiliated Drum Tower Hospital of Nanjing University Medical School321 Zhongshan RoadNanjing210008P. R. China
| | - Danqing Huang
- Department of Rheumatology and ImmunologyInstitute of Translational MedicineThe Affiliated Drum Tower Hospital of Nanjing University Medical School321 Zhongshan RoadNanjing210008P. R. China
| | - Jinglin Wang
- Department of Rheumatology and ImmunologyInstitute of Translational MedicineThe Affiliated Drum Tower Hospital of Nanjing University Medical School321 Zhongshan RoadNanjing210008P. R. China
| | - Yuanjin Zhao
- Department of Rheumatology and ImmunologyInstitute of Translational MedicineThe Affiliated Drum Tower Hospital of Nanjing University Medical School321 Zhongshan RoadNanjing210008P. R. China
- State Key Laboratory of BioelectronicsSchool of Biological Science and Medical EngineeringSoutheast University2 SipailouNanjing210096P. R. China
| | - Lingyun Sun
- Department of Rheumatology and ImmunologyThe First Affiliated Hospital of Anhui Medical University218 Jixi RoadHefei230022P.R. China
- Department of Rheumatology and ImmunologyInstitute of Translational MedicineThe Affiliated Drum Tower Hospital of Nanjing University Medical School321 Zhongshan RoadNanjing210008P. R. China
| |
Collapse
|
3
|
Ruan L, Cai X, Qian R, Bei S, Wu L, Cao J, Shen S. Live macrophages loaded with Fe 3O 4 and sulfasalazine for ferroptosis and photothermal therapy of rheumatoid arthritis. Mater Today Bio 2024; 24:100925. [PMID: 38226012 PMCID: PMC10788618 DOI: 10.1016/j.mtbio.2023.100925] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2023] [Revised: 11/26/2023] [Accepted: 12/18/2023] [Indexed: 01/17/2024] Open
Abstract
Rheumatoid arthritis (RA) is a systemic autoimmune disease characterized by the infiltration of inflammatory cells and proliferation of synovial cells. It can cause cartilage and bone damage as well as disability and is regarded as an incurable chronic disease. Available therapies cannot prevent the development of diseases due to the high toxicity of the therapeutic agents and the inefficient drug delivery. Ferroptosis, an iron-dependent manner of lipid peroxidative cell death, indicates great potential for RA therapy due to ability to damage the infiltrated inflammatory cells and proliferated fibroblast-like synoviocytes. Here, we use macrophages as vector to deliver Fe3O4 nanoparticles and sulfasalazine (SSZ) for ferroptosis and photothermal therapy of RA. The inherent property of migration towards the inflamed joints under the guidance of inflammatory factors enables macrophages to targetedly deliver the payload into the RA. Upon the irradiation of the near infrared light, the Fe3O4 nanoparticles convert the light into heat to damage the proliferated synovium. Meanwhile, the iron released from Fe3O4 nanoparticles works with SSZ to generate synergetic ferroptosis effect. The resident inflammatory cells and proliferated synovium are efficiently damaged by the ferroptosis and photothermal effect, showing pronounced therapeutic effect for RA.
Collapse
Affiliation(s)
- Li Ruan
- Affiliated Hospital of Jiangsu University, Zhenjiang, 212001, China
- College of Pharmaceutical Sciences, Jiangsu University, Zhenjiang, Jiangsu, 212013, China
| | - Xinxi Cai
- Affiliated Hospital of Jiangsu University, Zhenjiang, 212001, China
- College of Pharmaceutical Sciences, Jiangsu University, Zhenjiang, Jiangsu, 212013, China
| | - Rui Qian
- Affiliated Hospital of Jiangsu University, Zhenjiang, 212001, China
- College of Pharmaceutical Sciences, Jiangsu University, Zhenjiang, Jiangsu, 212013, China
| | - Shifang Bei
- The Affiliated People's Hospital, Jiangsu University, Zhenjiang, Jiangsu, 212001, China
| | - Lin Wu
- Affiliated Hospital of Jiangsu University, Zhenjiang, 212001, China
| | - Jin Cao
- College of Pharmaceutical Sciences, Jiangsu University, Zhenjiang, Jiangsu, 212013, China
| | - Song Shen
- College of Pharmaceutical Sciences, Jiangsu University, Zhenjiang, Jiangsu, 212013, China
| |
Collapse
|
4
|
Rubanová D, Skoroplyas S, Libánská A, Randárová E, Bryja J, Chorvátová M, Etrych T, Kubala L. Therapeutic activity and biodistribution of a nano-sized polymer-dexamethasone conjugate intended for the targeted treatment of rheumatoid arthritis. NANOMEDICINE : NANOTECHNOLOGY, BIOLOGY, AND MEDICINE 2024; 55:102716. [PMID: 38738529 DOI: 10.1016/j.nano.2023.102716] [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: 03/21/2023] [Revised: 08/02/2023] [Accepted: 10/21/2023] [Indexed: 05/14/2024]
Abstract
Rheumatoid arthritis is a chronic inflammatory autoimmune disease caused by alteration of the immune system. Current therapies have several limitations and the use of nanomedicines represents a promising strategy to overcome them. By employing a mouse model of adjuvant induced arthritis, we aimed to evaluate the biodistribution and therapeutic effects of glucocorticoid dexamethasone conjugated to a nanocarrier based on biocompatible N-(2-hydroxypropyl) methacrylamide copolymers. We observed an increased accumulation of dexamethasone polymer nanomedicines in the arthritic mouse paw using non-invasive fluorescent in vivo imaging and confirmed it by the analysis of tissue homogenates. The dexamethasone conjugate exhibited a dose-dependent healing effect on arthritis and an improved therapeutic outcome compared to free dexamethasone. Particularly, significant reduction of accumulation of RA mediator RANKL was observed. Overall, our data suggest that the conjugation of dexamethasone to a polymer nanocarrier by means of stimuli-sensitive spacer is suitable strategy for improving rheumatoid arthritis therapy.
Collapse
Affiliation(s)
- Daniela Rubanová
- Institute of Biophysics of the Czech Academy of Sciences, Královopolská 135, 612 00 Brno, Czech Republic; Department of Experimental Biology, Faculty of Science, Masaryk University, Kamenice 5, 625 00 Brno, Czech Republic
| | - Svitlana Skoroplyas
- Institute of Biophysics of the Czech Academy of Sciences, Královopolská 135, 612 00 Brno, Czech Republic; Department of Experimental Biology, Faculty of Science, Masaryk University, Kamenice 5, 625 00 Brno, Czech Republic
| | - Alena Libánská
- Institute of Macromolecular Chemistry of the Czech Academy of Sciences, Heyrovského nám. 2, 162 00 Prague, Czech Republic
| | - Eva Randárová
- Institute of Macromolecular Chemistry of the Czech Academy of Sciences, Heyrovského nám. 2, 162 00 Prague, Czech Republic
| | - Josef Bryja
- Institute of Biophysics of the Czech Academy of Sciences, Královopolská 135, 612 00 Brno, Czech Republic; Department of Experimental Biology, Faculty of Science, Masaryk University, Kamenice 5, 625 00 Brno, Czech Republic
| | - Michaela Chorvátová
- Institute of Biophysics of the Czech Academy of Sciences, Královopolská 135, 612 00 Brno, Czech Republic; Department of Experimental Biology, Faculty of Science, Masaryk University, Kamenice 5, 625 00 Brno, Czech Republic
| | - Tomáš Etrych
- Institute of Macromolecular Chemistry of the Czech Academy of Sciences, Heyrovského nám. 2, 162 00 Prague, Czech Republic
| | - Lukáš Kubala
- Institute of Biophysics of the Czech Academy of Sciences, Královopolská 135, 612 00 Brno, Czech Republic; International Clinical Research Center, St. Anne's University Hospital, Pekařská 53, 602 00 Brno, Czech Republic.
| |
Collapse
|
5
|
Faustino C, Duarte N, Pinheiro L. Triterpenes Drug Delivery Systems, a Modern Approach for Arthritis Targeted Therapy. Pharmaceuticals (Basel) 2023; 17:54. [PMID: 38256888 PMCID: PMC10819636 DOI: 10.3390/ph17010054] [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: 10/27/2023] [Revised: 12/22/2023] [Accepted: 12/26/2023] [Indexed: 01/24/2024] Open
Abstract
Arthritis is a major cause of disability. Currently available anti-arthritic drugs, such as disease-modifying anti-rheumatic drugs (DMARDs), have serious side-effects associated with long-term use. Triterpenoids are natural products with known anti-inflammatory properties, and many have revealed efficiency against arthritis both in vitro and in vivo in several animal models, with negligible cytotoxicity. However, poor bioavailability due to low water solubility and extensive metabolism upon oral administration hinder the therapeutic use of anti-arthritic triterpenoids. Therefore, drug delivery systems (DDSs) able to improve the pharmacokinetic profile of triterpenoids and achieve sustained drug release are useful alternatives for targeted delivery in arthritis treatment. Several DDSs have been described in the literature for triterpenoid delivery, including microparticulate and nanoparticulate DDSs, such as polymeric micro and nanoparticles (NPs), polymeric micelles, liposomes, micro and nanoemulsions, and hydrogels. These systems have shown superior therapeutic effects in arthritis compared to the free drugs and are similar to currently available anti-arthritic drugs without significant side-effects. This review focuses on nanocarriers for triterpenoid delivery in arthritis therapy, including osteoarthritis (OA), rheumatoid arthritis (RA) and gout that appeared in the literature in the last ten years.
