1
|
Dong L, Lou W, Xu C, Wang J. Naringenin cationic lipid-modified nanoparticles mitigate MASLD progression by modulating lipid homeostasis and gut microbiota. J Nanobiotechnology 2025; 23:168. [PMID: 40038718 DOI: 10.1186/s12951-025-03228-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2024] [Accepted: 02/11/2025] [Indexed: 03/06/2025] Open
Abstract
Naringenin (NAR) possesses various pharmacological activities including antioxidant, anti-inflammatory, and hepatoprotective effects. However, its therapeutic efficacy is limited by its hydrophobic and crystalline nature. This study aimed to investigate the therapeutic potential and molecular mechanisms of NAR efficiently loaded into cationic nanoparticles (NP-NAR) for treating metabolic dysfunction-associated steatotic liver disease (MASLD) in a mouse model. The results demonstrated that NP-NAR effectively ameliorated lipid metabolism dysbiosis, oxidative stress, insulin resistance, and inflammation in MASLD mice. Transcriptomic analysis and molecular data revealed that NP-NAR promoted fatty acid oxidation via activation of the PPAR signaling pathway, reduced hepatic lipid uptake and lipogenesis by inhibiting the expressions of key genes including CD36, ACC, and FASN. Moreover, NP-NAR modulated cholesterol metabolism by inhibiting the classical bile acid synthesis pathway. 16 S rDNA gene sequencing revealed a disbalanced gut microbiota in MASLD mice, whereas NP-NAR treatment statistically reversed the abundance changes of several intestinal bacteria at the phylum and genus levels, which partly contributed to the balance in intestinal metabolite production, including short-chain fatty acids. In conclusion, these findings suggest that NP-NAR may be a promising candidate for the treatment of obesity-associated MASLD, offering new insight into the mechanisms underlying NAR's efficacy against MASLD.
Collapse
Affiliation(s)
- Lu Dong
- School of Food Science and Engineering, South China University of Technology, Guangzhou, Guangdong Province, 510641, China
| | - Wenyong Lou
- School of Food Science and Engineering, South China University of Technology, Guangzhou, Guangdong Province, 510641, China
| | - Congfei Xu
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou, 511442, China.
| | - Juan Wang
- School of Food Science and Engineering, South China University of Technology, Guangzhou, Guangdong Province, 510641, China.
| |
Collapse
|
2
|
Sawan S, Kumari A, Majie A, Ghosh A, Karmakar V, Kumari N, Ghosh S, Gorain B. siRNA-based nanotherapeutic approaches for targeted delivery in rheumatoid arthritis. BIOMATERIALS ADVANCES 2025; 168:214120. [PMID: 39577366 DOI: 10.1016/j.bioadv.2024.214120] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Revised: 11/15/2024] [Accepted: 11/15/2024] [Indexed: 11/24/2024]
Abstract
Rheumatoid arthritis (RA), characterized as a systemic autoimmune ailment, predominantly results in substantial joint and tissue damage, affecting millions of individuals globally. Modern treatment modalities are being explored as the traditional RA therapy with non-specific immunosuppressive drugs showcased potential side effects and variable responses. Research potential with small interfering RNA (siRNA) depicted potential in the treatment of RA. These siRNA-based therapies could include genes encoding pro-inflammatory cytokines like TNF-α, IL-1, and IL-6, as well as other molecular targets such as RANK, p38 MAPK, TGF-β, Wnt/Fz complex, and HIF. By downregulating the expression of these genes, siRNA-based nanoformulations can attenuate inflammation, inhibit immune system dysregulation, and prevent tissue damage associated with RA. Strategies of delivering siRNA molecules through nanocarriers could be targeted to treat RA effectively, where specific genes associated with this autoimmune disease pathology can be selectively silenced. Additionally, simultaneous targeting of multiple molecular pathways may offer synergistic therapeutic benefits, potentially leading to more effective and safer therapeutic strategies for RA patients. This review critically highlights the in-depth pathology of RA, RNA interference-mediated molecular targets, and nanocarrier-based siRNA delivery strategies, along with the challenges and opportunities to harbor future solutions.
Collapse
Affiliation(s)
- Sweta Sawan
- Department of Pharmaceutical Sciences and Technology, Birla Institute of Technology, Mesra, Ranchi, India
| | - Ankita Kumari
- Department of Pharmaceutical Sciences and Technology, Birla Institute of Technology, Mesra, Ranchi, India
| | - Ankit Majie
- Department of Pharmaceutical Sciences and Technology, Birla Institute of Technology, Mesra, Ranchi, India
| | - Arya Ghosh
- Department of Pharmaceutical Sciences and Technology, Birla Institute of Technology, Mesra, Ranchi, India
| | - Varnita Karmakar
- Department of Pharmaceutical Sciences and Technology, Birla Institute of Technology, Mesra, Ranchi, India
| | - Nimmy Kumari
- Department of Pharmaceutical Sciences and Technology, Birla Institute of Technology, Mesra, Ranchi, India
| | - Santanu Ghosh
- Department of Pharmaceutical Sciences and Technology, Birla Institute of Technology, Mesra, Ranchi, India.
| | - Bapi Gorain
- Department of Pharmaceutical Sciences and Technology, Birla Institute of Technology, Mesra, Ranchi, India.
| |
Collapse
|
3
|
Rani L, Mathur P, Verma R, Kumar V, Mishra AK, Sahoo PK. Translation Research in Therapeutic Approaches from Conventional to Novel Nano-therapeutics for Rheumatoid Arthritis Treatment. Curr Rheumatol Rev 2025; 21:37-53. [PMID: 38629371 DOI: 10.2174/0115733971288433240408062359] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Revised: 03/15/2024] [Accepted: 03/18/2024] [Indexed: 04/05/2025]
Abstract
Rheumatoid arthritis is a systemic autoimmune disorder related to joint inflammation, bone erosion, and deformity. Numerous studies indicate that the causes and consequences of RA are still being debated, and therapeutic strategies are in the translation stage. Non-steroidal anti-inflammatory drugs continue to be often used to relieve pain. Still, due to their poor efficacy, failure to halt the spread of the disease, and undesirable adverse effects, they are no longer regarded as first-line treatments. The development of biologic DMRDs designed to reduce the inflammatory response led to substantial changes to the strategy for managing this disease. Although biologic DMRDs have made significant strides in the management of Rheumatoid arthritis, certain patients' lack of response to biological approaches and therapy cessation due to systemic toxicity are unresolved problems. Therefore, to improve the in vivo effect and reduce systemic adverse effects, new approaches are needed to proactively target and transport therapeutic molecules to target sites. The intriguing method of nanotechnology enables the encapsulation of drugs to prevent their deterioration and systemic adverse effects. The next generation of Rheumatoid arthritis therapies might be based on advances in nanomaterial-based drug delivery, Trojan horse, and antibody targeting approaches. This article presents an overview of the advancements in Rheumatoid arthritis therapy, ranging from traditional methods to recent cutting-edge, ongoing pre-clinical and clinical approaches.
