1
|
Xue Z, Li N, Du K, Shu J, Huang Z, Gao Z, Xie X, Li Q, Lu Y. Inhibiting synovial inflammation and promoting cartilage repair in rheumatoid arthritis using a matrix metalloproteinase-binding hydrogel. Mater Today Bio 2025; 32:101792. [PMID: 40343163 PMCID: PMC12059346 DOI: 10.1016/j.mtbio.2025.101792] [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: 02/25/2025] [Revised: 04/11/2025] [Accepted: 04/22/2025] [Indexed: 05/11/2025] Open
Abstract
Originating from synovial tissue, matrix metalloproteinase-9 (MMP-9) is a key inflammatory factor that promotes the formation and invasion of synovial pannus, leading to cartilage matrix destruction in rheumatoid arthritis (RA). However, clinical trials of systemic use of MMP-9 inhibitors are not successful due to severe side effects. Thus, locally inhibiting MMP-9 may be an alternative in the treatment of RA. Herein, we developed MMP-9 binding peptide-functionalized copper sulfide nanoparticles (CuS-T NPs) and delivered them with light crosslinking chondroitin sulfate methacrylate (ChSMA) hydrogel. We found that the CuS NP-doped hydrogels could inhibit synovial inflammation. Specifically, the CuS-T/ChSMA hydrogel could rapidly bind to MMP-9, thereby inhibiting not only the invasion of RA fibroblast-like synoviocytes but also the polarization of inflammatory M1-type macrophages. The underlying mechanism involved the inhibition of the MAPK pathway. Moreover, ChSMA hydrogel provided a cartilage matrix-mimic microenvironment and synergistically promoted the generation of collagen-2 and aggrecan with CuS NPs. In an adjuvant-induced arthritis mouse model, the intra-articular injection of ChSMA/CuS-T hydrogel significantly alleviated synovial inflammation and accelerated cartilage repair without causing any side effects, killing two birds with one stone in RA therapy.
Collapse
Affiliation(s)
- Zhanpeng Xue
- Department of Joint and Orthopedics, Orthopedic Center, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, 510282, China
| | - Nan Li
- Department of Joint and Orthopedics, Orthopedic Center, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, 510282, China
| | - Kaijun Du
- Department of Joint and Orthopedics, Orthopedic Center, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, 510282, China
| | - Jianxiong Shu
- Department of Joint and Orthopedics, Orthopedic Center, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, 510282, China
| | - Zhenwen Huang
- Department of Joint and Orthopedics, Orthopedic Center, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, 510282, China
| | - Zhifei Gao
- The Second School of Clinical Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, 510282, China
| | - Xiaobo Xie
- Department of Joint and Orthopedics, Orthopedic Center, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, 510282, China
| | - Qi Li
- Department of Joint and Orthopedics, Orthopedic Center, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, 510282, China
| | - Yao Lu
- Department of Joint and Orthopedics, Orthopedic Center, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, 510282, China
- The Second School of Clinical Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, 510282, China
- State Key Laboratory of Multi-organ Injury Prevention and Treatment, Southern Medical University, Guangzhou, Guangdong, 510515, China
| |
Collapse
|
2
|
Okasha RM, Mohamed AAH, Elhenawy AA, Alsehli MH, Alsaedi WH, Alblewi FF, Assirey EA, Alawad MO, Aljohani FS, Hrytani Z, Ibrahim I, Afifi TH. A novel avenue in the successful synthesis of Schiff base macromolecules via innovative plasma and classical approaches. Sci Rep 2025; 15:10840. [PMID: 40155415 PMCID: PMC11953434 DOI: 10.1038/s41598-025-94665-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2025] [Accepted: 03/17/2025] [Indexed: 04/01/2025] Open
Abstract
Schiff base macromolecules have been successfully synthesized, utilizing a classical chemistry route and a dielectric barrier discharge (DBD) plasma technique. The synthesis of monomeric units has been accomplished through typical reactions of aldehyde and amine functional molecules. The condensation polymerization of the Schiff base molecules has been instigated chemically, using p-toluene sulfonyl chloride in refluxed ethanol. The molecular weight of the obtained polymers was discovered to be 524, 664 and 1,503,228 for Schiff base polymers 4a and 4b, respectively. Additionally, the polymerization reactions were prompted, employing a Dielectric Barrier Discharge (DBD) atmospheric pressure air plasma technique. The DBD plasma demonstrated a very powerful routine to produce high molecular weights macromolecules with optimum condition at 5 min. duration time, which could be an ecofriendly strategy to acquire this class of materials. The new polymeric materials have been characterized utilizing FTIR and NMR spectroscopy. Moreover, the complexation of polymer 4b with various metal moieties, Ru (II), Co (II), Cu (II), and Ni (II), has been executed in order to have a comparative study of their antitumor activity against MCF-7, HCT-116, and HepG-2 cell lines. Furthermore, the density functional theory was exploited to optimize the polymers and their complexes, and their HOMO-LUMO and energy gap were calculated, which was utilized to examine the inter/intra molecular charge transfer. The molecular electrostatic potential map was similarly quantified to investigate the reactive sites that are present in the investigated molecules. The result for the docking study confirmed that these complexed polymers adopted numerous important interactions with the amino acid of the targeted enzyme.
Collapse
Affiliation(s)
- Rawda M Okasha
- Chemistry Department, College of Science, Taibah University, Medinah, 41411, Saudi Arabia.
| | - Abdel-Aleam H Mohamed
- Physics Department, College of Science, Taibah University, Medina, 30002, Saudi Arabia
- Physics Department, Faculty of Science, Beni-Suef University, Beni-Suef 62511, Egypt
| | - Ahmed A Elhenawy
- Chemistry Department, Faculty of Science, Al-Azhar University, Nasr City, 11884, Cairo, Egypt
- Chemistry Department, Faculty of Science and Art, AlBaha University, Mukhwah, 65731, Al Bahah, Saudi Arabia
| | - Mosa H Alsehli
- Chemistry Department, College of Science, Taibah University, Medinah, 41411, Saudi Arabia
| | - Wael H Alsaedi
- Chemistry Department, College of Science, Taibah University, Medinah, 41411, Saudi Arabia
| | - Fawzia F Alblewi
- Chemistry Department, College of Science, Taibah University, Medinah, 41411, Saudi Arabia
| | - Eman A Assirey
- Chemistry Department, College of Science, Taibah University, Medinah, 41411, Saudi Arabia
| | - Majed O Alawad
- Center of Excellence for Nanomaterials for Clean energy Applications, King Abdulaziz City for Science and Technology (KACST), Riyadh, 12354, Saudi Arabia
| | - Faizah S Aljohani
- Chemistry Department, College of Science, Taibah University, Medinah, 41411, Saudi Arabia
| | - Zainab Hrytani
- Chemistry Department, College of Science, Taibah University, Medinah, 41411, Saudi Arabia
| | - Israa Ibrahim
- Department of basic science, The applied college, Taibah University, Medina, Saudi Arabia
| | - Tarek H Afifi
- Chemistry Department, College of Science, Taibah University, Medinah, 41411, Saudi Arabia.
- Chemistry Department, Faculty of Science, Al-Azhar University, Nasr City, 11884, Cairo, Egypt.
| |
Collapse
|
3
|
Wang S, Li J, Ren F, Zhang J, Song W, Ren L. New Dawn in the Treatment of Rheumatoid Arthritis: Advanced Insight into Polymer Hydrogel Research. Gels 2025; 11:136. [PMID: 39996679 PMCID: PMC11855332 DOI: 10.3390/gels11020136] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2025] [Revised: 02/07/2025] [Accepted: 02/13/2025] [Indexed: 02/26/2025] Open
Abstract
As a chronic systemic autoimmune disease, rheumatoid arthritis (RA) not only damages joints and other organs or systems throughout the body but also torments patients' physical and mental health for a long time, seriously affecting their quality of life. According to incomplete statistics at present, the global prevalence of RA is approximately 0.5-1%, and the number of patients is increasing year by year. Currently, drug therapies are usually adopted for the treatment of RA, such as non-steroidal anti-inflammatory drugs (NSAIDs), disease-modifying antirheumatic drugs (DMARDs), glucocorticoids/steroids, and so on. However, traditional drug therapy has problems such as long half-lives, long treatment cycles requiring frequent drug administration, lack of specificity, and other possible adverse reactions (such as gastrointestinal side effects, skin stratum corneum barrier damage, and systemic toxicity), which greatly restrict the treatment of RA. In order to improve the limitations of traditional drug, physical, and surgical treatments for RA, a large number of related studies on the treatment of RA have been carried out. Among them, hydrogels have been widely used in the research on the treatment of RA due to their excellent biocompatibility, mechanical properties, and general adaptability. For example, hydrogels can be injected into the synovial cavity of joints as synovial fluid to reduce wear between joints, lubricate joints, and avoid synovial surface degradation. This article reviews the applications of hydrogels in the treatment of RA under different functions and the situation of hydrogels as carriers in the treatment of RA through different drug delivery routes and confirms the outstanding potential of hydrogels as drug carriers in the treatment of RA, which has great research significance.