Collapse
Affiliation(s)
| | - Noélia Duarte
- Research Institute for Medicines (iMed.ULisboa), Faculdade de Farmácia, Universidade de Lisboa, Avenida Prof. Gama Pinto, 1649-003 Lisbon, Portugal;
| | - Lídia Pinheiro
- Research Institute for Medicines (iMed.ULisboa), Faculdade de Farmácia, Universidade de Lisboa, Avenida Prof. Gama Pinto, 1649-003 Lisbon, Portugal;
| |
Collapse
|
6
|
He ZH, Zou JT, Chen X, Gong JS, Chen Y, Jin L, Liu YW, Rao SS, Yin H, Tan YJ, Wang Z, Du W, Li HM, Qian YX, Wang ZX, Wang YY, Wan TF, Luo Y, Zhu H, Chen CY, Xie H. Ångstrom-scale silver particles ameliorate collagen-induced and K/BxN-transfer arthritis in mice via the suppression of inflammation and osteoclastogenesis. Inflamm Res 2023; 72:2053-2072. [PMID: 37816881 DOI: 10.1007/s00011-023-01778-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Revised: 07/31/2023] [Accepted: 08/02/2023] [Indexed: 10/12/2023] Open
Abstract
OBJECTIVE Nanoparticles (NPs) hold a great promise in combating rheumatoid arthritis, but are often compromised by their toxicities because the currently used NPs are usually synthesized by chemical methods. Our group has previously fabricated Ångstrom-scale silver particles (AgÅPs) and demonstrated the anti-tumor and anti-sepsis efficacy of fructose-coated AgÅPs (F-AgÅPs). This study aimed to uncover the efficacy and mechanisms of F-AgÅPs for arthritis therapy. METHODS We evaluated the efficacy of F-AgÅPs in collagen-induced arthritis (CIA) mice. We also compared the capacities of F-AgÅPs, the commercial AgNPs, and the clinical drug methotrexate (MTX) in protecting against K/BxN serum-transfer arthritis (STA) mice. Moreover, we evaluated the effects of F-AgÅPs and AgNPs on inflammation, osteoclast formation, synoviocytes migration, and matrix metalloproteinases (MMPs) production in vitro and in vivo. Meanwhile, the toxicities of F-AgÅPs and AgNPs in vitro and in vivo were also tested. RESULTS F-AgÅPs significantly prevented bone erosion, synovitis, and cartilage damage, attenuated rheumatic pain, and improved the impaired motor function in mouse models of CIA or STA, the anti-rheumatic effects of which were comparable or stronger than AgNPs and MTX. Further studies revealed that F-AgÅPs exhibited similar or greater inhibitory abilities than AgNPs to suppress inflammation, osteoclast formation, synoviocytes migration, and MMPs production. No obvious toxicities were observed in vitro and in vivo after F-AgÅPs treatment. CONCLUSIONS F-AgÅPs can effectively alleviate arthritis without notable toxicities and their anti-arthritic effects are associated with the inhibition of inflammation, osteoclastogenesis, synoviocytes migration, and MMPs production. Our study suggests the prospect of F-AgÅPs as an efficient and low-toxicity agent for arthritis therapy.
Collapse
Affiliation(s)
- Ze-Hui He
- Department of Orthopedics, Movement System Injury and Repair Research Center, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Hunan Key Laboratory of Angmedicine, Changsha, Hunan, China
- Angmedicine Research Center, Central south university, Changsha, China
| | - Jing-Tao Zou
- Department of Orthopedics, Movement System Injury and Repair Research Center, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Hunan Key Laboratory of Angmedicine, Changsha, Hunan, China
- Angmedicine Research Center, Central south university, Changsha, China
| | - Xia Chen
- Department of Orthopedics, Movement System Injury and Repair Research Center, Xiangya Hospital, Central South University, Changsha, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Jiang-Shan Gong
- Department of Orthopedics, Movement System Injury and Repair Research Center, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Hunan Key Laboratory of Angmedicine, Changsha, Hunan, China
- Angmedicine Research Center, Central south university, Changsha, China
| | - Ya Chen
- Department of Orthopedics, Movement System Injury and Repair Research Center, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Hunan Key Laboratory of Angmedicine, Changsha, Hunan, China
- Angmedicine Research Center, Central south university, Changsha, China
| | - Ling Jin
- Department of Orthopedics, Movement System Injury and Repair Research Center, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Hunan Key Laboratory of Angmedicine, Changsha, Hunan, China
- Angmedicine Research Center, Central south university, Changsha, China
| | - Yi-Wei Liu
- Department of Orthopedics, Movement System Injury and Repair Research Center, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Hunan Key Laboratory of Angmedicine, Changsha, Hunan, China
- Angmedicine Research Center, Central south university, Changsha, China
| | - Shan-Shan Rao
- Department of Orthopedics, Movement System Injury and Repair Research Center, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Hunan Key Laboratory of Angmedicine, Changsha, Hunan, China
- Angmedicine Research Center, Central south university, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Hao Yin
- Department of Orthopedics, Movement System Injury and Repair Research Center, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Hunan Key Laboratory of Angmedicine, Changsha, Hunan, China
- Angmedicine Research Center, Central south university, Changsha, China
| | - Yi-Juan Tan
- Department of Orthopedics, Movement System Injury and Repair Research Center, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Hunan Key Laboratory of Angmedicine, Changsha, Hunan, China
- Angmedicine Research Center, Central south university, Changsha, China
| | - Zun Wang
- Department of Orthopedics, Movement System Injury and Repair Research Center, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Xiangya School of Nursing, Central South University, Changsha, Hunan, China
| | - Wei Du
- Department of Orthopedics, Movement System Injury and Repair Research Center, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Department of Rehabilitation, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
| | - Hong-Ming Li
- Department of Orthopedics, Movement System Injury and Repair Research Center, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Hunan Key Laboratory of Angmedicine, Changsha, Hunan, China
- Angmedicine Research Center, Central south university, Changsha, China
| | - Yu-Xuan Qian
- Department of Orthopedics, Movement System Injury and Repair Research Center, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Hunan Key Laboratory of Angmedicine, Changsha, Hunan, China
- Angmedicine Research Center, Central south university, Changsha, China
| | - Zhen-Xing Wang
- Department of Orthopedics, Movement System Injury and Repair Research Center, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Hunan Key Laboratory of Angmedicine, Changsha, Hunan, China
- Angmedicine Research Center, Central south university, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Yi-Yi Wang
- Department of Orthopedics, Movement System Injury and Repair Research Center, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Hunan Key Laboratory of Angmedicine, Changsha, Hunan, China
- Angmedicine Research Center, Central south university, Changsha, China
| | - Teng-Fei Wan
- Department of Orthopedics, Movement System Injury and Repair Research Center, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Hunan Key Laboratory of Angmedicine, Changsha, Hunan, China
- Angmedicine Research Center, Central south university, Changsha, China
| | - Yi Luo
- Department of Orthopedics, Movement System Injury and Repair Research Center, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Hunan Key Laboratory of Angmedicine, Changsha, Hunan, China
- Angmedicine Research Center, Central south university, Changsha, China
| | - Hao Zhu
- Department of Orthopedics, Movement System Injury and Repair Research Center, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Hunan Key Laboratory of Angmedicine, Changsha, Hunan, China
- Angmedicine Research Center, Central south university, Changsha, China
| | - Chun-Yuan Chen
- Department of Orthopedics, Movement System Injury and Repair Research Center, Xiangya Hospital, Central South University, Changsha, Hunan, China.
- Hunan Key Laboratory of Angmedicine, Changsha, Hunan, China.
- Angmedicine Research Center, Central south university, Changsha, China.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China.
| | - Hui Xie
- Department of Orthopedics, Movement System Injury and Repair Research Center, Xiangya Hospital, Central South University, Changsha, Hunan, China.
- Hunan Key Laboratory of Angmedicine, Changsha, Hunan, China.
- Angmedicine Research Center, Central south university, Changsha, China.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China.
| |
Collapse
|
7
|
Liu X, Chen S, Liu L, Chen Y. Cationic brush hybrid nanoparticles scavenge cell-free DNA to enhance rheumatoid arthritis treatment. Acta Biomater 2023; 170:215-227. [PMID: 37619897 DOI: 10.1016/j.actbio.2023.08.032] [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: 06/03/2023] [Revised: 08/01/2023] [Accepted: 08/16/2023] [Indexed: 08/26/2023]
Abstract
Abnormally high level of cell-free DNA (cfDNA) is one of the important causes of autoimmune diseases, which aggravate the symptoms of rheumatoid arthritis (RA). Recently, the utilization of cationic polymeric nanoparticles for scavenging cfDNA has emerged as a promising therapeutic strategy for the treatment of RA. However, the intravenous introduction of cationic polymeric nanoparticles into the circulation carries a risk of dissociation, causing toxicity. To realize the potential clinical translation, we employed a series of silica particles grafted with poly(2-(dimethylamino) ethyl methacrylate) (PDMA) (SiNP@PDMA) brush, which possess adjustable PDMA content (100, 200, and 300 degree of polymerization (DP)) and particle size (50, 100, and 200 nm diameter), to selectively scavenge cfDNA in inflamed joint cavity. We demonstrate that the binding affinity for cfDNA, cytotoxicity, circulation time in vivo and retention in the inflamed joint cavity are influenced by the core-shell structure of SiNP@PDMA, ultimately impacting therapeutic efficacy. Among them, SiNP@PDMA with 100 nm size and 200 DP of PDMA exhibit enhanced accumulation and prolonged retention time in inflammatory joint cavity, resulting in superior therapeutic effect. Therefore, in this study, applying the precisely tuning size and cation content of SiNP@PDMA, we demonstrated the factors to matter the therapeutic effect of cationic nanoparticles, which deepened the understanding of the anti-inflammatory therapies based on cfDNA scavenger for RA. STATEMENT OF SIGNIFICANCE: Inspired by the discovery that cfDNA would induce inappropriate immune responses to exacerbate the progress of RA, we innovatively employed SiNP@PDMA as a cfDNA scavenger to inhibit cfDNA-induced inflammation in RA. Increase in the cation content efficiently strengthened the binding between SiNP@PDMA and cfDNA, leading to an improvement in inhibitory effect of inflammation. In addition, we compared the behaviors of 50, 100 and 200 nm SiNP@PDMA in RA symptom suppression, local cfDNA scavenging and inflammation inhibition. The results demonstrated that SiNP100-PDMA200 outperformed other analogues, corresponding to their more favorable distribution in inflammatory articular cavity. Together, this study revealed the structure-property relationship of cfDNA scavengers for further development of safe and effective cfDNA scavenging system.