Collapse
Affiliation(s)
- Laxmi Rani
- Department of Pharmaceutics, Delhi Institute of Pharmaceutical Science and Research, DPSR University, Sector-3, MB Road Pushp Vihar, New Delhi, 110017, India
| | - Pooja Mathur
- Department of Pharmacy, School of Medical and Allied Sciences, G D Goenka University, Sohna, Gurugram, Haryana, 122103, India
| | - Ravinder Verma
- Department of Pharmaceutical Sciences, Chaudhary Bansi Lal University, Bhiwani, 127021, India
| | - Vivek Kumar
- Institute of Pharmacy, Shri Ram College of Pharmacy, Karnal, India
| | - Ashwini Kumar Mishra
- Department of Pharmaceutics, Delhi Institute of Pharmaceutical Science and Research, DPSR University, Sector-3, MB Road Pushp Vihar, New Delhi, 110017, India
| | - Pravat Kumar Sahoo
- Department of Pharmaceutics, Delhi Institute of Pharmaceutical Science and Research, DPSR University, Sector-3, MB Road Pushp Vihar, New Delhi, 110017, India
| |
Collapse
|
4
|
Park JS, Jeon H, Lee Y, Cheon SY, Lee D, Lim SG, Koo H. Rapid DNA Repair in Mesenchymal Stem Cells and Bone Regeneration by Nanoparticle-Based Codelivery of Nrf2-mRNA and Dexamethasone. ACS NANO 2024; 18:31877-31890. [PMID: 39520360 DOI: 10.1021/acsnano.4c08939] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/16/2024]
Abstract
Directional differentiation is a key factor determining the result of stem cell therapy. Herein, we developed a polyethylenimine (PEI)-coated poly(lactic-co-glycolic) acid (PLGA) nanoparticle (mPDN) carrying both nuclear factor erythroid 2-related factor 2 (Nrf2) mRNA and dexamethasone (Dex) to human mesenchymal stem cells (hMSCs). The combination of Dex and Nrf2-mRNA delivered by mPDN promoted the osteogenic differentiation of hMSCs. In particular, Nrf2-mRNA rapidly reduced the DNA damage caused by ROS due to early and efficient gene expression at 3 h after treatment, which was not achieved in traditional pDNA systems. High and rapid transfection, effective ROS-scavenging effect, and protection of mitochondrial dynamics were observed in hMSCs after treatment with the resulting Nrf2-mPDN. Osteogenic differentiation was also observed in 3D pellets for up to 5 weeks. Finally, the effects of rapid DNA repair in hMSCs by Nrf2-mPDN and on in vivo bone regeneration were evaluated in a rat femoral bone defect model using CT. This study demonstrated the potential of an NP-based codelivery system and efficient transfection of mRNA at early stages in hMSCs for bone regeneration and stem cell therapy.
Collapse
Affiliation(s)
- Ji Sun Park
- Department of Medical Life Sciences, Department of Medical Sciences (Graduate School), and Catholic Photomedicine Research Institute, College of Medicine, The Catholic University of Korea, 222 Banpo-daero, Seocho-gu, Seoul 06591, Republic of Korea
| | - Hayoung Jeon
- Department of Medical Life Sciences, Department of Medical Sciences (Graduate School), and Catholic Photomedicine Research Institute, College of Medicine, The Catholic University of Korea, 222 Banpo-daero, Seocho-gu, Seoul 06591, Republic of Korea
| | - Yeeun Lee
- Department of Medical Life Sciences, Department of Medical Sciences (Graduate School), and Catholic Photomedicine Research Institute, College of Medicine, The Catholic University of Korea, 222 Banpo-daero, Seocho-gu, Seoul 06591, Republic of Korea
| | - Seo Young Cheon
- Department of Medical Life Sciences, Department of Medical Sciences (Graduate School), and Catholic Photomedicine Research Institute, College of Medicine, The Catholic University of Korea, 222 Banpo-daero, Seocho-gu, Seoul 06591, Republic of Korea
| | - Donghyun Lee
- Department of Medical Life Sciences, Department of Medical Sciences (Graduate School), and Catholic Photomedicine Research Institute, College of Medicine, The Catholic University of Korea, 222 Banpo-daero, Seocho-gu, Seoul 06591, Republic of Korea
| | - Seong Gi Lim
- Department of Medical Life Sciences, Department of Medical Sciences (Graduate School), and Catholic Photomedicine Research Institute, College of Medicine, The Catholic University of Korea, 222 Banpo-daero, Seocho-gu, Seoul 06591, Republic of Korea
| | - Heebeom Koo
- Department of Medical Life Sciences, Department of Medical Sciences (Graduate School), and Catholic Photomedicine Research Institute, College of Medicine, The Catholic University of Korea, 222 Banpo-daero, Seocho-gu, Seoul 06591, Republic of Korea
| |
Collapse
|
5
|
Mao K, Wang J, Xie Q, Yang YG, Shen S, Sun T, Wang J. Cationic nanoparticles-based approaches for immune tolerance induction in vivo. J Control Release 2024; 366:425-447. [PMID: 38154540 DOI: 10.1016/j.jconrel.2023.12.044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Revised: 12/04/2023] [Accepted: 12/25/2023] [Indexed: 12/30/2023]
Abstract
The development of autoimmune diseases and the rejection of transplanted organs are primarily caused by an exaggerated immune response to autoantigens or graft antigens. Achieving immune tolerance is crucial for the effective treatment of these conditions. However, traditional therapies often have limited therapeutic efficacy and can result in systemic toxic effects. The emergence of nanomedicine offers a promising avenue for addressing immune-related diseases. Among the various nanoparticle formulations, cationic nanoparticles have demonstrated significant potential in inducing immune tolerance. In this review, we provide an overview of the underlying mechanism of autoimmune disease and organ transplantation rejection. We then highlight the recent advancements and advantages of utilizing cationic nanoparticles for inducing immune tolerance in the treatment of autoimmune diseases and the prevention of transplant rejection.