Collapse
Affiliation(s)
- Shuai Wang
- Key Laboratory of Bionic Engineering (Ministry of Education), College of Biological and Agricultural Engineering, Jilin University, Changchun 130022, China;
| | - Jinyang Li
- College of Biological and Agricultural Engineering, Jilin University, Changchun 130012, China;
| | - Fazhan Ren
- School of Agricultural Engineering and Food Science, Shandong University of Technology, Zibo 255000, China;
| | - Jiale Zhang
- College of Engineering, Northeast Agricultural University, Harbin 150030, China;
| | - Wei Song
- College of Engineering and Technology, Jilin Agricultural University, Changchun 130118, China;
| | - Lili Ren
- Key Laboratory of Bionic Engineering (Ministry of Education), College of Biological and Agricultural Engineering, Jilin University, Changchun 130022, China;
- National Key Laboratory of Automotive Chassis Integration and Bionics, Jilin University, Changchun 130022, China
| |
Collapse
|
4
|
Zhang L, Zheng G, Zhao W, He C, Huang Z. Coixol-Loaded Hydrogels Promote Osteochondral Defect Repair via Modulation of Ferroptosis and Autophagy in Chondrocytes. ACS Biomater Sci Eng 2025; 11:1096-1105. [PMID: 39818713 DOI: 10.1021/acsbiomaterials.4c01980] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2025]
Abstract
Osteoarthritis (OA) is a chronic multifactorial disease characterized by cartilage degeneration, pain, and reduced mobility. Current therapies primarily aim to relieve pain and restore function, but they often have limited effectiveness and side effects. Coixol, a bioactive compound from Coix lacryma-jobi L., exhibits anti-inflammatory and analgesic properties, suggesting potential benefits in OA treatment. This study explored the effects of coixol on OA chondrocytes. Primary chondrocytes from OA rats were isolated and treated with varying concentrations of coixol. Cell viability and proliferation were assessed by using CCK-8 assays. The expression of genes related to ferroptosis and autophagy was analyzed through RT-qPCR, Western blot, and immunofluorescence. Moreover, the study investigated the characteristics and performance of coixol-loaded PDLLA-PEG-PDLLA (PLEL)/gelatin sponge (GS) hydrogels (Coixol@PLEL/GS) for enhancing osteochondral defect repair by specifically targeting chondrocyte ferroptosis and autophagy. The characteristics of coixol-loaded PDLLA-PEG-PDLLA/gelatin sponge (Coixol@PLEL/GS) hydrogels were evaluated using cryo-scanning electron microscopy (SEM) or SEM, and coixol release kinetics were determined. In vivo, a rat osteochondral defect model was used to assess the efficacy of Coixol@PLEL/GS in osteochondral defect repair using International Cartilage Repair Society (ICRS) scores, Safranin O/Fast green staining, Toluidine blue staining, and immunofluorescence. Coixol significantly increased the viability and proliferation of OA chondrocytes in a dose-dependent manner. Furthermore, coixol inhibited ferroptosis and stimulated autophagy, as evidenced by the upregulation of related genes. In vivo, Coixol@PLEL/GS remarkably enhanced the repair of osteochondral defects compared to that of control groups. In conclusion, coixol protects OA chondrocytes by improving survival, inhibiting ferroptosis, and activating autophagy, highlighting its potential as a therapeutic strategy for OA treatment.
Collapse
Affiliation(s)
- Liqin Zhang
- The Affiliated Ganzhou Hospital of Nanchang University, Meiguan Avenue No. 16, Ganzhou 341000, China
| | - Guangping Zheng
- Ganzhou Municipal Key Laboratory of Bone and Joint Research, The Affiliated Ganzhou Hospital of Nanchang University, Meiguan Avenue No. 16, Ganzhou 341000, China
| | - Weicheng Zhao
- The Affiliated Ganzhou Hospital of Nanchang University, Meiguan Avenue No. 16, Ganzhou 341000, China
- Ganzhou Municipal Key Laboratory of Bone and Joint Research, The Affiliated Ganzhou Hospital of Nanchang University, Meiguan Avenue No. 16, Ganzhou 341000, China
| | - Chun He
- The Affiliated Ganzhou Hospital of Nanchang University, Meiguan Avenue No. 16, Ganzhou 341000, China
| | - Zhongming Huang
- The Affiliated Ganzhou Hospital of Nanchang University, Meiguan Avenue No. 16, Ganzhou 341000, China
- Ganzhou Municipal Key Laboratory of Bone and Joint Research, The Affiliated Ganzhou Hospital of Nanchang University, Meiguan Avenue No. 16, Ganzhou 341000, China
| |
Collapse
|
5
|
Bi J, Zeng J, Liu X, Mo C, Yao M, Zhang J, Yuan P, Jia B, Xu S. Drug delivery for age-related bone diseases: From therapeutic targets to common and emerging therapeutic strategies. Saudi Pharm J 2024; 32:102209. [PMID: 39697472 PMCID: PMC11653637 DOI: 10.1016/j.jsps.2024.102209] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2024] [Accepted: 11/22/2024] [Indexed: 12/20/2024] Open
Abstract
With the accumulation of knowledge on aging, people have gradually realized that among the many factors that cause individual aging, the accumulation of aging cells is an essential cause of organ degeneration and, ultimately, age-related diseases. Most cells present in the bone microenvironment gradually age over time, leading to an imbalance of osteogenesis, osteoclastogenesis, adipogenesis, and chondrogenesis. This imbalance contributes to age-related bone loss and the development of age-related bone diseases, such as osteoporosis. Bone aging can prolong the lifespan and delay the development of age-related diseases. Nanoparticles have controllable and stable physical and chemical properties and can precisely target different tissues and organs. By preparing multiple easily modified and biocompatible nanoparticles as different drug delivery carriers, specifically targeting various diseased tissues for controlled-release and sustained-release administration, the delivery efficiency of drugs can be significantly improved, and the toxicity and side effects of drugs can be substantially reduced, thereby improving the therapeutic effect of age-related bone diseases. In addition, other novel anti-aging strategies (such as stem cell exosomes) also have significant scientific and practical significance in anti-aging research on age-related bone diseases. This article reviews the research progress of various nano-drug-loaded particles and emerging anti-aging methods for treating age-related bone diseases, offering new insights and directions for precise targeted clinical therapies.