Collapse
Affiliation(s)
- Xingliang Liu
- School of Materials Science and Engineering, Key Laboratory for Polymeric Composite and Functional Materials of Ministry of Education, Sun Yat-sen University, Guangzhou 510275, China
| | - Shi Chen
- School of Materials Science and Engineering, Key Laboratory for Polymeric Composite and Functional Materials of Ministry of Education, Sun Yat-sen University, Guangzhou 510275, China
| | - Lixin Liu
- School of Materials Science and Engineering, Key Laboratory for Polymeric Composite and Functional Materials of Ministry of Education, Sun Yat-sen University, Guangzhou 510275, China; State Key Laboratory of Oncology in Southern China, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, China.
| | - Yongming Chen
- School of Materials Science and Engineering, Key Laboratory for Polymeric Composite and Functional Materials of Ministry of Education, Sun Yat-sen University, Guangzhou 510275, China; Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China; State Key Laboratory of Oncology in Southern China, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, China.
| |
Collapse
|
8
|
Wang P, Zhang Y, Lei H, Yu J, Zhou Q, Shi X, Zhu Y, Zhang D, Zhang P, Wang K, Dong K, Xing J, Dong Y. Hyaluronic acid-based M1 macrophage targeting and environmental responsive drug releasing nanoparticle for enhanced treatment of rheumatoid arthritis. Carbohydr Polym 2023; 316:121018. [PMID: 37321721 DOI: 10.1016/j.carbpol.2023.121018] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Revised: 04/30/2023] [Accepted: 05/09/2023] [Indexed: 06/17/2023]
Abstract
Herein, hyaluronic acid (HA) and β-cyclodextrin (β-CD) is used to form targeted drug delivery platform HCPC/DEX NPs with previously prepared carbon dots (CDs) as cross-linker, dexamethasone (DEX) is loaded for rheumatoid arthritis (RA) treatment. The drug loading capacity of β-CD and M1 macrophage targeting of HA were utilized for efficient delivery of DEX to the inflammatory joints. Because of the environmental responsive degradation of HA, DEX can be released in 24 h and inhibit the inflammatory response in M1 macrophages. The drug loading of NPs is 4.79 %. Cellular uptake evaluation confirmed that NPs can specifically target to M1 macrophages via HA ligands, the uptake of M1 macrophages is 3.7 times that of normal macrophages. In vivo experiments revealed that NPs can accumulate in RA joints to alleviate inflammation and accelerate cartilage healing, the accumulation can be observed in 24 h. The cartilage thickness increased to 0.45 mm after HCPC/DEX NPs treatment, indicating its good RA therapeutic effect. Importantly, this study was the first to utilize the potential acid and reactive oxygen species responsiveness of HA to release drug and prepare M1 macrophage targeting nanodrug for RA treatment, which provides a safe and effective RA therapeutic strategy.
Collapse
Affiliation(s)
- Pengchong Wang
- Department of Pharmacy, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China; Department of Pharmacy, Shaanxi Provincial People's Hospital, Xi'an, Shaanxi, China
| | - Ying Zhang
- Department of Pharmaceutics, School of Pharmacy, Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Hengyu Lei
- Department of Pharmaceutics, School of Pharmacy, Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Jie Yu
- Department of Pharmaceutics, School of Pharmacy, Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Qinyuan Zhou
- Department of Pharmaceutics, School of Pharmacy, Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Xianpeng Shi
- Department of Pharmacy, Shaanxi Provincial People's Hospital, Xi'an, Shaanxi, China
| | - Yaning Zhu
- Department of Pharmacy, Shaanxi Provincial People's Hospital, Xi'an, Shaanxi, China
| | - Dan Zhang
- Department of Pharmacy, Shaanxi Provincial People's Hospital, Xi'an, Shaanxi, China
| | - Peng Zhang
- Department of Pharmacy, Shaanxi Provincial People's Hospital, Xi'an, Shaanxi, China
| | - Ke Wang
- Department of Pharmaceutics, School of Pharmacy, Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Kai Dong
- Department of Pharmaceutics, School of Pharmacy, Xi'an Jiaotong University, Xi'an, Shaanxi, China.
| | - Jianfeng Xing
- Department of Pharmaceutics, School of Pharmacy, Xi'an Jiaotong University, Xi'an, Shaanxi, China.
| | - Yalin Dong
- Department of Pharmacy, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China.
| |
Collapse
|
9
|
Liu F, Dong Z, Li M, Sun J, Hou Z, Younas A, Wan X, Shang H, Zhang N. A macrophage plasma membrane-coated and DNA structured nanomedicine targets to alleviate rheumatoid arthritis via dual inhibition to TNF-α and NF-κB. Int J Pharm 2023:123188. [PMID: 37394158 DOI: 10.1016/j.ijpharm.2023.123188] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2023] [Revised: 06/10/2023] [Accepted: 06/27/2023] [Indexed: 07/04/2023]
Abstract
High heterogenicity of rheumatoid arthritis (RA) leads to poor response in many patients. Combined therapies that simultaneously inhibit multiple proinflammatory targets may improve anti-RA efficacy. However, which monotherapies to combine and how to achieve the combination are critical issues. Here, we design a macrophage plasma membrane-coated and DNA structured nanomedicine to achieve a dual inhibitory therapy to Tumor necrosis factor alpha (TNF-α) and NF-κB. An anti-NF-κB decoy oligodeoxynucleotides (dODN) is first conjugated to a DNA cage with precise numbers and locations (Cage-dODN). Meanwhile, an anti-TNF-α siRNA is anchored to extracted macrophage plasma membrane (siRNA@M). Subsequently, siRNA@M is used to encapsulate Cage-dODN to fabricate siRNA@M(Cage-dODN) (siMCO). The size and zeta potential of siMCO are 63.1 ± 15.7 nm and -20.7 ± 3.8 mV respectively. siMCO shows increased intracellular uptake by inflamed macrophages and enhanced accumulation in inflamed mouse paws. siMCO also reduces pro-inflammatory factors at genetic and protein levels, alleviates arthritic symptoms, and shows no influence to major blood components. These results show that siMCO is a potential targeted, efficient, and safe dual inhibitory therapy for the treatment of inflammatory arthritis. The macrophage plasma membrane can be utilized to improve the targeting, stability, and efficacy of DNA structured nanomedicines.
Collapse
Affiliation(s)
- Fenfen Liu
- Department of Pharmaceutics and Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases of Henan Province, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, Henan, P.R. China; School of Pharmaceutical Sciences, Zhengzhou University of Industrial Technology, Xinzheng City 451100, Henan, P.R.China.
| | - Zhuolin Dong
- Department of Pharmaceutics and Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases of Henan Province, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, Henan, P.R. China.
| | - Mengru Li
- Department of Pharmaceutics and Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases of Henan Province, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, Henan, P.R. China.
| | - Junhong Sun
- Department of Pharmaceutics and Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases of Henan Province, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, Henan, P.R. China; Bayinguoleng Mongolian Autonomous Prefecture People's Hospital, Korla 841000, Xinjiang Uygur Autonomous Region, P.R. China.
| | - Ziye Hou
- Department of Pharmaceutics and Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases of Henan Province, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, Henan, P.R. China.
| | - Ayesha Younas
- Department of Pharmaceutics and Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases of Henan Province, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, Henan, P.R. China.
| | - Xiangling Wan
- Department of Pharmaceutics and Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases of Henan Province, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, Henan, P.R. China.
| | - Hongtao Shang
- School of Sport Sciences (main campus), Zhengzhou University, Zhengzhou 450001, Henan, P.R. China.
| | - Nan Zhang
- Department of Pharmaceutics and Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases of Henan Province, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, Henan, P.R. China.
| |
Collapse
|
10
|
Guo R, Wang L, Huang J, Pang H, Wang L, Zhu B, Tang Y, Ma L, Qiu L. Ultrasound-Targeted Microbubble Destruction-Mediated Cell-Mimetic Nanodrugs for Treating Rheumatoid Arthritis. ACS Biomater Sci Eng 2023. [PMID: 37184981 DOI: 10.1021/acsbiomaterials.3c00475] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/17/2023]
Abstract
Rheumatoid arthritis (RA) is an autoimmune disease that mainly affects joints, and it can lead to disability and damage to vital organs if not diagnosed and treated in time. However, all current therapeutic agents for RA have limitations such as high dose, severe side effects, long-term use, and unsatisfactory therapeutic effects. The long-term use and dose escalation of methotrexate (MTX) may cause mild and severe side effects. To overcome the limitations, it is critical to target drug delivery to the inflamed joints. In this work, we constructed a folic acid-targeted and cell-mimetic nanodrug, MTX-loaded mesoporous silica composite nanoplatform (MMPRF), which can regulate drug release under ultrasound (US) and microbubble (MB) mediation. The targeted delivery and drug therapy were investigated through in vitro RAW264.7 cell experiments and in vivo collagen-induced arthritis animal experiments. The result showed that the targeting ability to the joints of MMPRF was strong and was more significant after US and MB mediation, which can potently reduce joint swelling, bone erosion, and inflammation in joints. This work indicated that the US- and MB-mediated MMPRF not only would be a promising method for synergistic targeted treatment of RA but also may show high potential for serving as a nanomedicine for many other biomedical fields.