Collapse
Affiliation(s)
- Kuirong Mao
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, Institute of Immunology, The First Hospital, Jilin University, Changchun, Jilin, China; International Center of Future Science, Jilin University, Changchun, Jilin, China; National-local Joint Engineering Laboratory of Animal Models for Human Diseases, Changchun, Jilin, China
| | - Jialiang Wang
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, Institute of Immunology, The First Hospital, Jilin University, Changchun, Jilin, China; National-local Joint Engineering Laboratory of Animal Models for Human Diseases, Changchun, Jilin, China
| | - Qianyue Xie
- Huafu International Department, Affiliated High School of South China Normal University, Guangzhou, Guangdong, China
| | - Yong-Guang Yang
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, Institute of Immunology, The First Hospital, Jilin University, Changchun, Jilin, China; International Center of Future Science, Jilin University, Changchun, Jilin, China; National-local Joint Engineering Laboratory of Animal Models for Human Diseases, Changchun, Jilin, China
| | - Song Shen
- School of Biomedical Sciences and Engineering, Guangzhou International Campus, South China University of Technology, Guangzhou, Guangdong, China
| | - Tianmeng Sun
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, Institute of Immunology, The First Hospital, Jilin University, Changchun, Jilin, China; International Center of Future Science, Jilin University, Changchun, Jilin, China; National-local Joint Engineering Laboratory of Animal Models for Human Diseases, Changchun, Jilin, China; State Key Laboratory of Supramolecular Structure and Materials, Jilin University, Changchun, Jilin, China.
| | - Jun Wang
- School of Biomedical Sciences and Engineering, Guangzhou International Campus, South China University of Technology, Guangzhou, Guangdong, China; National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou, Guangdong, China; Key Laboratory of Biomedical Engineering of Guangdong Province, and Innovatiion Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou, Guangdong, China.
| |
Collapse
|
6
|
Singh P, Singh M, Singh B, Sharma K, Kumar N, Singh D, Klair HS, Mastana S. Implications of siRNA Therapy in Bone Health: Silencing Communicates. Biomedicines 2024; 12:90. [PMID: 38255196 PMCID: PMC10813040 DOI: 10.3390/biomedicines12010090] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Revised: 12/27/2023] [Accepted: 12/28/2023] [Indexed: 01/24/2024] Open
Abstract
The global statistics of bone disorders, skeletal defects, and fractures are frightening. Several therapeutic strategies are being used to fix them; however, RNAi-based siRNA therapy is starting to prove to be a promising approach for the prevention of bone disorders because of its advanced capabilities to deliver siRNA or siRNA drug conjugate to the target tissue. Despite its 'bench-to-bedside' usefulness and approval by food and drug administration for five siRNA-based therapeutic medicines: Patisiran, Vutrisiran, Inclisiran, Lumasiran, and Givosiran, its use for the other diseases still remains to be resolved. By correcting the complications and complexities involved in siRNA delivery for its sustained release, better absorption, and toxicity-free activity, siRNA therapy can be harnessed as an experimental tool for the prevention of complex and undruggable diseases with a personalized medicine approach. The present review summarizes the findings of notable research to address the implications of siRNA in bone health for the restoration of bone mass, recovery of bone loss, and recuperation of bone fractures.
Collapse
Affiliation(s)
- Puneetpal Singh
- Department of Human Genetics, Punjabi University, Patiala 147002, Punjab, India; (M.S.); (B.S.); (K.S.); (N.K.)
| | - Monica Singh
- Department of Human Genetics, Punjabi University, Patiala 147002, Punjab, India; (M.S.); (B.S.); (K.S.); (N.K.)
| | - Baani Singh
- Department of Human Genetics, Punjabi University, Patiala 147002, Punjab, India; (M.S.); (B.S.); (K.S.); (N.K.)
| | - Kirti Sharma
- Department of Human Genetics, Punjabi University, Patiala 147002, Punjab, India; (M.S.); (B.S.); (K.S.); (N.K.)
| | - Nitin Kumar
- Department of Human Genetics, Punjabi University, Patiala 147002, Punjab, India; (M.S.); (B.S.); (K.S.); (N.K.)
| | - Deepinder Singh
- Vardhman Mahavir Health Care, Urban Estate, Ph-II, Patiala 147002, Punjab, India
| | | | - Sarabjit Mastana
- Human Genomics Laboratory, School of Sport, Exercise and Health Sciences, Loughborough University, Loughborough LE11 3TU, UK
| |
Collapse
|
7
|
Li X, Peng X, Zoulikha M, Boafo GF, Magar KT, Ju Y, He W. Multifunctional nanoparticle-mediated combining therapy for human diseases. Signal Transduct Target Ther 2024; 9:1. [PMID: 38161204 PMCID: PMC10758001 DOI: 10.1038/s41392-023-01668-1] [Citation(s) in RCA: 87] [Impact Index Per Article: 87.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 09/14/2023] [Accepted: 10/10/2023] [Indexed: 01/03/2024] Open
Abstract
Combining existing drug therapy is essential in developing new therapeutic agents in disease prevention and treatment. In preclinical investigations, combined effect of certain known drugs has been well established in treating extensive human diseases. Attributed to synergistic effects by targeting various disease pathways and advantages, such as reduced administration dose, decreased toxicity, and alleviated drug resistance, combinatorial treatment is now being pursued by delivering therapeutic agents to combat major clinical illnesses, such as cancer, atherosclerosis, pulmonary hypertension, myocarditis, rheumatoid arthritis, inflammatory bowel disease, metabolic disorders and neurodegenerative diseases. Combinatorial therapy involves combining or co-delivering two or more drugs for treating a specific disease. Nanoparticle (NP)-mediated drug delivery systems, i.e., liposomal NPs, polymeric NPs and nanocrystals, are of great interest in combinatorial therapy for a wide range of disorders due to targeted drug delivery, extended drug release, and higher drug stability to avoid rapid clearance at infected areas. This review summarizes various targets of diseases, preclinical or clinically approved drug combinations and the development of multifunctional NPs for combining therapy and emphasizes combinatorial therapeutic strategies based on drug delivery for treating severe clinical diseases. Ultimately, we discuss the challenging of developing NP-codelivery and translation and provide potential approaches to address the limitations. This review offers a comprehensive overview for recent cutting-edge and challenging in developing NP-mediated combination therapy for human diseases.