Collapse
Affiliation(s)
- Jiaming Bi
- Department of Endodontics, Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, Guangdong, China
| | - Jiawei Zeng
- Department of Endodontics, Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, Guangdong, China
| | - Xiaohao Liu
- Department of Periodontics, Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, Guangdong, China
| | - Chuzi Mo
- Department of Endodontics, Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, Guangdong, China
| | - Mingyan Yao
- Department of Endocrinology, Baoding No.1 Central Hospital, Baoding, China
| | - Jing Zhang
- Department of Cardiology, Affiliated Hospital of Hebei University, Baoding, China
| | - Peiyan Yuan
- Department of Endodontics, Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, Guangdong, China
| | - Bo Jia
- Department of Oral and Maxillofacial Surgery, Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, Guangdong, China
| | - Shuaimei Xu
- Department of Endodontics, Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, Guangdong, China
| |
Collapse
|
6
|
Ochoa-Sánchez C, Rodríguez-León E, Iñiguez-Palomares R, Rodríguez-Beas C. Brief Comparison of the Efficacy of Cationic and Anionic Liposomes as Nonviral Delivery Systems. ACS OMEGA 2024; 9:46664-46678. [PMID: 39619565 PMCID: PMC11603276 DOI: 10.1021/acsomega.4c06714] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/20/2024] [Revised: 10/28/2024] [Accepted: 11/05/2024] [Indexed: 01/05/2025]
Abstract
In recent decades, the development and application of nonviral vectors, such as liposomes and lipidic nanoparticles, for gene therapy and drug delivery have seen substantial progress. The interest in the physicochemical properties and structures of the complexes liposome/DNA and liposome/RNA is due to their potential to substitute viruses as carriers of drugs or genetic material into cells with minimal cytotoxicity, which could lead to their use in gene therapy. Initially, cationic liposomes were utilized as nonviral DNA delivery vectors; subsequently, different molecules, such as polymers, were incorporated to enhance transfection efficiency. Additionally, liposome/protein complexes have been developed as nonviral vectors for the treatment of diseases. The most relevant internalization pathways of these vectors and the few transfection results obtained using targeted and nontargeted liposomes are discussed below. The high cytotoxicity of cationic liposomes represents a significant challenge for the development of gene therapy and drug delivery. Anionic liposomes offer a promising alternative to address the limitations of conventional cationic liposomes, including immune response, short circulation time, and low toxicity. This review will discuss the advantages of cationic liposomes and the novel anionic liposome-based systems that have emerged as a result. The advent of novel designs and manufacturing techniques has facilitated the development of innovative systems, designated as lipid nanoparticles (LNPs), which serve as highly efficacious regulators of the immune system.
Collapse
Affiliation(s)
- Carlos Ochoa-Sánchez
- Physics Department, Universidad de Sonora. Rosales and Luis Encinas 8300, Hermosillo, Sonora 83000, México
| | - Ericka Rodríguez-León
- Physics Department, Universidad de Sonora. Rosales and Luis Encinas 8300, Hermosillo, Sonora 83000, México
| | - Ramón Iñiguez-Palomares
- Physics Department, Universidad de Sonora. Rosales and Luis Encinas 8300, Hermosillo, Sonora 83000, México
| | - César Rodríguez-Beas
- Physics Department, Universidad de Sonora. Rosales and Luis Encinas 8300, Hermosillo, Sonora 83000, México
| |
Collapse
|
7
|
Ye Q, Zhang M, Li S, Liu W, Xu C, Li Y, Xie R. Controlled Stimulus-Responsive Delivery Systems for Osteoarthritis Treatment. Int J Mol Sci 2024; 25:11799. [PMID: 39519350 PMCID: PMC11545989 DOI: 10.3390/ijms252111799] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2024] [Revised: 10/30/2024] [Accepted: 10/31/2024] [Indexed: 11/16/2024] Open
Abstract
Osteoarthritis (OA), a common and disabling degenerative joint disease, affects millions of people worldwide and imposes a considerable burden on patients and society due to its high prevalence and economic costs. The pathogenesis of OA is closely related to the progressive degradation of articular cartilage and the accompany inflammation; however, articular cartilage itself cannot heal and modulate the inflammation due to the lack of nerves, blood vessels, and lymph-vessels. Therefore, reliable and effective methods to treat OA remain highly desired. Local administration of drugs or bioactive materials by intra-articular injection of the delivery system represents a promising approach to treat OA, especially considering the prolonged joint retention, cartilage or chondrocytes targeting, and stimuli-responsive release to achieve precision OA therapy. This article summarizes and discusses the advances in the currently used delivery systems (nanoparticle, hydrogel, liposome, and microsphere) and then focuses on their applications in OA treatment from the perspective of endogenous stimulus (redox reactions, pH, enzymes, and temperature) and exogenous stimulus (near-infrared, magnetic, and ultrasound)-responsive release. Finally, the challenges and potential future directions for the development of nano-delivery systems are summarized.
Collapse
Affiliation(s)
- Qianwen Ye
- School of Medical Information Engineering, Gannan Medical University, Ganzhou 341000, China; (Q.Y.); (M.Z.); (S.L.); (W.L.)
- Jiangxi Provincial Key Laboratory of Tissue Engineering (2024SSY06291), Gannan Medical University, Ganzhou 341000, China;
| | - Mingshuo Zhang
- School of Medical Information Engineering, Gannan Medical University, Ganzhou 341000, China; (Q.Y.); (M.Z.); (S.L.); (W.L.)
- Jiangxi Provincial Key Laboratory of Tissue Engineering (2024SSY06291), Gannan Medical University, Ganzhou 341000, China;
| | - Shuyue Li
- School of Medical Information Engineering, Gannan Medical University, Ganzhou 341000, China; (Q.Y.); (M.Z.); (S.L.); (W.L.)
- Jiangxi Provincial Key Laboratory of Tissue Engineering (2024SSY06291), Gannan Medical University, Ganzhou 341000, China;
| | - Wenyue Liu
- School of Medical Information Engineering, Gannan Medical University, Ganzhou 341000, China; (Q.Y.); (M.Z.); (S.L.); (W.L.)
- Jiangxi Provincial Key Laboratory of Tissue Engineering (2024SSY06291), Gannan Medical University, Ganzhou 341000, China;
| | - Chunming Xu
- Jiangxi Provincial Key Laboratory of Tissue Engineering (2024SSY06291), Gannan Medical University, Ganzhou 341000, China;
- School of Basic Medicine, Gannan Medical University, Ganzhou 341000, China
| | - Yumei Li
- Jiangxi Provincial Key Laboratory of Tissue Engineering (2024SSY06291), Gannan Medical University, Ganzhou 341000, China;
- School of Basic Medicine, Gannan Medical University, Ganzhou 341000, China
| | - Renjian Xie
- School of Medical Information Engineering, Gannan Medical University, Ganzhou 341000, China; (Q.Y.); (M.Z.); (S.L.); (W.L.)
- Jiangxi Provincial Key Laboratory of Tissue Engineering (2024SSY06291), Gannan Medical University, Ganzhou 341000, China;
- Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases (Ministry of Education), Gannan Medical University, Ganzhou 341000, China
| |
Collapse
|
8
|
Shi TM, Chen XF, Ti H. Ferroptosis-Based Therapeutic Strategies toward Precision Medicine for Cancer. J Med Chem 2024; 67:2238-2263. [PMID: 38306267 DOI: 10.1021/acs.jmedchem.3c01749] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2024]
Abstract
Ferroptosis is a type of iron-dependent programmed cell death characterized by the dysregulation of iron metabolism and the accumulation of lipid peroxides. This nonapoptotic mode of cell death is implicated in various physiological and pathological processes. Recent findings have underscored its potential as an innovative strategy for cancer treatment, particularly against recalcitrant malignancies that are resistant to conventional therapies. This article focuses on ferroptosis-based therapeutic strategies for precision cancer treatment, covering the molecular mechanisms of ferroptosis, four major types of ferroptosis inducers and their inhibitory effects on diverse carcinomas, the detection of ferroptosis by fluorescent probes, and their implementation in image-guided therapy. These state-of-the-art tactics have manifested enhanced selectivity and efficacy against malignant carcinomas. Given that the administration of ferroptosis in cancer therapy is still at a burgeoning stage, some major challenges and future perspectives are discussed for the clinical translation of ferroptosis into precision cancer treatment.