Collapse
Affiliation(s)
- Ruiqian Guo
- Department of Ultrasound, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Lei Wang
- Department of Ultrasound, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu 610072, China
- Chinese Academy of Sciences Sichuan Translational Medicine Research Hospital, Chengdu 610072, China
| | - Jianbo Huang
- Department of Ultrasound, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Houqing Pang
- Department of Ultrasound, West China Second University Hospital, Sichuan University, Chengdu 610041, China
| | - Liyun Wang
- Department of Ultrasound, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Bihui Zhu
- Department of Ultrasound, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Yuanjiao Tang
- Department of Ultrasound, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Lang Ma
- Department of Ultrasound, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Li Qiu
- Department of Ultrasound, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu 610041, China
| |
Collapse
|
11
|
Vasdev N, Pawar B, Gupta T, Mhatre M, Tekade RK. A Bird's Eye View of Various Cell-Based Biomimetic Nanomedicines for the Treatment of Arthritis. Pharmaceutics 2023; 15:1150. [PMID: 37111636 PMCID: PMC10146206 DOI: 10.3390/pharmaceutics15041150] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Revised: 03/26/2023] [Accepted: 04/02/2023] [Indexed: 04/08/2023] Open
Abstract
Arthritis is the inflammation and tenderness of the joints because of some metabolic, infectious, or constitutional reasons. Existing arthritis treatments help in controlling the arthritic flares, but more advancement is required to cure arthritis meticulously. Biomimetic nanomedicine represents an exceptional biocompatible treatment to cure arthritis by minimizing the toxic effect and eliminating the boundaries of current therapeutics. Various intracellular and extracellular pathways can be targeted by mimicking the surface, shape, or movement of the biological system to form a bioinspired or biomimetic drug delivery system. Different cell-membrane-coated biomimetic systems, and extracellular-vesicle-based and platelets-based biomimetic systems represent an emerging and efficient class of therapeutics to treat arthritis. The cell membrane from various cells such as RBC, platelets, macrophage cells, and NK cells is isolated and utilized to mimic the biological environment. Extracellular vesicles isolated from arthritis patients can be used as diagnostic tools, and plasma or MSCs-derived extracellular vesicles can be used as a therapeutic target for arthritis. Biomimetic systems guide the nanomedicines to the targeted site by hiding them from the surveillance of the immune system. Nanomedicines can be functionalized using targeted ligand and stimuli-responsive systems to reinforce their efficacy and minimize off-target effects. This review expounds on various biomimetic systems and their functionalization for the therapeutic targets of arthritis treatment, and discusses the challenges for the clinical translation of the biomimetic system.
Collapse
Affiliation(s)
| | | | | | | | - Rakesh Kumar Tekade
- National Institute of Pharmaceutical Education and Research (NIPER), Ahmedabad, Department of Pharmaceuticals, Ministry of Chemicals and Fertilizers, Opposite Air Force Station, Palaj, Gandhinagar 382355, Gujarat, India
| |
Collapse
|
12
|
Tao J, Yang P, Gao M, Zhang F, Wu Y, Jiang Y, Ning Y, Li Z, Ai F. Reversing inflammatory microenvironment by a single intra-articular injection of multi-stimulus responsive lipogel to relieve rheumatoid arthritis and promote joint repair. Mater Today Bio 2023; 20:100622. [PMID: 37056918 PMCID: PMC10085779 DOI: 10.1016/j.mtbio.2023.100622] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Revised: 03/23/2023] [Accepted: 03/30/2023] [Indexed: 04/03/2023] Open
Abstract
Rheumatoid arthritis (RA) is a common chronic disease dominated by inflammatory synovitis, which is characterized with hyperplastic synovium, up-regulated matrix metalloproteinase (MMP) expression, hypoxic joint cavity and excessive reactive oxygen species (ROS) accumulation. Such local adverse microenvironment in RA joints further exacerbates the infiltration of synovial inflammatory cells, especially M1-type macrophages. Regulating intra-articular pathological conditions, eliminating excess M1 macrophages or converting them to an anti-inflammatory M2 phenotype may break the vicious progression circle. Herein, we develop a multi-stimulus responsive lipogel as effective platform to relieve RA symptoms and promote articular cartilage recovery via reversing its inflammatory microenvironment. The injectable lipogel is fabricated by loading polydopamine nanoparticles and methotrexate into a thermosensitive gel, and intra-articularly injected to form the therapeutic depot (PDA/MTX@TSG) in situ. The gel degrades slowly under esterase hydrolysis, and maintains sustained drug release in physiological conditions. Meanwhile, it can 1) induce a reversible gel-sol phase transition upon mild photothermal treatment (external NIR light control), and 2) specifically respond to MMP-rich RA microenvironment (internal enzymatic hydrolysis effect). Such stimulus-responsive system can deliver therapeutic components in a controllable manner, and significantly reverse adverse inflammatory microenvironment of RA joints through ROS eliminating, hypoxia alleviating, and M1-M2 macrophage polarization effects. Animal experiments indicate that observable RA relief and joint repair are realized after a single lipogel injection combined with NIR irradiation. Our study highlights the importance of altering local RA microenvironment via anti-inflammatory macrophage polarization, and therefore presents a potent therapeutic strategy for RA treatment in clinical intervention.
Collapse
Affiliation(s)
- Jun Tao
- The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330000, PR China
| | - Peng Yang
- The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330000, PR China
| | - Mingying Gao
- Pingyang Hospital Affiliated to Wenzhou Medical University, Wenzhou, Zhejiang 325400, PR China
| | - Fan Zhang
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, PR China
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Wenzhou, Zhejiang 325000, PR China
| | - Yongzhong Wu
- Department of Orthopaedics, The Second People's Hospital of Jingdezhen, Jingdezhen, Jiangxi 333099, PR China
| | - Yan Jiang
- The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330000, PR China
| | - Yunxuan Ning
- The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330000, PR China
- Corresponding author. : .
| | - Zhenglin Li
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, PR China
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Wenzhou, Zhejiang 325000, PR China
- Corresponding author.School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, PR China .
| | - Fanrong Ai
- School of Advanced Manufacturing, Nanchang University, Nanchang, Jiangxi 330031, PR China
- Corresponding author. .
| |
Collapse
|
13
|
Yu X, Mai Y, Wei Y, Yu N, Gao T, Yang J. Therapeutic potential of tolerance-based peptide vaccines in autoimmune diseases. Int Immunopharmacol 2023; 116:109740. [PMID: 36696858 DOI: 10.1016/j.intimp.2023.109740] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2022] [Revised: 01/04/2023] [Accepted: 01/13/2023] [Indexed: 01/24/2023]
Abstract
Autoimmune diseases are caused by the dysfunction of the body's immune regulatory system, which leads to the recognition of self-antigens and the destruction of self-tissues and is mediated by immune cells such as T and B cells, and affects 5-10% of the population worldwide. Current treatments such as non-steroidal anti-inflammatory drugs and glucocorticoids can only relieve symptoms of the disease and are accompanied by serious side effects that affect patient quality of life. The recent rise in antigen-specific therapies, especially vaccines carrying autoantigenic peptides, promises to change this disadvantage, where research has increased dramatically in the last decade. This therapy established specific immune tolerance by delivering peptide fragments containing disease-specific self-antigen epitopes to suppress excessive immune responses, thereby exerting a therapeutic effect, with high safety and specificity. This article presents the latest progress on the treatment of autoimmune diseases with autoantigen peptide vaccines. It includes the construction of peptide vaccine delivery system, the mechanism of inducing immune tolerance and its application.
Collapse
Affiliation(s)
- Xueting Yu
- Department of Pharmaceutics, School of Pharmacy, Ningxia Medical University, Yinchuan, Ningxia, China
| | - Yaping Mai
- School of Science and Technology Centers, Ningxia Medical University, Yinchuan, Ningxia, China
| | - Yaya Wei
- Department of Pharmaceutics, School of Pharmacy, Ningxia Medical University, Yinchuan, Ningxia, China
| | - Na Yu
- Department of Pharmaceutical Preparation, General Hospital of Ningxia Medical University, Yinchuan, Ningxia, China
| | - Ting Gao
- Department of Pharmaceutical Preparation, General Hospital of Ningxia Medical University, Yinchuan, Ningxia, China.
| | - Jianhong Yang
- Department of Pharmaceutics, School of Pharmacy, Ningxia Medical University, Yinchuan, Ningxia, China.
| |
Collapse
|
14
|
Zhang L, Meng W, Chen X, Wu L, Chen M, Zhou Z, Chen Y, Yuan L, Chen M, Chen J, Shui P. Multifunctional Nanoplatform for Mild Microwave-Enhanced Thermal, Antioxidative, and Chemotherapeutic Treatment of Rheumatoid Arthritis. ACS APPLIED MATERIALS & INTERFACES 2023; 15:10341-10355. [PMID: 36790223 DOI: 10.1021/acsami.2c19198] [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: 06/18/2023]
Abstract
Rheumatoid arthritis (RA) is usually associated with excessive proliferation of M1-type proinflammatory macrophages, resulting in severe hypoxia and excess reactive oxygen species (ROS) in the joint cavity. Inhibiting M1-type proinflammatory macrophages and/or repolarizing them into M2 phenotype anti-inflammatory cells by alleviating hypoxia and scavenging ROS could be a promising strategy for RA treatment. In this work, a microwave-sensitive metal-organic framework of UiO-66-NH2 is constructed for coating a nanoenzyme of cerium oxide (CeO2) and loading with the drug celastrol (Cel) to give UiO-66-NH2/CeO2/Cel, which is ultimately wrapped with hyaluronic acid (HA) to form a nanocomposite UiO-66-NH2/CeO2/Cel@HA (UCCH). With the microwave-susceptible properties of UiO-66-NH2, the thermal effect of microwaves can eliminate the excessive proliferation of inflammatory cells. In addition, superoxide-like and catalase-like activities originating from CeO2 in UCCH are boosted to scavenge ROS and accelerate the decomposition of H2O2 to produce O2 under microwave irradiation. The nonthermal effect of microwaves could synergistically promote the repolarization of M1-type macrophages into the M2 phenotype. Accompanied by the release of the anti-RA chemotherapeutic drug Cel, UCCH can efficiently ameliorate RA in vitro and in vivo through microwave-enhanced multisynergistic effects. This strategy could inspire the design of other multisynergistic platforms enhanced by microwaves to exploit new treatment modalities in RA therapies.