Collapse
Affiliation(s)
- Xiaotong Li
- School of Pharmacy, China Pharmaceutical University, Nanjing, 2111198, PR China
| | - Xiuju Peng
- School of Pharmacy, China Pharmaceutical University, Nanjing, 2111198, PR China
| | - Makhloufi Zoulikha
- School of Pharmacy, China Pharmaceutical University, Nanjing, 2111198, PR China
| | - George Frimpong Boafo
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410013, PR China
| | - Kosheli Thapa Magar
- School of Pharmacy, China Pharmaceutical University, Nanjing, 2111198, PR China
| | - Yanmin Ju
- School of Pharmacy, China Pharmaceutical University, Nanjing, 2111198, PR China.
| | - Wei He
- Shanghai Skin Disease Hospital, Tongji University School of Medicine, Shanghai, 200443, China.
| |
Collapse
|
8
|
Kumari A, Kaur A, Aggarwal G. The emerging potential of siRNA nanotherapeutics in treatment of arthritis. Asian J Pharm Sci 2023; 18:100845. [PMID: 37881798 PMCID: PMC10594572 DOI: 10.1016/j.ajps.2023.100845] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Revised: 07/14/2023] [Accepted: 08/12/2023] [Indexed: 10/27/2023] Open
Abstract
RNA interference (RNAi) using small interfering RNA (siRNA) has shown potential as a therapeutic option for the treatment of arthritis by silencing specific genes. However, siRNA delivery faces several challenges, including stability, targeting, off-target effects, endosomal escape, immune response activation, intravascular degradation, and renal clearance. A variety of nanotherapeutics like lipidic nanoparticles, liposomes, polymeric nanoparticles, and solid lipid nanoparticles have been developed to improve siRNA cellular uptake, protect it from degradation, and enhance its therapeutic efficacy. Researchers are also investigating chemical modifications and bioconjugation to reduce its immunogenicity. This review discusses the potential of siRNA nanotherapeutics as a therapeutic option for various immune-mediated diseases, including rheumatoid arthritis, osteoarthritis, etc. siRNA nanotherapeutics have shown an upsurge of interest and the future looks promising for such interdisciplinary approach-based modalities that combine the principles of molecular biology, nanotechnology, and formulation sciences.
Collapse
Affiliation(s)
- Anjali Kumari
- School of Pharmaceutical Sciences, Delhi Pharmaceutical Sciences and Research University, New Delhi 110017, India
| | - Amanpreet Kaur
- Centre for Advanced Formulation Technology, Delhi Pharmaceutical Sciences and Research, New Delhi 110017, India
- School of Pharmaceutical Sciences, Delhi Pharmaceutical Sciences and Research University, New Delhi 110017, India
| | - Geeta Aggarwal
- Centre for Advanced Formulation Technology, Delhi Pharmaceutical Sciences and Research, New Delhi 110017, India
- School of Pharmaceutical Sciences, Delhi Pharmaceutical Sciences and Research University, New Delhi 110017, India
| |
Collapse
|
9
|
Zhang X, Liu Y, Xiao C, Guan Y, Gao Z, Huang W. Research Advances in Nucleic Acid Delivery System for Rheumatoid Arthritis Therapy. Pharmaceutics 2023; 15:1237. [PMID: 37111722 PMCID: PMC10145518 DOI: 10.3390/pharmaceutics15041237] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Revised: 03/31/2023] [Accepted: 04/05/2023] [Indexed: 04/29/2023] Open
Abstract
Rheumatoid arthritis (RA) is a chronic inflammatory autoimmune disease that affects the lives of nearly 1% of the total population worldwide. With the understanding of RA, more and more therapeutic drugs have been developed. However, lots of them possess severe side effects, and gene therapy may be a potential method for RA treatment. A nanoparticle delivery system is vital for gene therapy, as it can keep the nucleic acids stable and enhance the efficiency of transfection in vivo. With the development of materials science, pharmaceutics and pathology, more novel nanomaterials and intelligent strategies are applied to better and safer gene therapy for RA. In this review, we first summarized the existing nanomaterials and active targeting ligands used for RA gene therapy. Then, we introduced various gene delivery systems for RA treatment, which may enlighten the relevant research in the future.
Collapse
Affiliation(s)
- Xintong Zhang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China; (X.Z.); (Y.L.); (C.X.)
- Beijing Key Laboratory of Drug Delivery Technology and Novel Formulations, Department of Pharmaceutics, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Yanhong Liu
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China; (X.Z.); (Y.L.); (C.X.)
- Beijing Key Laboratory of Drug Delivery Technology and Novel Formulations, Department of Pharmaceutics, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Congcong Xiao
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China; (X.Z.); (Y.L.); (C.X.)
- Beijing Key Laboratory of Drug Delivery Technology and Novel Formulations, Department of Pharmaceutics, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Youyan Guan
- Department of Urology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China;
| | - Zhonggao Gao
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China; (X.Z.); (Y.L.); (C.X.)
- Beijing Key Laboratory of Drug Delivery Technology and Novel Formulations, Department of Pharmaceutics, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Wei Huang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China; (X.Z.); (Y.L.); (C.X.)
- Beijing Key Laboratory of Drug Delivery Technology and Novel Formulations, Department of Pharmaceutics, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| |
Collapse
|
10
|
Wen J, Li H, Dai H, Hua S, Long X, Li H, Ivanovski S, Xu C. Intra-articular nanoparticles based therapies for osteoarthritis and rheumatoid arthritis management. Mater Today Bio 2023; 19:100597. [PMID: 36910270 PMCID: PMC9999238 DOI: 10.1016/j.mtbio.2023.100597] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Revised: 02/19/2023] [Accepted: 02/24/2023] [Indexed: 02/27/2023] Open
Abstract
Osteoarthritis (OA) and rheumatoid arthritis (RA) are chronic and progressive inflammatory joint diseases that affect a large population worldwide. Intra-articular administration of various therapeutics is applied to alleviate pain, prevent further progression, and promote cartilage regeneration and bone remodeling in both OA and RA. However, the effectiveness of intra-articular injection with traditional drugs is uncertain and controversial due to issues such as rapid drug clearance and the barrier afforded by the dense structure of cartilage. Nanoparticles can improve the efficacy of intra-articular injection by facilitating controlled drug release, prolonged retention time, and enhanced penetration into joint tissue. This review systematically summarizes nanoparticle-based therapies for OA and RA management. Firstly, we explore the interaction between nanoparticles and joints, including articular fluids and cells. This is followed by a comprehensive analysis of current nanoparticles designed for OA/RA, divided into two categories based on therapeutic mechanisms: direct therapeutic nanoparticles and nanoparticles-based drug delivery systems. We highlight nanoparticle design for tissue/cell targeting and controlled drug release before discussing challenges of nanoparticle-based therapies for efficient OA and RA treatment and their future clinical translation. We anticipate that rationally designed local injection of nanoparticles will be more effective, convenient, and safer than the current therapeutic approach.