Collapse
Affiliation(s)
- Tong-Mei Shi
- School of Chinese Materia Medica, Guangdong Pharmaceutical University, Guangzhou 510006, P. R. China
| | - Xiao-Fei Chen
- Guangdong Provincial Key Laboratory of Chemical Measurement and Emergency Test Technology, Institute of Analysis, Guangdong Academy of Sciences, China National Analytical Center, Guangzhou, Guangzhou 510070, P. R. China
| | - Huihui Ti
- School of Chinese Materia Medica, Guangdong Pharmaceutical University, Guangzhou 510006, P. R. China
- Guangdong Province Precise Medicine Big Data of Traditional Chinese Medicine Engineering Technology Research Center, Guangdong Pharmaceutical University, Guangzhou 510006, P. R. China
| |
Collapse
|
9
|
Alghamdi R, Pertusati F, Prokopovich P. Poly-beta-amino-ester licofelone conjugates development for osteoarthritis treatment. RSC Adv 2024; 14:15-28. [PMID: 38173598 PMCID: PMC10758810 DOI: 10.1039/d3ra04967a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2023] [Accepted: 11/16/2023] [Indexed: 01/05/2024] Open
Abstract
Disease-modifying osteoarthritis drugs (DMOADs) are a new therapeutic class for osteoarthritis (OA) prevention or inhibition of the disease development. Unfortunately, none of the DMOADs have been clinically approved due to their poor therapeutic performances in clinical trials. The joint environment has played a role in this process by limiting the amount of drug effectively delivered as well as the time that the drug stays within the joint space. The current study aimed to improve the delivery of the DMOADs into cartilage tissue by increasing uptake and retention time of the DMOADs within the tissue. Licofelone was used a model DMOAD due to its significant therapeutic effect against OA progression as shown in the recent phase III clinical trial. For this purpose licofelone was covalently conjugated to the two different A16 and A87 poly-beta-amino-ester (PBAEs) polymers taking advantage of their hydrolysable, cytocompatible, and cationic nature. We have shown cartilage uptake of the licofelone-PBAE conjugates increased 18 times and retention in tissues was prolonged by 37 times compared to the equivalent dose of the free licofelone. Additionally, these licofelone conjugates showed no detrimental effect on the chondrocyte viability. In conclusion, the cationic A87 and A16 PBAE polymers increased the amount of licofelone within the cartilage, which could potentially enhance the therapeutic effect and pharmacokinetic performance of this drug and other DMOADs clinically.
Collapse
Affiliation(s)
- Raed Alghamdi
- School of Pharmacy and Pharmaceutical Sciences, Cardiff University Redwood Building, King Edward VII Avenue Cardiff Wales CF10 3NB UK
| | - Fabrizio Pertusati
- School of Pharmacy and Pharmaceutical Sciences, Cardiff University Redwood Building, King Edward VII Avenue Cardiff Wales CF10 3NB UK
| | - Polina Prokopovich
- School of Pharmacy and Pharmaceutical Sciences, Cardiff University Redwood Building, King Edward VII Avenue Cardiff Wales CF10 3NB UK
| |
Collapse
|
10
|
Kamankesh M, Yadegar A, Llopis-Lorente A, Liu C, Haririan I, Aghdaei HA, Shokrgozar MA, Zali MR, Miri AH, Rad-Malekshahi M, Hamblin MR, Wacker MG. Future Nanotechnology-Based Strategies for Improved Management of Helicobacter pylori Infection. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2302532. [PMID: 37697021 DOI: 10.1002/smll.202302532] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Revised: 07/25/2023] [Indexed: 09/13/2023]
Abstract
Helicobacter pylori (H. pylori) is a recalcitrant pathogen, which can cause gastric disorders. During the past decades, polypharmacy-based regimens, such as triple and quadruple therapies have been widely used against H. pylori. However, polyantibiotic therapies can disturb the host gastric/gut microbiota and lead to antibiotic resistance. Thus, simpler but more effective approaches should be developed. Here, some recent advances in nanostructured drug delivery systems to treat H. pylori infection are summarized. Also, for the first time, a drug release paradigm is proposed to prevent H. pylori antibiotic resistance along with an IVIVC model in order to connect the drug release profile with a reduction in bacterial colony counts. Then, local delivery systems including mucoadhesive, mucopenetrating, and cytoadhesive nanobiomaterials are discussed in the battle against H. pylori infection. Afterward, engineered delivery platforms including polymer-coated nanoemulsions and polymer-coated nanoliposomes are poposed. These bioinspired platforms can contain an antimicrobial agent enclosed within smart multifunctional nanoformulations. These bioplatforms can prevent the development of antibiotic resistance, as well as specifically killing H. pylori with no or only slight negative effects on the host gastrointestinal microbiota. Finally, the essential checkpoints that should be passed to confirm the potential effectiveness of anti-H. pylori nanosystems are discussed.
Collapse
Affiliation(s)
- Mojtaba Kamankesh
- Polymer Chemistry Department, School of Science, University of Tehran, PO Box 14155-6455, Tehran, 14144-6455, Iran
| | - Abbas Yadegar
- Foodborne and Waterborne Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, 1985717411, Iran
| | - Antoni Llopis-Lorente
- Instituto Interuniversitario de Investigación de Reconocimiento Molecular y Desarrollo Tecnológico (IDM), Universitat Politècnica de València, Universitat de València, CIBER de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Insituto de Salud Carlos III, Valencia, 46022, Spain
| | - Chenguang Liu
- College of Marine Life Science, Ocean University of China, Qingdao, 266003, P.R. China
| | - Ismaeil Haririan
- Department of Pharmaceutical Biomaterials and Medical Biomaterials Research Center, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, 1417614411, Iran
| | - Hamid Asadzadeh Aghdaei
- Basic and Molecular Epidemiology of Gastrointestinal Disorders Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, 1985717411, Iran
| | | | - Mohammad Reza Zali
- Gastroenterology and Liver Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, 1985717411, Iran
| | - Amir Hossein Miri
- Department of Pharmaceutical Biomaterials and Medical Biomaterials Research Center, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, 1417614411, Iran
| | - Mazda Rad-Malekshahi
- Department of Pharmaceutical Biomaterials and Medical Biomaterials Research Center, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, 1417614411, Iran
| | - Michael R Hamblin
- Laser Research Centre, Faculty of Health Science, University of Johannesburg, Doornfontein, 2028, South Africa
| | - Matthias G Wacker
- Department of Pharmacy, Faculty of Science, National University of Singapore, 4 Science Drive 2, Singapore, 117545, Singapore
| |
Collapse
|
11
|
Liu L, Tang H, Wang Y. Polymeric biomaterials: Advanced drug delivery systems in osteoarthritis treatment. Heliyon 2023; 9:e21544. [PMID: 38034809 PMCID: PMC10682535 DOI: 10.1016/j.heliyon.2023.e21544] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Revised: 10/23/2023] [Accepted: 10/23/2023] [Indexed: 12/02/2023] Open
Abstract
Polymeric biomaterials have emerged as a highly promising candidate for drug delivery systems (DDS), exhibiting significant potential to enhance the therapeutic landscape of osteoarthritis (OA) therapy. Their remarkable capacity to manifest desirable physicochemical attributes, coupled with their excellent biocompatibility and biodegradability, has greatly expanded their utility in pharmacotherapeutic applications. Nevertheless, an urgent necessity exists for a comprehensive synthesis of the most recent advances in polymeric DDS, providing valuable guidance for their implementation in the context of OA therapy. This review is dedicated to summarizing and examining recent developments in the utilization of polymeric DDS for OA therapy. Initially, we present an overview of the intricate pathophysiology characterizing OA and underscore the prevailing limitations inherent to current treatment modalities. Subsequently, we introduce diverse categories of polymeric DDS, including hydrogels, nanofibers, and microspheres, elucidating their inherent advantages and limitations. Moreover, we discuss and summarize the delivery of bioactive agents through polymeric biomaterials for OA therapy, emphasizing key findings and emerging trends. Finally, we highlight prospective directions for advancing polymeric DDS, offering a promising approach to enhance their translational potential for OA therapy.