Collapse
Affiliation(s)
- Lianying Zhang
- School of Pharmacy, Southwest Medical University, Luzhou 646000, China
| | - Wei Meng
- Guangdong Provincial Key Laboratory of New Drug Screening, Guangzhou Key Laboratory of Drug Research for Emerging Virus Prevention and Treatment, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Xiaotong Chen
- Guangdong Provincial Key Laboratory of New Drug Screening, Guangzhou Key Laboratory of Drug Research for Emerging Virus Prevention and Treatment, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Libo Wu
- School of Pharmacy, Southwest Medical University, Luzhou 646000, China
| | - Mingwa Chen
- Guangdong Provincial Key Laboratory of New Drug Screening, Guangzhou Key Laboratory of Drug Research for Emerging Virus Prevention and Treatment, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Zhaoxi Zhou
- Guangdong Provincial Key Laboratory of New Drug Screening, Guangzhou Key Laboratory of Drug Research for Emerging Virus Prevention and Treatment, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Yongjian Chen
- Guangdong Provincial Key Laboratory of New Drug Screening, Guangzhou Key Laboratory of Drug Research for Emerging Virus Prevention and Treatment, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Lixia Yuan
- School of Chinese Medicine, Southern Medical University, Guangzhou 510515, China
| | - Ming Chen
- The People's Hospital of Gaozhou, Maoming 525200, China
| | - Jinxiang Chen
- Guangdong Provincial Key Laboratory of New Drug Screening, Guangzhou Key Laboratory of Drug Research for Emerging Virus Prevention and Treatment, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Pixian Shui
- School of Pharmacy, Southwest Medical University, Luzhou 646000, China
| |
Collapse
|
15
|
Han H, Xing J, Chen W, Jia J, Li Q. Fluorinated polyamidoamine dendrimer-mediated miR-23b delivery for the treatment of experimental rheumatoid arthritis in rats. Nat Commun 2023; 14:944. [PMID: 36805456 PMCID: PMC9941585 DOI: 10.1038/s41467-023-36625-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Accepted: 02/10/2023] [Indexed: 02/22/2023] Open
Abstract
In rheumatoid arthritis (RA), insufficient apoptosis of macrophages and excessive generation of pro-inflammatory cytokines are intimately connected, accelerating the development of disease. Here, a fluorinated polyamidoamine dendrimer (FP) is used to deliver miR-23b to reduce inflammation by triggering the apoptosis of as well as inhibiting the inflammatory response in macrophages. Following the intravenous injection of FP/miR-23b nanoparticles in experimental RA models, the nanoparticles show therapeutic efficacy with inhibition of inflammatory response, reduced bone and cartilage erosion, suppression of synoviocyte infiltration and the recovery of mobility. Moreover, the nanoparticles accumulate in the inflamed joint and are non-specifically captured by synoviocytes, leading to the restoration of miR-23b expression in the synovium. The miR-23b nanoparticles target Tab2, Tab3 and Ikka to regulate the activation of NF-κB pathway in the hyperplastic synovium, thereby promoting anti-inflammatory and anti-proliferative responses. Additionally, the intravenous administration of FP/miR-23b nanoparticles do not induce obvious systemic toxicity. Overall, our work demonstrates that the combination of apoptosis induction and inflammatory inhibition could be a promising approach in the treatment of RA and possibly other autoimmune diseases.
Collapse
Affiliation(s)
- Haobo Han
- Key Laboratory for Molecular Enzymology and Engineering of Ministry of Education, School of Life Sciences, Jilin University, 130012, Changchun, China
| | - Jiakai Xing
- Key Laboratory for Molecular Enzymology and Engineering of Ministry of Education, School of Life Sciences, Jilin University, 130012, Changchun, China
| | - Wenqi Chen
- Key Laboratory for Molecular Enzymology and Engineering of Ministry of Education, School of Life Sciences, Jilin University, 130012, Changchun, China
| | - Jiaxin Jia
- Key Laboratory for Molecular Enzymology and Engineering of Ministry of Education, School of Life Sciences, Jilin University, 130012, Changchun, China
| | - Quanshun Li
- Key Laboratory for Molecular Enzymology and Engineering of Ministry of Education, School of Life Sciences, Jilin University, 130012, Changchun, China.
| |
Collapse
|
16
|
Logesh K, Raj B, Bhaskaran M, Thirumaleshwar S, Gangadharappa H, Osmani R, Asha Spandana K. Nanoparticulate drug delivery systems for the treatment of rheumatoid arthritis: A comprehensive review. J Drug Deliv Sci Technol 2023. [DOI: 10.1016/j.jddst.2023.104241] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/10/2023]
|
17
|
Haemmerich D, Ramajayam KK, Newton DA. Review of the Delivery Kinetics of Thermosensitive Liposomes. Cancers (Basel) 2023; 15:cancers15020398. [PMID: 36672347 PMCID: PMC9856714 DOI: 10.3390/cancers15020398] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Revised: 01/02/2023] [Accepted: 01/03/2023] [Indexed: 01/10/2023] Open
Abstract
Thermosensitive liposomes (TSL) are triggered nanoparticles that release the encapsulated drug in response to hyperthermia. Combined with localized hyperthermia, TSL enabled loco-regional drug delivery to tumors with reduced systemic toxicities. More recent TSL formulations are based on intravascular triggered release, where drug release occurs within the microvasculature. Thus, this delivery strategy does not require enhanced permeability and retention (EPR). Compared to traditional nanoparticle drug delivery systems based on EPR with passive or active tumor targeting (typically <5%ID/g tumor), TSL can achieve superior tumor drug uptake (>10%ID/g tumor). Numerous TSL formulations have been combined with various drugs and hyperthermia devices in preclinical and clinical studies over the last four decades. Here, we review how the properties of TSL dictate delivery and discuss the advantages of rapid drug release from TSL. We show the benefits of selecting a drug with rapid extraction by tissue, and with quick cellular uptake. Furthermore, the optimal characteristics of hyperthermia devices are reviewed, and impact of tumor biology and cancer cell characteristics are discussed. Thus, this review provides guidelines on how to improve drug delivery with TSL by optimizing the combination of TSL, drug, and hyperthermia method. Many of the concepts discussed are applicable to a variety of other triggered drug delivery systems.
Collapse
Affiliation(s)
- Dieter Haemmerich
- Department of Pediatrics, Medical University of South Carolina, Charleston, SC 29425, USA
- Department of Bioengineering, Clemson University, Clemson, SC 29634, USA
- Correspondence:
| | - Krishna K. Ramajayam
- Department of Pediatrics, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Danforth A. Newton
- Department of Pediatrics, Medical University of South Carolina, Charleston, SC 29425, USA
| |
Collapse
|
18
|
Inflammation-responsive nanoparticles suppress lymphatic clearance for prolonged arthritis therapy. J Control Release 2022; 352:700-711. [PMID: 36347402 DOI: 10.1016/j.jconrel.2022.11.005] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Revised: 10/24/2022] [Accepted: 11/02/2022] [Indexed: 11/11/2022]
Abstract
The clearance of nanomedicine in inflamed joints has been accelerated due to the increased lymph angiogenesis and lymph flow in arthritic sites. To maximize the therapeutic efficacy for rheumatoid arthritis (RA), it is necessary to facilitate targeted delivery and extended drug retention in inflamed synovium simultaneously. In general, nanosized particles are more likely to achieve prolonged circulation and targeted delivery. While drug carriers with larger dimension might be more beneficial for extending drug retention. To balance the conflicting requirements, an inflammation-responsive shape transformable nanoparticle, comprised of amyloid β-derived KLVFF peptide and polysialic acid (PSA), coupled with therapeutic agent dexamethasone (Dex) via an acid-sensitive linker, was fabricated and termed as Dex-KLVFF-PSA (DKPNPs). Under physiological condition, DKPNPs can keep stable nanosized morphology, and PSA shell could endow DKPNPs with long circulation and active targeting to arthritic sites. While in inflamed joints, acidic pH-triggered Dex dissociation or macrophages-induced specific binding with PSA would induce the re-assembly of DKPNPs from nanoparticles to nanofibers. Our results reveal that intravenously injected DKPNPs display prolonged in vivo circulation and preferential distribution in inflamed joints, where DKPNPs undergo shape transition to fibrous structures, leading to declined lymphatic clearance and prolonged efficacy. Overall, our dual-stimulus responsive transformable nanoparticle offers an intelligent solution to achieve enhanced therapeutic efficacy in RA.
Collapse
|
19
|
Jeong M, Jung Y, Yoon J, Kang J, Lee SH, Back W, Kim H, Sailor MJ, Kim D, Park JH. Porous Silicon-Based Nanomedicine for Simultaneous Management of Joint Inflammation and Bone Erosion in Rheumatoid Arthritis. ACS NANO 2022; 16:16118-16132. [PMID: 36214219 DOI: 10.1021/acsnano.2c04491] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
The lack of drugs that target both disease progression and tissue preservation makes it difficult to effectively manage rheumatoid arthritis (RA). Here, we report a porous silicon-based nanomedicine that efficiently delivers an antirheumatic drug to inflamed synovium while degrading into bone-remodeling products. Methotrexate (MTX) is loaded into the porous silicon nanoparticles using a calcium silicate based condenser chemistry. The calcium silicate-porous silicon nanoparticle constructs (pCaSiNPs) degrade and release the drug preferentially in an inflammatory environment. The biodegradation products of the pCaSiNP drug carrier are orthosilicic acid and calcium ions, which exhibit immunomodulatory and antiresorptive effects. In a mouse model of collagen-induced arthritis, systemically administered MTX-loaded pCaSiNPs accumulate in the inflamed joints and ameliorate the progression of RA at both early and established stages of the disease. The disease state readouts show that the combination is more effective than the monotherapies.