Collapse
Affiliation(s)
- Juan Wen
- School of Dentistry, The University of Queensland, Brisbane, Queensland, 4006, Australia
- Centre for Orofacial Regeneration, Reconstruction and Rehabilitation (COR3), School of Dentistry, The University of Queensland, Brisbane, Queensland, 4006, Australia
| | - Huimin Li
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) and Key Laboratory of Oral Biomedicine Ministry of Education, School and Hospital of Stomatology, Wuhan University, Wuhan, 430079, China
- Department of Oral and Maxillofacial Surgery, School and Hospital of Stomatology, Wuhan University, Wuhan, 430079, China
| | - Huan Dai
- School of Dentistry, The University of Queensland, Brisbane, Queensland, 4006, Australia
- Centre for Orofacial Regeneration, Reconstruction and Rehabilitation (COR3), School of Dentistry, The University of Queensland, Brisbane, Queensland, 4006, Australia
| | - Shu Hua
- School of Dentistry, The University of Queensland, Brisbane, Queensland, 4006, Australia
- Centre for Orofacial Regeneration, Reconstruction and Rehabilitation (COR3), School of Dentistry, The University of Queensland, Brisbane, Queensland, 4006, Australia
| | - Xing Long
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) and Key Laboratory of Oral Biomedicine Ministry of Education, School and Hospital of Stomatology, Wuhan University, Wuhan, 430079, China
| | - Huang Li
- Department of Orthodontics, Nanjing Stomatological Hospital, Medical School of Nanjing University, Nanjing, Jiangsu, 210009, China
| | - Sašo Ivanovski
- School of Dentistry, The University of Queensland, Brisbane, Queensland, 4006, Australia
- Centre for Orofacial Regeneration, Reconstruction and Rehabilitation (COR3), School of Dentistry, The University of Queensland, Brisbane, Queensland, 4006, Australia
- Corresponding author. School of Dentistry, The University of Queensland, Brisbane, Queensland, 4006, Australia.
| | - Chun Xu
- School of Dentistry, The University of Queensland, Brisbane, Queensland, 4006, Australia
- Centre for Orofacial Regeneration, Reconstruction and Rehabilitation (COR3), School of Dentistry, The University of Queensland, Brisbane, Queensland, 4006, Australia
- Corresponding author. School of Dentistry, The University of Queensland, Brisbane, Queensland, 4006, Australia.
| |
Collapse
|
11
|
Paesa M, Alejo T, Garcia-Alvarez F, Arruebo M, Mendoza G. New insights in osteoarthritis diagnosis and treatment: Nano-strategies for an improved disease management. WILEY INTERDISCIPLINARY REVIEWS. NANOMEDICINE AND NANOBIOTECHNOLOGY 2023; 15:e1844. [PMID: 35965293 DOI: 10.1002/wnan.1844] [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: 12/02/2021] [Revised: 06/02/2022] [Accepted: 07/12/2022] [Indexed: 11/07/2022]
Abstract
Osteoarthritis (OA) is a common chronic joint pathology that has become a predominant cause of disability worldwide. Even though the origin and evolution of OA rely on different factors that are not yet elucidated nor understood, the development of novel strategies to treat OA has emerged in the last years. Cartilage degradation is the main hallmark of the pathology though alterations in bone and synovial inflammation, among other comorbidities, are also involved during OA progression. From a molecular point of view, a vast amount of signaling pathways are implicated in the progression of the disease, opening up a wide plethora of targets to attenuate or even halt OA. The main purpose of this review is to shed light on the recent strategies published based on nanotechnology for the early diagnosis of the disease as well as the most promising nano-enabling therapeutic approaches validated in preclinical models. To address the clinical issue, the key pathways involved in OA initiation and progression are described as the main potential targets for OA prevention and early treatment. Furthermore, an overview of current therapeutic strategies is depicted. Finally, to solve the drawbacks of current treatments, nanobiomedicine has shown demonstrated benefits when using drug delivery systems compared with the administration of the equivalent doses of the free drugs and the potential of disease-modifying OA drugs when using nanosystems. We anticipate that the development of smart and specific bioresponsive and biocompatible nanosystems will provide a solid and promising basis for effective OA early diagnosis and treatment. This article is categorized under: Diagnostic Tools > In Vivo Nanodiagnostics and Imaging Implantable Materials and Surgical Technologies > Nanotechnology in Tissue Repair and Replacement.
Collapse
Affiliation(s)
- Monica Paesa
- Department of Chemical Engineering, Aragon Institute of Nanoscience (INA), University of Zaragoza, Aragón Materials Science Institute, ICMA, Zaragoza, Spain
| | - Teresa Alejo
- Department of Chemical Engineering, Aragon Institute of Nanoscience (INA), University of Zaragoza, Aragón Materials Science Institute, ICMA, Zaragoza, Spain
- Health Research Institute Aragon (IIS Aragon), Zaragoza, Spain
| | - Felicito Garcia-Alvarez
- Health Research Institute Aragon (IIS Aragon), Zaragoza, Spain
- Hospital Clínico Universitario Lozano Blesa, Department of Orthopedic Surgery & Traumatology, University of Zaragoza, Zaragoza, Spain
| | - Manuel Arruebo
- Department of Chemical Engineering, Aragon Institute of Nanoscience (INA), University of Zaragoza, Aragón Materials Science Institute, ICMA, Zaragoza, Spain
- Health Research Institute Aragon (IIS Aragon), Zaragoza, Spain
- Networking Research Center on Bioengineering, Biomaterials and Nanomedicine, CIBER-BBN, Madrid, Spain
| | - Gracia Mendoza
- Health Research Institute Aragon (IIS Aragon), Zaragoza, Spain
- Networking Research Center on Bioengineering, Biomaterials and Nanomedicine, CIBER-BBN, Madrid, Spain
| |
Collapse
|
12
|
Tekade M, Pingale P, Gupta R, Pawar B, Tekade RK, Sharma MC. Recent Advances in Polymer-Based Nanomaterials for Non-Invasive Photothermal Therapy of Arthritis. Pharmaceutics 2023; 15:pharmaceutics15030735. [PMID: 36986596 PMCID: PMC10058747 DOI: 10.3390/pharmaceutics15030735] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Revised: 01/25/2023] [Accepted: 02/08/2023] [Indexed: 02/25/2023] Open
Abstract
To date, nanomaterials have been widely used for the treatment and diagnosis of rheumatoid arthritis. Amongst various nanomaterials, polymer-based nanomaterials are becoming increasingly popular in nanomedicine due to their functionalised fabrication and easy synthesis, making them biocompatible, cost-effective, biodegradable, and efficient nanocarriers for the delivery of drugs to a specific target cell. They act as photothermal reagents with high absorption in the near-infrared region that can transform near-infrared light into localised heat with fewer side effects, provide easier integration with existing therapies, and offer increased effectiveness. They have been combined with photothermal therapy to understand the chemical and physical activities behind the stimuli-responsiveness of polymer nanomaterials. In this review article, we provide detailed information regarding the recent advances in polymer nanomaterials for the non-invasive photothermal treatment of arthritis. The synergistic effect of polymer nanomaterials and photothermal therapy has enhanced the treatment and diagnosis of arthritis and reduced the side effects of drugs in the joint cavity. In addition, further novel challenges and future perspectives must be resolved to advance polymer nanomaterials for the photothermal therapy of arthritis.