Collapse
Affiliation(s)
- Lin Liu
- Department of Emergency, Honghui Hospital of Xi'an Jiaotong University, Xi'an City, Shaanxi Province, 710054, China
| | - Haifeng Tang
- Department of Emergency, Honghui Hospital of Xi'an Jiaotong University, Xi'an City, Shaanxi Province, 710054, China
| | - Yanjun Wang
- Department of Emergency, Honghui Hospital of Xi'an Jiaotong University, Xi'an City, Shaanxi Province, 710054, China
| |
Collapse
|
12
|
Zhang X, Zhang M, Huang S, Ohtani K, Xu L, Guo Y. Engineered Polymeric Nanovector for Intracellular Peptide Delivery in Antitumor Therapy. Int J Nanomedicine 2023; 18:5343-5363. [PMID: 37746048 PMCID: PMC10517702 DOI: 10.2147/ijn.s427536] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Accepted: 09/13/2023] [Indexed: 09/26/2023] Open
Abstract
Objective This study aimed to deliver a polypeptide from the Bax-BH3 domain (BHP) through the synthesis of self-assembled amphiphile nanovectors (NVs) and to assess their potential for cancer therapeutic applications and biological safety in vitro and in vivo. These findings provide valuable options for cancer intervention and a novel approach for the rational design of therapeutics. Methods We studied the antitumor activity of BHP by preparing RGDfK-PHPMA-b-Poly (MMA-alt-(Rhob-MA)) (RPPMMRA) and encapsulating it in BHP-NV. We also performed a series of characterizations and property analyses of RPPMMRA, including its size, stability, and drug-carrying capacity. The biocompatibility of RPPMMRA was evaluated in terms of cytotoxicity and hemolytic effects. The pro-apoptotic capacity of BHP was evaluated in vitro using mitochondrial membrane potential, flow cytometry, and apoptosis visualization techniques. The potential therapeutic effects of BHP on tumors were explored using reverse molecular docking. We also investigated the in vivo proapoptotic effect of BHP-NV in a nude mouse tumor model. Results NVs were successfully prepared with hydrated particle sizes ranging from 189.6 nm to 256.6 nm, spherical overall, and were able to remain stable in different media for 72 h with drug loading up to 15.2%. The NVs were be successfully internalized within 6 h with good biocompatibility. Neither BHP nor NV showed significant toxicity when administered alone, however, BHP-NV demonstrated significant side effects in vitro and in vivo. The apoptosis rate increased significantly from 14.13% to 66.34%. Experiments in vivo showed that BHP-NV exhibited significant apoptotic and tumor-suppressive effects. Conclusion A targeted fluorescent NV with high drug delivery efficiency and sustained release protected the active center of BHP, constituting BHP-NV for targeted delivery. RPPMMRA demonstrated excellent biocompatibility, stability, and drug loading ability, whereas and BHP-NV demonstrated potent antitumor effects in vivo and in vitro.
Collapse
Affiliation(s)
- Xi Zhang
- Key Laboratory for Molecular Enzymology and Engineering, the Ministry of Education, National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun, 130012, People’s Republic of China
| | - Mingming Zhang
- Key Laboratory for Molecular Enzymology and Engineering, the Ministry of Education, National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun, 130012, People’s Republic of China
| | - Sijun Huang
- Key Laboratory for Molecular Enzymology and Engineering, the Ministry of Education, National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun, 130012, People’s Republic of China
| | - Kiyoshi Ohtani
- Department of Biomedical Sciences, School of Biological and Environmental Sciences, Kwansei Gakuin University, Sanda, Hyogo, 669-1337, Japan
| | - Li Xu
- Key Laboratory for Molecular Enzymology and Engineering, the Ministry of Education, National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun, 130012, People’s Republic of China
| | - Yi Guo
- Key Laboratory for Molecular Enzymology and Engineering, the Ministry of Education, National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun, 130012, People’s Republic of China
| |
Collapse
|
13
|
Deshmukh R. Rheumatoid arthritis: Pathophysiology, current therapeutic strategies and recent advances in targeted drug delivery system. MATERIALS TODAY COMMUNICATIONS 2023; 35:105877. [DOI: 10.1016/j.mtcomm.2023.105877] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
|
14
|
Li S, Cao P, Chen T, Ding C. Latest insights in disease-modifying osteoarthritis drugs development. Ther Adv Musculoskelet Dis 2023; 15:1759720X231169839. [PMID: 37197024 PMCID: PMC10184265 DOI: 10.1177/1759720x231169839] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Accepted: 03/29/2023] [Indexed: 05/19/2023] Open
Abstract
Osteoarthritis (OA) is a prevalent and severely debilitating disease with an unmet medical need. In order to alleviate OA symptoms or prevent structural progression of OA, new drugs, particularly disease-modifying osteoarthritis drugs (DMOADs), are required. Several drugs have been reported to attenuate cartilage loss or reduce subchondral bone lesions in OA and thus potentially be DMOADs. Most biologics (including interleukin-1 (IL-1) and tumor necrosis factor (TNF) inhibitors), sprifermin, and bisphosphonates failed to yield satisfactory results when treating OA. OA clinical heterogeneity is one of the primary reasons for the failure of these clinical trials, which can require different therapeutic approaches based on different phenotypes. This review describes the latest insights into the development of DMOADs. We summarize in this review the efficacy and safety profiles of various DMOADs targeting cartilage, synovitis, and subchondral bone endotypes in phase 2 and 3 clinical trials. To conclude, we summarize the reasons for clinical trial failures in OA and suggest possible solutions.
Collapse
Affiliation(s)
- Shengfa Li
- Clinical Research Center, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Peihua Cao
- Clinical Research Center, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Tianyu Chen
- Clinical Research Center, Zhujiang Hospital, Southern Medical University, Guangzhou, China
- Department of Orthopedics, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
| | - Changhai Ding
- Clinical Research Center, Zhujiang Hospital, Southern Medical University, 261 Industry Road, Guangzhou 510515, China
- Menzies Institute for Medical Research, University of Tasmania, Hobart, TAS, Australia
- Clinical Research Center, The Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, China
| |
Collapse
|
15
|
Wu W, Liu J, Lin X, He Z, Zhang H, Ji L, Gong P, Zhou F, Liu W. Dual-functional MOFs-based hybrid microgel advances aqueous lubrication and anti-inflammation. J Colloid Interface Sci 2023; 644:200-210. [PMID: 37116318 DOI: 10.1016/j.jcis.2023.04.071] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Revised: 04/03/2023] [Accepted: 04/17/2023] [Indexed: 04/30/2023]
Abstract
This paper demonstrates the hybridization of copolymer microgel with drug-loaded metal-organic frameworks nanoparticles that can achieve excellent aqueous lubricating performance and anti-inflammatory effect for synergistic treatment of osteoarthritis (OA). Poly(ethylene glycol)-graft-poly(N-isopropylacrylamide) (PEG-g-PNIPAm) microgel layer is grown on the MIL-101(Cr) surface via one-pot soap-free emulsion polymerization method. The lower critical solution temperature of the MIL-101(Cr)@PEG-g-PNIPAm hybrid is raised significantly by incorporating PEG chains into the PNIPAm microgel matrix, which greatly enhances the high-temperature aqueous dispersion stability. The hybrid microgel demonstrated reversibly thermo-sensitive swelling-collapsing behavior to modulate the optical properties and hydrodynamic size. Using as aqueous lubricating additives, the hybrid reduces over 64% and 97% in friction coefficient and wear volume. Also, the hybrid supports desirable temperature-controlled lubrication modulation due to their reversible thermo-responsive behavior, which is benefit to joint lubrication of OA. After encapsulating anti-inflammatory diclofenac sodium (DS), the DS-MIL-101(Cr)@PEG-g-PNIPAm shows thermo-responsive drug release in aqueous media, which can improve the drug-delivery efficiency. By co-culturing the DS-loaded hybrid with human normal chondrocytes, we demonstrate good biocompatibility and anti-inflammatory effect on the chondrocytes with inflammation by regulating the expression of OA-related genes and proteins. Our work establishes multifunctional MOFs-based hybrid microgel systems for advanced colloids modulation and biomedical application.