Collapse
Affiliation(s)
- Moonkyoung Jeong
- Department of Bio and Brain Engineering and KAIST Institute for Health Science and Technology, Korea Advanced Institute of Science and Technology (KAIST), Daejeon34141, Republic of Korea
| | - Yuna Jung
- Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul02447, Republic of Korea
| | - Junyong Yoon
- Department of Bio and Brain Engineering and KAIST Institute for Health Science and Technology, Korea Advanced Institute of Science and Technology (KAIST), Daejeon34141, Republic of Korea
| | | | - Seo Hyeon Lee
- Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul02447, Republic of Korea
| | - Woojin Back
- Department of Bio and Brain Engineering and KAIST Institute for Health Science and Technology, Korea Advanced Institute of Science and Technology (KAIST), Daejeon34141, Republic of Korea
| | - Hyoyeon Kim
- Department of Bio and Brain Engineering and KAIST Institute for Health Science and Technology, Korea Advanced Institute of Science and Technology (KAIST), Daejeon34141, Republic of Korea
| | | | - Dokyoung Kim
- Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul02447, Republic of Korea
| | - Ji-Ho Park
- Department of Bio and Brain Engineering and KAIST Institute for Health Science and Technology, Korea Advanced Institute of Science and Technology (KAIST), Daejeon34141, Republic of Korea
| |
Collapse
|
20
|
Xu Y, Zhang Z, He J, Chen Z. Immune Effects of Macrophages in Rheumatoid Arthritis: A Bibliometric Analysis From 2000 to 2021. Front Immunol 2022; 13:903771. [PMID: 36172378 PMCID: PMC9510364 DOI: 10.3389/fimmu.2022.903771] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Accepted: 08/11/2022] [Indexed: 01/03/2023] Open
Abstract
Rheumatoid arthritis (RA) is a chronic autoimmune inflammatory disease characterized by macrophage activation. The current characteristics, hotspots, and research frontiers of macrophage-related RA were analyzed using bibliometric analysis. Relatedpapers published from 2000 to 2021 in the Web of Science database were retrieved. The diagrams were generated and analyzed using the bibliometric software package. VOSviewer and CiteSpace were used to evaluate and visualize the research trends and hotspots in macrophage-related RA. A total of 7253 original articles were obtained. Global research on macrophage-related RA is in an advanced stage of development, with core authors, teams and research institutions emerging. United States has published the most papers, received the most citations, and had the highest H-index over the last 22 years. The University of Amsterdam and the journal of Arthritis and Rheumatism are the most productive research institutions and journals. Tak PP's (St Vincent's Hospital) paper has the highest publication and citation scores. The keywords "bone loss" and "polarization" have the highest frequency. Additionally, the study of macrophage polarization in RA has been research focus in recent years. This study demonstrates that research on macrophages in RA will continue. China is a significant producer, whereas the United States is an influential nation in this regard. In the last decade, most studies have concentrated on fundamental research. Recent studies have shown how macrophages play a role in controlling and weakening inflammation, and drug delivery and mechanism have come to the fore.
Collapse
Affiliation(s)
- YunLing Xu
- Department of Basic Medical, Zhejiang Academy of Traditional Chinese Medicine, Hangzhou, China
| | - Zhongmin Zhang
- College of Pharmacy, Guangxi University of Chinese Medicine, Nanning, China
| | - Jiaolong He
- Department of Intensive Care, First Affiliated Hospital of Jishou University, Jishou, China
| | - Zhenxing Chen
- College of Pharmacy, Guangxi University of Chinese Medicine, Nanning, China
| |
Collapse
|
21
|
Ruvalcaba-Ontiveros RI, González-Chávez SA, Carrasco-Hernández AR, López-Loeza SM, Castellanos-Ponce I, Vázquez-Olvera G, Neri-Flores MÁ, Espino-Solís GP, Duarte-Moller JA, Pacheco-Tena C, Esparza-Ponce HE. Treatment with silica-gold nanostructures decreases inflammation-related gene expression in collagen-induced arthritis. Biomater Sci 2022; 10:5216-5229. [PMID: 35903989 DOI: 10.1039/d2bm00498d] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Gold salts have been used to treat rheumatoid arthritis (RA) since the 1940s, and, with advances in nanotechnology, the use of nanogold provides multiple options for anti-inflammatory therapies. This paper presents the synthesis and characterization of silica-gold nanostructures (SGNs) and their therapeutic effect in collagen-induced arthritis (CIA) in DBA/1 mice. At the end of the treatment, the synovial membranes, kidneys, livers, and spleens were dissected and analyzed by inductively coupled plasma mass spectroscopy (ICP) showing less than 0.0001 and 0.1% of the administered doses of Au and Si, respectively. Remains of the SGNs were visually identified in the synovial membrane by scanning electron microscopy (SEM), and the bone density of the hind paws was observed by computerized tomography (CT) indicating a reduction of porosity in the CIA-experimental group. The DNA microarray analysis carried out with RNA obtained from the hind paws showed 2628 differentially expressed genes (DEGs) by SGNs. The bioinformatic analysis showed that DEGs were significantly associated with several inflammatory signalling pathways including chemokines, cytokine-cytokine receptor interaction, PI3K-Akt, TNF, IL-17, NFκβ, MAPK, and RA. SGNs downregulated relevant inflammatory genes in the arthritic joints, including Tnf, Ifng, Il6, and Cxcl5; immunohistochemistry (IHC) confirmed the reduction of TNFα, IL-6, NFκβ, and VEGF in the joints due to the effect of SGNs. TNFα and IL-6 were also reduced in the serum of DBA/1 mice treated with SGNs.
Collapse
Affiliation(s)
- Rosa Isela Ruvalcaba-Ontiveros
- Centro de Investigación en Materiales Avanzados, S.C., Av. Miguel de Cervantes Saavedra 120, Chihuahua, Chihuahua, 31136, Mexico.
| | - Susana Aideé González-Chávez
- Laboratorio PABIOM, Facultad de Medicina y Ciencias Biomédicas, Universidad Autónoma de Chihuahua, Circuito Universitario Campus II, Chihuahua, Chihuahua, 31125, Mexico.
| | - Anel Rocío Carrasco-Hernández
- Centro de Investigación en Materiales Avanzados, S.C., Av. Miguel de Cervantes Saavedra 120, Chihuahua, Chihuahua, 31136, Mexico.
| | - Salma Marcela López-Loeza
- Laboratorio PABIOM, Facultad de Medicina y Ciencias Biomédicas, Universidad Autónoma de Chihuahua, Circuito Universitario Campus II, Chihuahua, Chihuahua, 31125, Mexico.
| | - Ivonne Castellanos-Ponce
- Laboratorio PABIOM, Facultad de Medicina y Ciencias Biomédicas, Universidad Autónoma de Chihuahua, Circuito Universitario Campus II, Chihuahua, Chihuahua, 31125, Mexico.
| | - Gregorio Vázquez-Olvera
- Centro de Investigación en Materiales Avanzados, S.C., Av. Miguel de Cervantes Saavedra 120, Chihuahua, Chihuahua, 31136, Mexico.
| | - Miguel Ángel Neri-Flores
- Centro de Investigación en Materiales Avanzados, S.C., Av. Miguel de Cervantes Saavedra 120, Chihuahua, Chihuahua, 31136, Mexico.
| | - Gerardo Pavel Espino-Solís
- Translational Research Laboratory and National Laboratory of Flow Cytometry, Autonomous University of Chihuahua, Circuito Universitario Campus II, Chihuahua, Chihuahua, 31109, Mexico.
| | - José Alberto Duarte-Moller
- División de Ciencias e Ingeniería, Universidad de Sonora, Unidad Regional Sur, Lázaro Cárdenas del Río 100. Colonia Francisco Villa, Navojoa Son. 85880, Mexico.
| | - César Pacheco-Tena
- Laboratorio PABIOM, Facultad de Medicina y Ciencias Biomédicas, Universidad Autónoma de Chihuahua, Circuito Universitario Campus II, Chihuahua, Chihuahua, 31125, Mexico.
| | - Hilda Esperanza Esparza-Ponce
- Centro de Investigación en Materiales Avanzados, S.C., Av. Miguel de Cervantes Saavedra 120, Chihuahua, Chihuahua, 31136, Mexico.
| |
Collapse
|
22
|
The protein corona modulates the inflammation inhibition by cationic nanoparticles via cell-free DNA scavenging. Bioact Mater 2022; 13:249-259. [PMID: 35224306 PMCID: PMC8843952 DOI: 10.1016/j.bioactmat.2021.10.044] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2021] [Revised: 10/06/2021] [Accepted: 10/28/2021] [Indexed: 12/25/2022] Open
Abstract
A central paradigm in nanomedicine is that when synthetic nanoparticles (NPs) enter the body, they are immediately cloaked by a corona of macromolecules (mostly proteins) that mediates the role of the physico-chemical properties in the NP biological functions (the “coronation paradigm”). In this work, we focused on the assessment of the “coronation paradigm” for cationic NPs (cNPs) used as rheumatoid arthritis (RA) drugs due to their ability to scavenge cell-free DNA (cfDNA). We fabricated series of cNPs uniformly coated with single or di-hydroxyl groups and different types of amino groups and showed that hydroxylated nanoparticles displayed a prolonged retention in inflamed joints and greater anti-inflammatory effect in collagen-induced arthritis (CIA) rats than the non-hydroxylated analogues. Especially, the cNPs with secondary amines and a di-hydroxyl shell showed the best performance among the tested cNPs. Proteomic analysis showed that the cNPs with a di-hydroxyl shell adsorbed less opsonin proteins than the cNPs carrying mono hydroxyl groups and non-hydroxylated ones, which may provide a mechanistic explanation for the different biodistribution profiles of cNPs. Thus, this study suggests that the protein corona mediates the effects of the surface chemistry on the fate and functions of cNPs as anti-RA drugs. cNPs with hydroxyl shell decreased side-effects to cells and CIA rats without decreasing the ability to scavenge cfDNA. Hydroxyl shell reduced the adsorption of proteins, thus prolonged the circulation and site accumulation of cNPs. The composition of the protein corona formed onto the cNPs in the serum can be modulated by their synthetic identity.