Collapse
Affiliation(s)
- Muktika Tekade
- School of Pharmacy, Devi Ahilya Vishwavidyalaya, Takshila Campus, Khandwa Road, Indore 452001, Madhya Pradesh, India
- Correspondence: (M.T.); (R.K.T.)
| | - Prashant Pingale
- Department of Pharmaceutics, Sir Dr. M.S. Gosavi College of Pharmaceutical Education and Research, Nashik 422005, Maharashtra, India
| | - Rachna Gupta
- National Institute of Pharmaceutical Education and Research (NIPER), Ahmedabad, Palaj, Opp. Air Force Station, Gandhinagar 382355, Gujarat, India
| | - Bhakti Pawar
- National Institute of Pharmaceutical Education and Research (NIPER), Ahmedabad, Palaj, Opp. Air Force Station, Gandhinagar 382355, Gujarat, India
| | - Rakesh Kumar Tekade
- National Institute of Pharmaceutical Education and Research (NIPER), Ahmedabad, Palaj, Opp. Air Force Station, Gandhinagar 382355, Gujarat, India
- Correspondence: (M.T.); (R.K.T.)
| | - Mukesh Chandra Sharma
- School of Pharmacy, Devi Ahilya Vishwavidyalaya, Takshila Campus, Khandwa Road, Indore 452001, Madhya Pradesh, India
| |
Collapse
|
13
|
Nano-Based Co-Delivery System for Treatment of Rheumatoid Arthritis. Molecules 2022; 27:molecules27185973. [PMID: 36144709 PMCID: PMC9503141 DOI: 10.3390/molecules27185973] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Revised: 08/07/2022] [Accepted: 08/12/2022] [Indexed: 11/21/2022] Open
Abstract
A systemic autoimmune condition known as rheumatoid arthritis (RA) has a significant impact on patients’ quality of life. Given the complexity of RA’s biology, no single treatment can totally block the disease’s progression. The combined use of co-delivery regimens integrating various diverse mechanisms has been widely acknowledged as a way to make up for the drawbacks of single therapy. These days, co-delivery systems have been frequently utilized for co-treatment, getting over drug limitations, imaging of inflammatory areas, and inducing reactions. Various small molecules, nucleic acid drugs, and enzyme-like agents intended for co-delivery are frequently capable of producing the ability to require positive outcomes. In addition, the excellent response effect of phototherapeutic agents has led to their frequent use for delivery together with chemotherapeutics. In this review, we discuss different types of nano-based co-delivery systems and their advantages, limitations, and future directions. In addition, we review the prospects and predicted challenges for the combining of phototherapeutic agents with conventional drugs, hoping to provide some theoretical support for future in-depth studies of nano-based co-delivery systems and phototherapeutic agents.
Collapse
|
14
|
Li Y, Liang Q, Zhou L, Cao Y, Yang J, Li J, Liu J, Bi J, Liu Y. An ROS-responsive artesunate prodrug nanosystem co-delivers dexamethasone for rheumatoid arthritis treatment through the HIF-1α/NF-κB cascade regulation of ROS scavenging and macrophage repolarization. Acta Biomater 2022; 152:406-424. [DOI: 10.1016/j.actbio.2022.08.054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Revised: 07/17/2022] [Accepted: 08/23/2022] [Indexed: 11/01/2022]
|
15
|
Gargano G, Oliva F, Oliviero A, Maffulli N. Small interfering RNAs in the management of human rheumatoid arthritis. Br Med Bull 2022; 142:34-43. [PMID: 35488320 PMCID: PMC9351475 DOI: 10.1093/bmb/ldac012] [Citation(s) in RCA: 101] [Impact Index Per Article: 33.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 03/30/2022] [Indexed: 11/13/2022]
Abstract
BACKGROUND Rheumatoid arthritis (RA) has unclear pathogenesis, but the molecules that feed its inflammatory state are known. Small interfering RNAs (siRNAs) are useful to identify molecular targets and evaluate the efficacy of specific drugs, and can themselves be used for therapeutic purposes. SOURCES OF DATA A systematic search of different databases to March 2022 was performed to define the role of siRNAs in RA therapy. Twenty suitable studies were identified. AREAS OF AGREEMENT Small interfering RNAs can be useful in the study of inflammatory processes in RA, and identify possible therapeutic targets and drug therapies. AREAS OF CONTROVERSY Many genes and cytokines participate in the inflammatory process of RA and can be regulated with siRNA. However, it is difficult to determine whether the responses to siRNAs and other drugs studied in human cells in vitro are similar to the responses in vivo. GROWING POINTS Inflammatory processes can be affected by the gene dysregulation of siRNAs on inflammatory cytokines. AREAS TIMELY FOR DEVELOPING RESEARCH To date, it is not possible to determine whether the pharmacological response of siRNAs on cells in vitro would be similar to what takes place in vivo for the diseases studied so far.