Collapse
Affiliation(s)
- Wei Wu
- State Key Laboratory of Solidification Processing, Center of Advanced Lubrication and Seal Materials, School of Materials Science and Engineering, Northwestern Polytechnical University, Xi'an 710072, PR China
| | - Jianxi Liu
- State Key Laboratory of Solidification Processing, Center of Advanced Lubrication and Seal Materials, School of Materials Science and Engineering, Northwestern Polytechnical University, Xi'an 710072, PR China.
| | - Xiao Lin
- Key Lab for Space Biosciences and Biotechnology, Research Center for Special Medicine and Health Systems Engineering, School of Life Sciences, Northwestern Polytechnical University, Xi'an 710072, PR China
| | - Zhengze He
- State Key Laboratory of Solidification Processing, Center of Advanced Lubrication and Seal Materials, School of Materials Science and Engineering, Northwestern Polytechnical University, Xi'an 710072, PR China
| | - Hui Zhang
- Key Lab for Space Biosciences and Biotechnology, Research Center for Special Medicine and Health Systems Engineering, School of Life Sciences, Northwestern Polytechnical University, Xi'an 710072, PR China
| | - Le Ji
- Department of Orthopedic Surgery, Shaanxi Provincial People's Hospital, Xi'an 710068, PR China
| | - Peiwei Gong
- State Key Laboratory of Solidification Processing, Center of Advanced Lubrication and Seal Materials, School of Materials Science and Engineering, Northwestern Polytechnical University, Xi'an 710072, PR China
| | - Feng Zhou
- State Key Laboratory of Solid Lubrication, Lanzhou Institute of Chemical Physics, Chinese Academy of Sciences, Lanzhou 730000, PR China
| | - Weimin Liu
- State Key Laboratory of Solidification Processing, Center of Advanced Lubrication and Seal Materials, School of Materials Science and Engineering, Northwestern Polytechnical University, Xi'an 710072, PR China; State Key Laboratory of Solid Lubrication, Lanzhou Institute of Chemical Physics, Chinese Academy of Sciences, Lanzhou 730000, PR China
| |
Collapse
|
16
|
Bruno MC, Cristiano MC, Celia C, d'Avanzo N, Mancuso A, Paolino D, Wolfram J, Fresta M. Injectable Drug Delivery Systems for Osteoarthritis and Rheumatoid Arthritis. ACS NANO 2022; 16:19665-19690. [PMID: 36512378 DOI: 10.1021/acsnano.2c06393] [Citation(s) in RCA: 71] [Impact Index Per Article: 23.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/17/2023]
Abstract
Joint diseases are one of the most common causes of morbidity and disability worldwide. The main diseases that affect joint cartilage are osteoarthritis and rheumatoid arthritis, which require chronic treatment focused on symptomatic relief. Conventional drugs administered through systemic or intra-articular routes have low accumulation and/or retention in articular cartilage, causing dose-limiting toxicities and reduced efficacy. Therefore, there is an urgent need to develop improved strategies for drug delivery, in particular, the use of micro- and nanotechnology-based methods. Encapsulation of therapeutic agents in delivery systems reduces drug efflux from the joint and protects against rapid cellular and enzymatic clearance following intra-articular injection. Consequently, the use of drug delivery systems decreases side effects and increases therapeutic efficacy due to enhanced drug retention in the intra-articular space. Additionally, the frequency of intra-articular administration is reduced, as delivery systems enable sustained drug release. This review summarizes various advanced drug delivery systems, such as nano- and microcarriers, developed for articular cartilage diseases.
Collapse
Affiliation(s)
- Maria Chiara Bruno
- Department of Health Sciences, School of Pharmacy and Nutraceuticals, University "Magna Græcia" of Catanzaro, Campus Universitario "S. Venuta", Building of BioSciences, Viale S. Venuta, Germaneto-Catanzaro, I-88100, Italy
| | - Maria Chiara Cristiano
- Department of Experimental and Clinical Medicine, School of Pharmacy and Nutraceuticals, University "Magna Græcia" of Catanzaro, Campus Universitario "S. Venuta", Building of BioSciences, Viale S. Venuta, Germaneto-Catanzaro, I-88100, Italy
| | - Christian Celia
- Department of Pharmacy, University of Chieti - Pescara "G. d'Annunzio", Via dei Vestini 31, Chieti, I-66100, Italy
- Laboratory of Drug Targets Histopathology, Institute of Cardiology, Lithuanian University of Health Sciences, A. Mickeviciaus g. 9, LT-44307, Kaunas, Lithuania
| | - Nicola d'Avanzo
- Department of Health Sciences, School of Pharmacy and Nutraceuticals, University "Magna Græcia" of Catanzaro, Campus Universitario "S. Venuta", Building of BioSciences, Viale S. Venuta, Germaneto-Catanzaro, I-88100, Italy
- Department of Pharmacy, University of Chieti - Pescara "G. d'Annunzio", Via dei Vestini 31, Chieti, I-66100, Italy
| | - Antonia Mancuso
- Department of Experimental and Clinical Medicine, School of Pharmacy and Nutraceuticals, University "Magna Græcia" of Catanzaro, Campus Universitario "S. Venuta", Building of BioSciences, Viale S. Venuta, Germaneto-Catanzaro, I-88100, Italy
| | - Donatella Paolino
- Department of Experimental and Clinical Medicine, School of Pharmacy and Nutraceuticals, University "Magna Græcia" of Catanzaro, Campus Universitario "S. Venuta", Building of BioSciences, Viale S. Venuta, Germaneto-Catanzaro, I-88100, Italy
| | - Joy Wolfram
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, QLD 4072, Australia
- School of Chemical Engineering, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Massimo Fresta
- Department of Health Sciences, School of Pharmacy and Nutraceuticals, University "Magna Græcia" of Catanzaro, Campus Universitario "S. Venuta", Building of BioSciences, Viale S. Venuta, Germaneto-Catanzaro, I-88100, Italy
| |
Collapse
|
17
|
Advances in Polymeric Colloids for Cancer Treatment. Polymers (Basel) 2022; 14:polym14245445. [PMID: 36559812 PMCID: PMC9788371 DOI: 10.3390/polym14245445] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Revised: 12/01/2022] [Accepted: 12/05/2022] [Indexed: 12/14/2022] Open
Abstract
Polymer colloids have remarkable features and are gaining importance in many areas of research including medicinal science. Presently, the innovation of cancer drugs is at the top in the world. Polymer colloids have been used as drug delivery and diagnosis agents in cancer treatment. The polymer colloids may be of different types such as micelles, liposomes, emulsions, cationic carriers, and hydrogels. The current article describes the state-of-the-art polymer colloids for the treatment of cancer. The contents of this article are about the role of polymeric nanomaterials with special emphasis on the different types of colloidal materials and their applications in targeted cancer therapy including cancer diagnoses. In addition, attempts are made to discuss future perspectives. This article will be useful for academics, researchers, and regulatory authorities.
Collapse
|
18
|
Xu XL, Xue Y, Ding JY, Zhu ZH, Wu XC, Song YJ, Cao YL, Tang LG, Ding DF, Xu JG. Nanodevices for deep cartilage penetration. Acta Biomater 2022; 154:23-48. [PMID: 36243371 DOI: 10.1016/j.actbio.2022.10.007] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Revised: 09/30/2022] [Accepted: 10/04/2022] [Indexed: 12/14/2022]
Abstract
Osteoarthritis (OA) is a degenerative joint disease and is the main cause of chronic pain and functional disability in adults. Articular cartilage is a hydrated soft tissue that is composed of normally quiescent chondrocytes at a low density, a dense network of collagen fibrils with a pore size of 60-200 nm, and aggrecan proteoglycans with high-density negative charge. Although certain drugs, nucleic acids, and proteins have the potential to slow the progression of OA and restore the joints, these treatments have not been clinically applied owing to the lack of an effective delivery system capable of breaking through the cartilage barrier. Recently, the development of nanotechnology for delivery systems renders new ideas and treatment methods viable in overcoming the limited penetration. In this review, we focus on current research on such applications of nanotechnology, including exosomes, protein-based cationic nanocarriers, cationic liposomes/solid lipid nanoparticles, amino acid-based nanocarriers, polyamide derivatives-based nanocarriers, manganese dioxide, and carbon nanotubes. Exosomes are the smallest known nanoscale extracellular vesicles, and they can quickly deliver nucleic acids or proteins to the required depth. Through electrostatic interactions, nanocarriers with appropriate balance in cationic property and particle size have a strong ability to penetrate cartilage. Although substantial preclinical evidence has been obtained, further optimization is necessary for clinical transformation. STATEMENT OF SIGNIFICANCE: The dense cartilage matrix with high-negative charge was associated with reduced therapeutic effect in osteoarthritis patients with deep pathological changes. However, a systematic review in nanodevices for deep cartilage penetration is still lacking. Current approaches to assure penetration of nanosystems into the depth of cartilage were reviewed, including nanoscale extracellular vesicles from different cell lines and nanocarriers with appropriate balance in cationic property and size particle. Moreover, nanodevices entering clinical trials and further optimization were also discussed, providing important guiding significance to future research.