Collapse
|
23
|
Li C, Zheng X, Hu M, Jia M, Jin R, Nie Y. Recent progress in therapeutic strategies and biomimetic nanomedicines for rheumatoid arthritis treatment. Expert Opin Drug Deliv 2022; 19:883-898. [PMID: 35760767 DOI: 10.1080/17425247.2022.2094364] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
INTRODUCTION Rheumatoid arthritis (RA) is an autoimmune systemic disease in which inflammatory and immune cells accumulate in inflamed joints. Researchers aimed at the characteristics of RA to achieve the effect of treating RA through different therapeutic strategies, and have used various endogenous materials to design drug-loaded nanoparticles that can target RA by binding to cell adhesion molecules or chemokines. In some cases, the nanoparticles can respond to the characteristics of the microenvironment. AREAS COVERED This article reviews the recent advances in the treatment of RA from two aspects of therapeutic strategies and delivery strategies. Therapeutic strategies mainly include neutralization of inflammatory factors, promotion of inflammatory cell apoptosis, ROS scavenger, immunosuppression, and bone tissue repair. The drug delivery strategy is mainly described from two aspects: chemically functionalized biomimetic nanoparticles and endogenous nanoparticles. EXPERT OPINION Biomimetic NPs may be effective drug carriers for targeted RA treatment. NPs can reduce the clearance of mononuclear phagocytes, prolong the blood circulation time, and improve the targeting ability. With the deepening of research, more and more biomimetic NPs have entered the clinical trial stage. However, safe and scalable preparation methods are needed to improve their clinical applicability.
Collapse
Affiliation(s)
- Chunhong Li
- Department of Pharmaceutical Sciences, School of Pharmacy, Southwest Medical University, Luzhou 646000, Sichuan, China.,National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, 610064, P. R. China
| | - Xiu Zheng
- Department of Pharmaceutical Sciences, School of Pharmacy, Southwest Medical University, Luzhou 646000, Sichuan, China
| | - Mei Hu
- Pharmacy Laboratory, School of Pharmacy, Southwest Medical University, Luzhou 646000, Sichuan, China
| | - Ming Jia
- Department of Pharmaceutical Sciences, School of Pharmacy, Southwest Medical University, Luzhou 646000, Sichuan, China
| | - Rongrong Jin
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, 610064, P. R. China
| | - Yu Nie
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, 610064, P. R. China
| |
Collapse
|
24
|
Wang X, Cao W, Sun C, Wang Y, Wang M, Wu J. Development of pH-sensitive dextran-based methotrexate nanodrug for rheumatoid arthritis therapy through inhibition of JAK-STAT pathways. Int J Pharm 2022; 622:121874. [PMID: 35636630 DOI: 10.1016/j.ijpharm.2022.121874] [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: 01/16/2022] [Revised: 04/18/2022] [Accepted: 05/24/2022] [Indexed: 11/08/2022]
Abstract
Rheumatoid arthritis (RA) is a chronic and symmetrical autoimmune disease that primarily characterized with articular synovial hyperplasia, joint swelling, cartilage and bone destruction. The in-depth understanding of the role of immune signaling pathway inhibitors provides inspiration for the construction of new and more effective strategy for RA therapy. In this study, by loading methotrexate (MTX) into an acetalated dextran biopolymer, AcDEX, we developed a pH-sensitive, MTX-loaded and molecularly targeted nanodrug MTX@pH-AcDEX NPs) to decrease the toxicity of MTX and simultaneously enhance its therapeutic effect. The resultant MTX@pH-AcDEX NPs showed the spherical morphology and notable pH-responsiveness with high drug loading of 88.32%. As demonstrated in vitro and in vivo, the reduced cytotoxicity of both RAW264.7 cells and LPS-activated RAW264.7 cells treated with MTX@pH-AcDEX NPs was found compared to free MTX. Upon intravenous administration into adjuvant-induced arthritis (AIA) rat model, the nanodrug had potent pharmacokinetic and pharmacodynamic profiles, which can accumulate in RA lesions and release MTX inhibitors for regulating the JAK-STAT pathways. As a result, the MTX@pH-AcDEX NPs achieved the cartilage and bone protective and a better anti-inflammatory effect with negligible systemic toxicity, suggesting the strong potential of safe and effective nanodrug for RA therapy as well as other autoimmune diseases.
Collapse
Affiliation(s)
- Xianbin Wang
- Department of Rheumatology and Immunology, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai 264000, China
| | - Wenjun Cao
- Department of Rheumatology and Immunology, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai 264000, China
| | - Chuanfen Sun
- Department of Rheumatology and Immunology, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai 264000, China
| | - Yutie Wang
- Department of Rheumatology and Immunology, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai 264000, China
| | - Mingyu Wang
- Department of Rheumatology and Immunology, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai 264000, China.
| | - Jiarong Wu
- Department of Rheumatology and Immunology, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai 264000, China.
| |
Collapse
|
25
|
Rahman MM, Islam F, Afsana Mim S, Khan MS, Islam MR, Haque MA, Mitra S, Emran TB, Rauf A. Multifunctional Therapeutic Approach of Nanomedicines against Inflammation in Cancer and Aging. JOURNAL OF NANOMATERIALS 2022; 2022:1-19. [DOI: 10.1155/2022/4217529] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/01/2023]
Abstract
Cancer is a fatal disorder that affects people across the globe, yet existing therapeutics are ineffective. The development of submicrometer transport for optimizing the biodistribution of systemically provided medications is the focus of nanomedicine. Nanoparticle- (NP-) based treatments may enable the development of novel therapeutic approaches to combat this deadly disorder. In multifunctional, multimodal imaging, and drug delivery carriers, NPs generally play a major role. They have emerged as potential strategies for the invention of innovative therapeutic procedures in the last decade. The exponential growth of nanotechnologies in recent years has increased public awareness of the application of these innovative therapeutic approaches. Many tumor-targeted nanomedicines have been studied in cancer therapy, and there is clear evidence for a significant improvement in the therapeutic index of antineoplastic drugs. Age-related factors such as metabolic and physiological alterations in old age and inadequate animal models are currently understudied in nanomedicine and pharmacology. This review highlighted the most important targeting approaches, as well as public awareness, therapeutic advancements, and future prospects in age-related metabolic variations, and tumor-targeted nanomedicine studies.
Collapse
Affiliation(s)
- Md. Mominur Rahman
- Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, Dhaka 1207, Bangladesh
| | - Fahadul Islam
- Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, Dhaka 1207, Bangladesh
| | - Sadia Afsana Mim
- Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, Dhaka 1207, Bangladesh
| | - Md. Shajib Khan
- Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, Dhaka 1207, Bangladesh
| | - Md. Rezaul Islam
- Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, Dhaka 1207, Bangladesh
| | - Md. Anamul Haque
- Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, Dhaka 1207, Bangladesh
| | - Saikat Mitra
- Department of Pharmacy, Faculty of Pharmacy, University of Dhaka, Dhaka 1000, Bangladesh
| | - Talha Bin Emran
- Department of Pharmacy, BGC Trust University Bangladesh, Chittagong 4381, Bangladesh
| | - Abdur Rauf
- Department of Chemistry, University of Swabi, Anbar, Swabi, Khyber Pakhtunkhwa, Pakistan
| |
Collapse
|
26
|
Abdel-Hakem NE, Talaat RM, Samaka RM, Bassyouniy IH, EL-Shahat M, Alkawareek MY, Alkilany AM. Therapeutic outcomes and biodistribution of gold nanoparticles in collagen-induced arthritis animal model. J Drug Deliv Sci Technol 2022. [DOI: 10.1016/j.jddst.2021.102944] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
|
27
|
Li X, Wang H, Zou X, Su H, Li C. Methotrexate-loaded folic acid of solid-phase synthesis conjugated gold nanoparticles targeted treatment for rheumatoid arthritis. Eur J Pharm Sci 2021; 170:106101. [PMID: 34936935 DOI: 10.1016/j.ejps.2021.106101] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Revised: 12/02/2021] [Accepted: 12/16/2021] [Indexed: 12/14/2022]
Abstract
PURPOSE Methotrexate (MTX) is a first-line drug for rheumatoid arthritis (RA). Targeting of MTX to inflamed joints is essential to the prevention of potential toxicity and improving therapeutic effects. Gold nanoparticles (GNPs) are characterized by controllable particle sizes and good biocompatibilities, therefore, they are promising drug delivery systems. We aimed at developing a GNPs drug delivery system incorporating MTX and folic acid (FA) with strong efficacies against RA. METHODS MTX-Cys-FA was synthesized through solid-phase organic synthesis. Then, it was coupled with sulfhydryl groups in GNPs, thereby successfully preparing a GNPs/MTX-Cys-FA nanoconjugate with targeting properties. Physical and chemical techniques were used to characterize it. Moreover, we conducted its stability, release, pharmacokinetics, biodistribution and cell cytotoxicity, cell uptake, cell migration, as well as its therapeutic effect on CIA rats. The histopathology was conducted to investigate anti-RA effects of GNPs/MTX-Cys-FA nanoconjugates. RESULTS The GNPs/MTX-Cys-FA nanoconjugate exhibited a spherical appearance, had a particle size of 103.06 nm, a zeta potential of -33.68 mV, drug loading capacity of 11.04 %, and an encapsulation efficiency of 73.61%. Cytotoxicity experiments revealed that GNPs had good biocompatibilities while GNPs/MTX-Cys-FA exhibited excellent drug-delivery abilities. Cell uptake and migration experiment showed that nanoconjugates containing FA by LPS activated mouse mononuclear macrophages (RAW264.7) was significantly increased, and they exerted significant inhibitory effects on human fibroblast-like synoviocytes (HFLS) of RA (p<0.01). In addition, the nanoconjugate prolonged blood circulation time of MTX in collagen-induced arthritis (CIA) rats (p<0.01), enhanced MTX accumulation in inflamed joints (p<0.01), enhanced their therapeutic effects (p<0.01), and reduced toxicity to major organs (p<0.01). CONCLUSION GNPs/MTX-Cys-FA nanoconjugates provide effective approaches for RA targeted therapeutic strategies.