Collapse
Affiliation(s)
- Giuseppe Gargano
- Department of Trauma and Orthopaedic Surgery, AOU San Giovanni di Dio e Ruggi D'Aragona, Via San Leonardo 1, Salerno 84131, Italy.,Department of Medicine, Surgery and Dentistry, University of Salerno, Via S. Allende, Baronissi SA 84081, Italy
| | - Francesco Oliva
- Department of Trauma and Orthopaedic Surgery, AOU San Giovanni di Dio e Ruggi D'Aragona, Via San Leonardo 1, Salerno 84131, Italy.,Department of Medicine, Surgery and Dentistry, University of Salerno, Via S. Allende, Baronissi SA 84081, Italy
| | - Antonio Oliviero
- Department of Trauma and Orthopaedic Surgery, AOU San Giovanni di Dio e Ruggi D'Aragona, Via San Leonardo 1, Salerno 84131, Italy.,Department of Medicine, Surgery and Dentistry, University of Salerno, Via S. Allende, Baronissi SA 84081, Italy
| | - Nicola Maffulli
- Department of Trauma and Orthopaedic Surgery, AOU San Giovanni di Dio e Ruggi D'Aragona, Via San Leonardo 1, Salerno 84131, Italy.,Department of Medicine, Surgery and Dentistry, University of Salerno, Via S. Allende, Baronissi SA 84081, Italy.,Queen Mary University of London, Barts and the London School of Medicine and Dentistry, Centre for Sports and Exercise Medicine, Mile End Hospital, 275 Bancroft Road, London E1 4DG, UK.,School of Pharmacy and Bioengineering, Keele University School of Medicine, Thornburrow Drive, Stoke on Trent, ST4 7QB, UK
| |
Collapse
|
16
|
Polymer nanotherapeutics to correct autoimmunity. J Control Release 2022; 343:152-174. [PMID: 34990701 DOI: 10.1016/j.jconrel.2021.12.036] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Revised: 12/24/2021] [Accepted: 12/28/2021] [Indexed: 12/11/2022]
Abstract
The immune system maintains homeostasis and protects the body from pathogens, mutated cells, and other harmful substances. When immune homeostasis is disrupted, excessive autoimmunity will lead to diseases. To inhibit the unexpected immune responses and reduce the impact of treatment on immunoprotective functions, polymer nanotherapeutics, such as nanomedicines, nanovaccines, and nanodecoys, were developed as part of an advanced strategy for precise immunomodulation. Nanomedicines transport cytotoxic drugs to target sites to reduce the occurrence of side effects and increase the stability and bioactivity of various immunomodulating agents, especially nucleic acids and cytokines. In addition, polymer nanomaterials carrying autoantigens used as nanovaccines can induce antigen-specific immune tolerance without interfering with protective immune responses. The precise immunomodulatory function of nanovaccines has broad prospects for the treatment of immune related-diseases. Besides, nanodecoys, which are designed to protect the body from various pathogenic substances by intravenous administration, are a simple and relatively noninvasive treatment. Herein, we have discussed and predicted the application of polymer nanotherapeutics in the correction of autoimmunity, including treating autoimmune diseases, controlling hypersensitivity, and avoiding transplant rejection.
Collapse
|
17
|
Wang P, Wu M, Li R, Cai Z, Zhang H. Fabrication of a Double-Network Hydrogel Based on Carboxymethylated Curdlan/Polyacrylamide with Highly Mechanical Performance for Cartilage Repair. ACS APPLIED POLYMER MATERIALS 2021; 3:5857-5869. [DOI: 10.1021/acsapm.1c01094] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
Affiliation(s)
- Pengguang Wang
- Advanced Rheology Institute, Department of Polymer Science and Engineering, School of Chemistry and Chemical Engineering, Frontiers Science Center for Transformative Molecules, Shanghai Key Laboratory of Electrical Insulation and Thermal Aging, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Min Wu
- Advanced Rheology Institute, Department of Polymer Science and Engineering, School of Chemistry and Chemical Engineering, Frontiers Science Center for Transformative Molecules, Shanghai Key Laboratory of Electrical Insulation and Thermal Aging, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Ruiqi Li
- Advanced Rheology Institute, Department of Polymer Science and Engineering, School of Chemistry and Chemical Engineering, Frontiers Science Center for Transformative Molecules, Shanghai Key Laboratory of Electrical Insulation and Thermal Aging, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Zhixiang Cai
- Advanced Rheology Institute, Department of Polymer Science and Engineering, School of Chemistry and Chemical Engineering, Frontiers Science Center for Transformative Molecules, Shanghai Key Laboratory of Electrical Insulation and Thermal Aging, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Hongbin Zhang
- Advanced Rheology Institute, Department of Polymer Science and Engineering, School of Chemistry and Chemical Engineering, Frontiers Science Center for Transformative Molecules, Shanghai Key Laboratory of Electrical Insulation and Thermal Aging, Shanghai Jiao Tong University, Shanghai 200240, China
| |
Collapse
|
18
|
Li X, Dai B, Guo J, Zheng L, Guo Q, Peng J, Xu J, Qin L. Nanoparticle-Cartilage Interaction: Pathology-Based Intra-articular Drug Delivery for Osteoarthritis Therapy. NANO-MICRO LETTERS 2021; 13:149. [PMID: 34160733 PMCID: PMC8222488 DOI: 10.1007/s40820-021-00670-y] [Citation(s) in RCA: 64] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Accepted: 05/19/2021] [Indexed: 05/03/2023]
Abstract
Osteoarthritis is the most prevalent chronic and debilitating joint disease, resulting in huge medical and socioeconomic burdens. Intra-articular administration of agents is clinically used for pain management. However, the effectiveness is inapparent caused by the rapid clearance of agents. To overcome this issue, nanoparticles as delivery systems hold considerable promise for local control of the pharmacokinetics of therapeutic agents. Given the therapeutic programs are inseparable from pathological progress of osteoarthritis, an ideal delivery system should allow the release of therapeutic agents upon specific features of disorders. In this review, we firstly introduce the pathological features of osteoarthritis and the design concept for accurate localization within cartilage for sustained drug release. Then, we review the interactions of nanoparticles with cartilage microenvironment and the rational design. Furthermore, we highlight advances in the therapeutic schemes according to the pathology signals. Finally, armed with an updated understanding of the pathological mechanisms, we place an emphasis on the development of "smart" bioresponsive and multiple modality nanoparticles on the near horizon to interact with the pathological signals. We anticipate that the exploration of nanoparticles by balancing the efficacy, safety, and complexity will lay down a solid foundation tangible for clinical translation.