Collapse
Affiliation(s)
- Xiao-Ling Xu
- Shulan International Medical College, Zhejiang Shuren University, Hangzhou 310015, China
| | - Yan Xue
- Shanghai Yangzhi Rehabilitation Hospital (Shanghai Sunshine Rehabilitation Centre), School of Medicine, Tongji University, Shanghai 201613, China
| | - Jia-Ying Ding
- School of Rehabilitation Science, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Zhi-Heng Zhu
- School of Rehabilitation Science, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Xi-Chen Wu
- School of Rehabilitation Science, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Yong-Jia Song
- School of Rehabilitation Science, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Yue-Long Cao
- Shi's Center of Orthopedics and Traumatology, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Long-Guang Tang
- International Institutes of Medicine, The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu 322000, China.
| | - Dao-Fang Ding
- School of Rehabilitation Science, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China.
| | - Jian-Guang Xu
- School of Rehabilitation Science, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China.
| |
Collapse
|
19
|
Feng C, Pan L, Qin X, Li D, Chen T, Lin Z, Li G, Wang Q. Inflammation-homing "living drug depot" for efficient arthritis treatment. Acta Biomater 2022; 150:324-336. [PMID: 35840107 DOI: 10.1016/j.actbio.2022.07.013] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Revised: 06/29/2022] [Accepted: 07/06/2022] [Indexed: 11/28/2022]
Abstract
Delivering therapeutic agents efficiently to inflamed joints remains an intractable problem in rheumatoid arthritis (RA) treatment due to the complicated physiological barriers. Circulating monocytes could selectively migrate to inflamed sites and differentiate into resident macrophages to aggravate RA. Therefore, a drug carrier that can be specifically internalized by circulating monocytes and switch monocytes into anti-inflammatory phenotype when reaching inflamed sites, might bypass the in vivo physiological barriers and achieve efficient RA therapy. Herein, we design a dextran sulfate (DS) functionalized nanoparticle (ZDNP) to selectively deliver anti-inflammatory agent dexamethasone (Dex) to circulating monocytes via the scavenger receptors on monocytes. Monocytes engulfing drug-loaded ZDNP could subsequently home to arthritic joints and act as a "living drug depot" to combat RA. Results revealed that ZDNP could be preferentially internalized by circulating monocytes when intravenously administrated in vivo. In a rat arthritic model, we found that circulating monocytes remarkably facilitated drug distribution and retention in inflamed joints. Moreover, monocytes engulfing drug-loaded nanoparticles exhibited favorable anti-inflammatory ability and M2-biased differentiation. Our work offers a facile approach to achieve site-directed anti-inflammatory therapy by taking advantage of the inflammation-homing ability of circulating monocytes. STATEMENT OF SIGNIFICANCE: Circulating monocytes can migrate to inflamed sites and then differentiate into macrophages to aggravate arthritis. Therefore, a drug carrier that can be specifically internalized by circulating monocytes and switch monocytes into anti-inflammatory phenotype when reaching inflamed sites may achieve efficient arthritis therapy. Here, we designed a monocyte-targeting nanoparticle (ZDNP) to selectively deliver anti-inflammatory Dex to circulating monocytes. When injected intravenously, ZDNP was effectively internalized by circulating monocytes via a scavenger receptor and subsequently was transported to arthritic joints, where monocytes engulfing the drug-loaded nanoparticles could switch to an anti-inflammatory phenotype to inhibit arthritis progress. We provide detailed evidence about the in vivo fate of ZDNP and unravel how monocytes act as a "living drug depot" to achieve site-directed arthritis therapy.
Collapse
Affiliation(s)
- Chenglan Feng
- Key Laboratory of Advanced Technologies of Materials, Ministry of Education and School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu 610031, China
| | - Lihua Pan
- Key Laboratory of Advanced Technologies of Materials, Ministry of Education and School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu 610031, China
| | - Xianyan Qin
- Key Laboratory of Advanced Technologies of Materials, Ministry of Education and School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu 610031, China
| | - Daming Li
- Key Laboratory of Advanced Technologies of Materials, Ministry of Education and School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu 610031, China
| | - Tao Chen
- Key Laboratory of Advanced Technologies of Materials, Ministry of Education and School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu 610031, China
| | - Zhicong Lin
- Key Laboratory of Advanced Technologies of Materials, Ministry of Education and School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu 610031, China
| | - Guojiao Li
- Key Laboratory of Advanced Technologies of Materials, Ministry of Education and School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu 610031, China
| | - Qin Wang
- Key Laboratory of Advanced Technologies of Materials, Ministry of Education and School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu 610031, China.
| |
Collapse
|
20
|
Zhu C, Han S, Zeng X, Zhu C, Pu Y, Sun Y. Multifunctional thermo-sensitive hydrogel for modulating the microenvironment in Osteoarthritis by polarizing macrophages and scavenging RONS. J Nanobiotechnology 2022; 20:221. [PMID: 35526013 PMCID: PMC9077879 DOI: 10.1186/s12951-022-01422-9] [Citation(s) in RCA: 44] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Accepted: 04/13/2022] [Indexed: 02/06/2023] Open
Abstract
Osteoarthritis (OA) is a common degenerative joint disease that can lead to disability. Blocking the complex malignant feedback loop system dominated by oxidative stress and pro-inflammatory factors is the key to treating OA. Here, we develop a multifunctional composite thermo-sensitive hydrogel (HPP@Cu gel), which is utilized by Poloxamer 407 (P407) and hyaluronic acid (HA) mixture as the gel matrix, then physically mixed with copper nanodots (Cu NDs) and platelet-rich plasma (PRP). Cu NDs is a novel nano-scavenger of reactive oxygen and nitrogen species (RONS) with efficient free radical scavenging activity. HPP@Cu gel is injected into the articular cavity, where it form an in situ gel that slowly released Cu NDs, HA, and PRP, prolonging the duration of drug action. Our results indicate that HPP@Cu gel could efficiently remove RONS from inflammatory sites and promote repolarization of macrophages to an anti-inflammatory phenotype. The HPP@Cu gel therapy dramatically reduces cartilage degradation and inflammatory factor production in OA rats. This study provides a reliable reference for the application of injectable hydrogels in inflammatory diseases associated with oxidative stress.
Collapse
Affiliation(s)
- Chunrong Zhu
- Department of Pharmaceutics, School of Pharmacy, Qingdao University, Qingdao, 266021, People's Republic of China
| | - Shangcong Han
- Department of Pharmaceutics, School of Pharmacy, Qingdao University, Qingdao, 266021, People's Republic of China
| | - Xianhu Zeng
- Department of Pharmaceutics, School of Pharmacy, Qingdao University, Qingdao, 266021, People's Republic of China
| | - Chunxiao Zhu
- Department of Pharmaceutics, School of Pharmacy, Qingdao University, Qingdao, 266021, People's Republic of China
| | - Yuji Pu
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, 610064, People's Republic of China
| | - Yong Sun
- Department of Pharmaceutics, School of Pharmacy, Qingdao University, Qingdao, 266021, People's Republic of China.
| |
Collapse
|
21
|
Development of an intelligent, stimuli-responsive transdermal system for efficient delivery of Ibuprofen against rheumatoid arthritis. Int J Pharm 2021; 610:121242. [PMID: 34737113 DOI: 10.1016/j.ijpharm.2021.121242] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Revised: 10/23/2021] [Accepted: 10/25/2021] [Indexed: 12/26/2022]
Abstract
The present study aimed to fabricate and evaluate the therapeutic efficacy of pH-responsive Ibuprofen (IB) nanoparticles (NPs) loaded transdermal hydrogel against rheumatoid arthritis (RA). The IB loaded Eudragit® L 100 (EL 100) nanoparticles were formulated through a modified nanoprecipitation technique and optimized using central composite design software. The optimized NPs were loaded into Carbopol® 934-based hydrogel by solvent evaporation method and were analyzed for physicochemical characteristics. The mean particle size of the prepared NPs was 48 nm with an entrapment efficiency of 90%. The transdermal hydrogel showed a pH-responsive sustained drug release and high penetration through the skin. Moreover, the prepared nanocarrier system exhibited therapeutic efficacy at inflamed joints' sites both in acute and chronic RA mice model. The therapeutic efficacy of the prepared formulation was confirmed through the results of various behavioral, biochemical, and cytokines-based assays. Similarly, the assessment of histopathological and radiological images, as well as the skin irritation studies further strengthens the potential use of the prepared formulation through the transdermal route. The current findings suggested that IB loaded pH-responsive NPs based transdermal hydrogel can be used as an efficient agent to manage RA.