Collapse
Affiliation(s)
- Xuena Li
- College of Pharmacy, Yanbian University, No. 977, Gongyuan Road, Yanji 133000, China
| | - Huanhui Wang
- College of Pharmacy, Yanbian University, No. 977, Gongyuan Road, Yanji 133000, China
| | - Xiaotong Zou
- College of Pharmacy, Yanbian University, No. 977, Gongyuan Road, Yanji 133000, China
| | - Hui Su
- Department of Pharmacy, The Sixth Affiliated Hospital of Harbin Medical University, No. 142 road, Zhongyuan Avenue, Harbin 150028, China
| | - Cheng Li
- College of Medicine, Yanbian University, No. 977, Gongyuan Road, Yanji 133000, China; Department of Pharmacy, Affiliated Hospital of Yanbian University, No. 1327, Juzi Street, Yanji 133000, China.
| |
Collapse
|
28
|
Targeting immunosuppressor cells with nanoparticles in autoimmunity: How far have we come to? Cell Immunol 2021; 368:104412. [PMID: 34340162 DOI: 10.1016/j.cellimm.2021.104412] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2021] [Revised: 07/20/2021] [Accepted: 07/23/2021] [Indexed: 12/24/2022]
Abstract
Autoimmunity is the assault of immune response towards self-antigens, resulting to inflammation and tissue injury. It is staged into three phases and caused by malfunction of immune tolerance. In our body, immune tolerance is synchronized by several immunosuppressor cells such as regulatory T cells and B cells as well as myeloid-derived suppressor cells, which are prominently dysregulated in autoimmunity. Hence, targeting these cell populations serve as a significant potential in the therapy of autoimmunity. Nanotechnology with its advantageous properties is shown to be a remarkable tool as drug delivery system in this field. This review focused on the development of therapeutics in autoimmune diseases utilizing various nanoparticles formulation based on two targeting approaches in autoimmunity, passive and active targeting. Lastly, this review outlined the approved present nanomedicines as well as in clinical evaluations and issues regarding the lack of translation of these nanomedicines into the market, despite the abundant of positive experimental observations.
Collapse
|
29
|
Chen L, Wang Y, Sun L, Yan J, Mao H. Nanomedicine Strategies for Anti-Inflammatory Treatment of Noninfectious Arthritis. Adv Healthc Mater 2021; 10:e2001732. [PMID: 33870656 DOI: 10.1002/adhm.202001732] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2020] [Revised: 03/09/2021] [Indexed: 02/06/2023]
Abstract
Noninfectious arthritis (NIA) comprises a class of chronic and progressive inflammatory disorders that require early-stage management to prevent disease progression. The most common forms include osteoarthritis, rheumatoid arthritis, ankylosing spondylitis, and gouty arthritis. Current treatments involve nonsteroidal anti-inflammatory drugs, disease-modifying antirheumatic drugs and glucocorticoids to alleviate clinical symptoms, although regular use of these can result in a high risk of chronic kidney disease and heart failure, as well as severe adverse gastrointestinal effects. Nanomedicine offers unique opportunities to address these challenges and improve therapeutic efficacy due to its ability to deliver therapeutics locally in a sustained manner, thus extending the half-life, improving bioavailability, and reducing the side effects of these agents. This review includes a comprehensive analysis of the mechanisms of various treatment options for NIA and highlights recent progress and emerging strategies in treating NIA with nanomedicine platforms, particularly related to long-term biosafety and nonspecific targeting in designing nanomedicine delivery systems.
Collapse
Affiliation(s)
- Long Chen
- Department of Orthopedics Guizhou Provincial People's Hospital Guiyang Guizhou 550000 China
- Translational Tissue Engineering Center Johns Hopkins School of Medicine Baltimore MD 21287 USA
- Institute for NanoBioTechnology Johns Hopkins University Baltimore MD 21218 USA
| | - Yuanzheng Wang
- Department of Orthopedics Guizhou Provincial People's Hospital Guiyang Guizhou 550000 China
| | - Li Sun
- Department of Orthopedics Guizhou Provincial People's Hospital Guiyang Guizhou 550000 China
| | - Jerry Yan
- Institute for NanoBioTechnology Johns Hopkins University Baltimore MD 21218 USA
- Department of Biomedical Engineering School of Medicine Johns Hopkins University Baltimore MD 21205 USA
| | - Hai‐Quan Mao
- Translational Tissue Engineering Center Johns Hopkins School of Medicine Baltimore MD 21287 USA
- Institute for NanoBioTechnology Johns Hopkins University Baltimore MD 21218 USA
- Department of Biomedical Engineering School of Medicine Johns Hopkins University Baltimore MD 21205 USA
- Department of Materials Science and Engineering Whiting School of Engineering Johns Hopkins University Baltimore MD 21218 USA
| |
Collapse
|
30
|
Hosseinikhah SM, Barani M, Rahdar A, Madry H, Arshad R, Mohammadzadeh V, Cucchiarini M. Nanomaterials for the Diagnosis and Treatment of Inflammatory Arthritis. Int J Mol Sci 2021; 22:3092. [PMID: 33803502 PMCID: PMC8002885 DOI: 10.3390/ijms22063092] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Revised: 03/04/2021] [Accepted: 03/16/2021] [Indexed: 12/12/2022] Open
Abstract
Nanomaterials have received increasing attention due to their unique chemical and physical properties for the treatment of rheumatoid arthritis (RA), the most common complex multifactorial joint-associated autoimmune inflammatory disorder. RA is characterized by an inflammation of the synovium with increased production of proinflammatory cytokines (IL-1, IL-6, IL-8, and IL-10) and by the destruction of the articular cartilage and bone, and it is associated with the development of cardiovascular disorders such as heart attack and stroke. While a number of imaging tools allow for the monitoring and diagnosis of inflammatory arthritis, and despite ongoing work to enhance their sensitivity and precision, the proper assessment of RA remains difficult particularly in the early stages of the disease. Our goal here is to describe the benefits of applying various nanomaterials as next-generation RA imaging and detection tools using contrast agents and nanosensors and as improved drug delivery systems for the effective treatment of the disease.
Collapse
Affiliation(s)
- Seyedeh Maryam Hosseinikhah
- Nanotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad 91886-17871, Iran;
| | - Mahmood Barani
- Department of Chemistry, Shahid Bahonar University of Kerman, Kerman 761691411, Iran;
| | - Abbas Rahdar
- Department of Physics, Faculty of Science, University of Zabol, Zabol 538-9861, Iran
| | - Henning Madry
- Center of Experimental Orthopaedics, Saarland University Medical Center, D-66421 Homburg/Saar, Germany;
| | - Rabia Arshad
- Department of Pharmacy, Quaid-i-Azam University, Islamabad 45320, Pakistan;
| | - Vahideh Mohammadzadeh
- Department of Pharmaceutical Nanotechnology, School of Pharmacy, Mashhad University of Medical Science, Mashhad 91886-17871, Iran;
| | - Magali Cucchiarini
- Center of Experimental Orthopaedics, Saarland University Medical Center, D-66421 Homburg/Saar, Germany;
| |
Collapse
|
31
|
Bilal M, Qindeel M, Nunes LV, Duarte MTS, Ferreira LFR, Soriano RN, Iqbal HMN. Marine-Derived Biologically Active Compounds for the Potential Treatment of Rheumatoid Arthritis. Mar Drugs 2020; 19:10. [PMID: 33383638 PMCID: PMC7823916 DOI: 10.3390/md19010010] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Revised: 12/05/2020] [Accepted: 12/10/2020] [Indexed: 02/05/2023] Open
Abstract
Rheumatoid arthritis (RA) is a chronic, systemic autoimmune disease with a prevalence rate of up to 1% and is significantly considered a common worldwide public health concern. Commercially, several traditional formulations are available to treat RA to some extent. However, these synthetic compounds exert toxicity and considerable side effects even at lower therapeutic concentrations. Considering the above-mentioned critiques, research is underway around the world in finding and exploiting potential alternatives. For instance, marine-derived biologically active compounds have gained much interest and are thus being extensively utilized to confront the confines of in practice counterparts, which have become ineffective for 21st-century medical settings. The utilization of naturally available bioactive compounds and their derivatives can minimize these synthetic compounds' problems to treat RA. Several marine-derived compounds exhibit anti-inflammatory and antioxidant properties and can be effectively used for therapeutic purposes against RA. The results of several studies ensured that the extraction of biologically active compounds from marine sources could provide a new and safe source for drug development against RA. Finally, current challenges, gaps, and future perspectives have been included in this review.
Collapse
Affiliation(s)
- Muhammad Bilal
- School of Life Science and Food Engineering, Huaiyin Institute of Technology, Huaian 223003, China
| | - Maimoona Qindeel
- Department of Pharmacy, Quaid-i-Azam University, Islamabad 45320, Pakistan;
| | - Leonardo Vieira Nunes
- Department of Medicine, Federal University of Juiz de Fora, Juiz de Fora-MG 36036-900, Brazil;
| | | | - Luiz Fernando Romanholo Ferreira
- Graduate Program in Process Engineering, Tiradentes University (UNIT), Av. Murilo Dantas, 300, Farolândia, Aracaju-Sergipe 49032-490, Brazil;
- Institute of Technology and Research (ITP), Tiradentes University (UNIT), Av. Murilo Dantas, 300, Farolândia, Aracaju-Sergipe 49032-490, Brazil
| | - Renato Nery Soriano
- Division of Physiology and Biophysics, Department of Basic Life Sciences, Federal University of Juiz de Fora, Governador Valadares-MG 35010-180, Brazil;
| | - Hafiz M. N. Iqbal
- School of Engineering and Sciences, Tecnologico de Monterrey, Monterrey 64849, Mexico
| |
Collapse
|