Collapse
|
22
|
Nanodispersions of Polyelectrolytes Based on Humic Substances: Isolation, Physico-Chemical Characterization and Evaluation of Biological Activity. Pharmaceutics 2021; 13:pharmaceutics13111954. [PMID: 34834368 PMCID: PMC8623726 DOI: 10.3390/pharmaceutics13111954] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Revised: 11/12/2021] [Accepted: 11/16/2021] [Indexed: 12/16/2022] Open
Abstract
Natural polyelectrolytes, including in the form of complexes with colloidal particles, are increasingly used in pharmacy due to the possibility of regulated attachment of medicinal substances and their targeted delivery to the target organ. However, the formation, stability, and molecular-mass characteristics of polyelectrolyte nanodispersions (ND) vary depending on the nature and composition of the medium of their origin. This is due to the lack of standardized approaches to quality control and regulatory documentation for most natural ND. In this paper, we first introduced the isolation, followed by investigations into their physico-chemical properties and bioactivity. Using the dried droplet method, we were able to detect the “coffee ring effect”. Fractographic studies of the surface structure of EHA and FA dried samples using SEM showed its heterogeneity and the presence of submicron particles encapsulated in the internal molecular cavities of polyelectrolyte. FTIR spectroscopy revealed the ND chemical structure of benzo-α-pyron and benzo-γ-pyron, consisting of nanoparticles and a branched frame part. The main elements detected by X-ray fluorescence in humic substance extract and fulvic acid include Si, P, S, K, Ca, Mn, Fe, Cu, Zn, whereas Fe is in high concentrations. The UV-spectra and fluorescent radiation demonstrated the possibility of studying the effect of the fulvate chromone structure on its optical properties. It is shown that dilution of the initial solutions of polyelectrolytes 1:10 contributes to the detection of smaller nanoparticles and an increase in the absolute value of the negative ζ-potential as a factor of ND stability. A study of the EHS effect on the SARS-CoV-2 virus infectious titer in the Vero E6 cell showed the effective against virus both in the virucidal scheme (the SI is 11.90–22.43) and treatment/prevention scheme (the SI is 34.85–57.33). We assume that polyelectrolyte ND prevent the binding of the coronavirus spike glycoprotein to the receptor. Taking into account the results obtained, we expect that the developed approach can become unified for the standardization of the ND natural polyelectrolytes complex, which has great prospects for use in pharmacy and medicine as a drug with antiviral activity.
Collapse
|
23
|
Yu P, Liu Y, Xie J, Li J. Spatiotemporally controlled calcitonin delivery: Long-term and targeted therapy of skeletal diseases. J Control Release 2021; 338:486-504. [PMID: 34481022 DOI: 10.1016/j.jconrel.2021.08.056] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2021] [Revised: 08/29/2021] [Accepted: 08/30/2021] [Indexed: 02/05/2023]
Abstract
Bone is a connective tissue that support the entire body and protect the internal organs. However, there are great challenges on curing intractable skeletal diseases such as hypercalcemia, osteoporosis and osteoarthritis. To address these issues, calcitonin (CT) therapy is an effective treatment alternative to regulate calcium metabolism and suppress inflammation response, which are closely related to skeletal diseases. Traditional calcitonin formulation requires frequent administration due to the low bioavailability resulting from the short half-life and abundant calcitonin receptors distributed through the whole body. Therefore, long-term and targeted calcitonin delivery systems (LCDS and TCDS) have been widely explored as the popular strategies to overcome the intrinsic limitations of calcitonin and improve the functions of calcium management and inflammation inhibition in recent years. In this review, we first explain the physiological effects of calcitonin on bone remodeling: (i) inhibitory effects on osteoclasts and (ii) facilitated effects on osteoblasts. Then we summarized four strategies for spatiotemporally controlled delivery of calcitonin: micro-/nanomedicine (e.g. inorganic micro-/nanomedicine, polymeric micro-/nanomedicine and supramolecular assemblies), hydrogels (especially thermosensitive hydrogels), prodrug (PEGylation and targeting design) and hybrid biomaterials. Subsequently, we discussed the application of LCDS and TCDS in treating hypercalcemia, osteoporosis, and arthritis. Understanding and analyzing these advanced calcitonin delivery applications are essential for future development of calcitonin therapies toward skeletal diseases with superior efficacy in clinic.
Collapse
Affiliation(s)
- Peng Yu
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu 610065, PR China
| | - Yanpeng Liu
- Hangzhou Global Scientific and Technological Innovation Center, Zhejiang University, Hangzhou 311200, PR China
| | - Jing Xie
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu 610065, PR China.
| | - Jianshu Li
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu 610065, PR China; State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, PR China; Med-X Center for Materials, Sichuan University, Chengdu 610041, PR China.
| |
Collapse
|
24
|
Yang J, Yue L, Yang Z, Miao Y, Ouyang R, Hu Y. Metal-Based Nanomaterials: Work as Drugs and Carriers against Viral Infections. NANOMATERIALS (BASEL, SWITZERLAND) 2021; 11:2129. [PMID: 34443959 PMCID: PMC8400983 DOI: 10.3390/nano11082129] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Revised: 08/05/2021] [Accepted: 08/12/2021] [Indexed: 01/08/2023]
Abstract
Virus infection is one of the threats to the health of organisms, and finding suitable antiviral agents is one of the main tasks of current researchers. Metal ions participate in multiple key reaction stages of organisms and maintain the important homeostasis of organisms. The application of synthetic metal-based nanomaterials as an antiviral therapy is a promising new research direction. Based on the application of synthetic metal-based nanomaterials in antiviral therapy, we summarize the research progress of metal-based nanomaterials in recent years. This review analyzes the three inhibition pathways of metal nanomaterials as antiviral therapeutic materials against viral infections, including direct inactivation, inhibition of virus adsorption and entry, and intracellular virus suppression; it further classifies and summarizes them according to their inhibition mechanisms. In addition, the use of metal nanomaterials as antiviral drug carriers and vaccine adjuvants is summarized. The analysis clarifies the antiviral mechanism of metal nanomaterials and broadens the application in the field of antiviral therapy.
Collapse
Affiliation(s)
- Junlei Yang
- Institute of Bismuth Science, University of Shanghai for Science and Technology, Shanghai 200093, China; (J.Y.); (Z.Y.); (Y.M.)
- Institut Pasteur of Shanghai, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Yueyang Road 320, Shanghai 200031, China
| | - Lihuan Yue
- CAS Key Laboratory of Molecular Virology & Immunology, Institutional Center for Shared Technologies and Facilities, Pathogen Discovery and Big Data Center, Institut Pasteur of Shanghai, Chinese Academy of Sciences, Yueyang Road 320, Shanghai 200031, China;
- Department of Bioengineering, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Zhu Yang
- Institute of Bismuth Science, University of Shanghai for Science and Technology, Shanghai 200093, China; (J.Y.); (Z.Y.); (Y.M.)
- Institut Pasteur of Shanghai, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Yueyang Road 320, Shanghai 200031, China
| | - Yuqing Miao
- Institute of Bismuth Science, University of Shanghai for Science and Technology, Shanghai 200093, China; (J.Y.); (Z.Y.); (Y.M.)
| | - Ruizhuo Ouyang
- Institute of Bismuth Science, University of Shanghai for Science and Technology, Shanghai 200093, China; (J.Y.); (Z.Y.); (Y.M.)
| | - Yihong Hu
- Institut Pasteur of Shanghai, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Yueyang Road 320, Shanghai 200031, China
- CAS Key Laboratory of Molecular Virology & Immunology, Institutional Center for Shared Technologies and Facilities, Pathogen Discovery and Big Data Center, Institut Pasteur of Shanghai, Chinese Academy of Sciences, Yueyang Road 320, Shanghai 200031, China;
| |
Collapse
|
25
|
Lima F, Melo WG, Braga MDF, Vieira E, Câmara JV, Pierote JJ, Argôlo Neto N, Silva Filho E, Fialho AC. Chitosan-based hydrogel for treatment of temporomandibular joint arthritis. POLIMEROS 2021. [DOI: 10.1590/0104-1428.20210026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
|