Collapse
Affiliation(s)
- Xu Li
- Musculoskeletal Research Laboratory, Department of Orthopedics and Traumatology, The Chinese University of Hong Kong, Sha Tin, Hong Kong, SAR, People's Republic of China
- Joint Laboratory of Chinese Academic of Science and Hong Kong for Biomaterials, The Chinese University of Hong Kong, Sha Tin, Hong Kong, SAR, People's Republic of China
- Innovative Orthopedic Biomaterial and Drug Translational Research Laboratory, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Sha Tin, Hong Kong, SAR, People's Republic of China
| | - Bingyang Dai
- Musculoskeletal Research Laboratory, Department of Orthopedics and Traumatology, The Chinese University of Hong Kong, Sha Tin, Hong Kong, SAR, People's Republic of China
- Joint Laboratory of Chinese Academic of Science and Hong Kong for Biomaterials, The Chinese University of Hong Kong, Sha Tin, Hong Kong, SAR, People's Republic of China
- Innovative Orthopedic Biomaterial and Drug Translational Research Laboratory, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Sha Tin, Hong Kong, SAR, People's Republic of China
| | - Jiaxin Guo
- Musculoskeletal Research Laboratory, Department of Orthopedics and Traumatology, The Chinese University of Hong Kong, Sha Tin, Hong Kong, SAR, People's Republic of China
- Joint Laboratory of Chinese Academic of Science and Hong Kong for Biomaterials, The Chinese University of Hong Kong, Sha Tin, Hong Kong, SAR, People's Republic of China
- Innovative Orthopedic Biomaterial and Drug Translational Research Laboratory, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Sha Tin, Hong Kong, SAR, People's Republic of China
| | - Lizhen Zheng
- Musculoskeletal Research Laboratory, Department of Orthopedics and Traumatology, The Chinese University of Hong Kong, Sha Tin, Hong Kong, SAR, People's Republic of China
- Joint Laboratory of Chinese Academic of Science and Hong Kong for Biomaterials, The Chinese University of Hong Kong, Sha Tin, Hong Kong, SAR, People's Republic of China
- Innovative Orthopedic Biomaterial and Drug Translational Research Laboratory, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Sha Tin, Hong Kong, SAR, People's Republic of China
| | - Quanyi Guo
- Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma and War Injuries PLA, Institute of Orthopedics, Chinese PLA General Hospital, Beijing, People's Republic of China
| | - Jiang Peng
- Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma and War Injuries PLA, Institute of Orthopedics, Chinese PLA General Hospital, Beijing, People's Republic of China
| | - Jiankun Xu
- Musculoskeletal Research Laboratory, Department of Orthopedics and Traumatology, The Chinese University of Hong Kong, Sha Tin, Hong Kong, SAR, People's Republic of China.
- Joint Laboratory of Chinese Academic of Science and Hong Kong for Biomaterials, The Chinese University of Hong Kong, Sha Tin, Hong Kong, SAR, People's Republic of China.
- Innovative Orthopedic Biomaterial and Drug Translational Research Laboratory, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Sha Tin, Hong Kong, SAR, People's Republic of China.
| | - Ling Qin
- Musculoskeletal Research Laboratory, Department of Orthopedics and Traumatology, The Chinese University of Hong Kong, Sha Tin, Hong Kong, SAR, People's Republic of China.
- Joint Laboratory of Chinese Academic of Science and Hong Kong for Biomaterials, The Chinese University of Hong Kong, Sha Tin, Hong Kong, SAR, People's Republic of China.
- Innovative Orthopedic Biomaterial and Drug Translational Research Laboratory, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Sha Tin, Hong Kong, SAR, People's Republic of China.
| |
Collapse
|
19
|
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: 13] [Impact Index Per Article: 3.3] [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
|
20
|
Zhao M, Zhu T, Chen J, Cui Y, Zhang X, Lee RJ, Sun F, Li Y, Teng L. PLGA/PCADK composite microspheres containing hyaluronic acid-chitosan siRNA nanoparticles: A rational design for rheumatoid arthritis therapy. Int J Pharm 2021; 596:120204. [PMID: 33493604 DOI: 10.1016/j.ijpharm.2021.120204] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2020] [Revised: 12/14/2020] [Accepted: 01/01/2021] [Indexed: 12/27/2022]
Abstract
Myeloid cell leukemia-1 (Mcl-1), a member of the Bcl-2 anti-apoptotic family, is overexpressed in the synovial macrophages of patients with rheumatoid arthritis (RA). Small interfering RNA (siRNA) Mcl-1 can induce macrophage apoptosis in the joints and is a potential therapeutic target of RA. Nevertheless, the application of siRNA is limited owing to its instability and susceptibility to degradation in vivo. To address these shortcomings, we developed composite microspheres (MPs) loaded with hyaluronic acid (HA)-chitosan (CS) nanoparticles (NPs). First, we synthesized HA-CS/siRNA NPs (HCNPs) using ionotropic gelation process. Then, HCNPs, as an internal aqueous phase, were loaded into poly (D, L-lactide-co-glycolide) (PLGA) and poly (cyclohexane-1,4-diyl acetone dimethylene ketal) (PCADK) MPs using the double emulsion method. The NPs-in-MPs (NiMPs) composite system provided sustained release of NPs, protected siRNA against nuclease degradation in the serum, and could readily cross the cellular membrane. In addition, we evaluated the advantages of NiMPs in an adjuvant-induced arthritis rat model. Our experimental results demonstrate that NiMPs have greater pharmacodynamic effects than common MPs. Meanwhile, compared with HCNPs, NiMPs reduced the frequency of drug administration. Therefore, NiMPs are a promising and novel siRNA delivery vehicle for RA therapy.
Collapse
Affiliation(s)
- Menghui Zhao
- School of Life Sciences, Jilin University, Changchun, Jilin, China
| | - Tianyu Zhu
- School of Life Sciences, Jilin University, Changchun, Jilin, China
| | - Jicong Chen
- School of Life Sciences, Jilin University, Changchun, Jilin, China
| | - Yaxin Cui
- School of Life Sciences, Jilin University, Changchun, Jilin, China
| | - Xueyan Zhang
- School of Life Sciences, Jilin University, Changchun, Jilin, China
| | - Robert J Lee
- School of Life Sciences, Jilin University, Changchun, Jilin, China; College of Pharmacy, the Ohio State University, Columbus, OH, USA
| | - Fengying Sun
- School of Life Sciences, Jilin University, Changchun, Jilin, China
| | - Youxin Li
- School of Life Sciences, Jilin University, Changchun, Jilin, China.
| | - Lesheng Teng
- School of Life Sciences, Jilin University, Changchun, Jilin, China.
| |
Collapse
|
21
|
Zheng M, Jia H, Wang H, Liu L, He Z, Zhang Z, Yang W, Gao L, Gao X, Gao F. Application of nanomaterials in the treatment of rheumatoid arthritis. RSC Adv 2021; 11:7129-7137. [PMID: 35423287 PMCID: PMC8695100 DOI: 10.1039/d1ra00328c] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Accepted: 02/02/2021] [Indexed: 12/20/2022] Open
Abstract
Rheumatoid Arthritis (RA) is a chronic autoimmune disease, which mainly causes inflammation of the synovial joints and destruction of cartilage and bone tissue. At present, a variety of clinical drugs have been applied in the treatment of rheumatoid arthritis. With the development of nanotechnology, more and more nano-drugs have been applied in the treatment of rheumatoid arthritis due to the unique physical and chemical properties of nanomaterials. Treatment of RA with nanomaterials can improve bioavailability and selectively target damaged joint tissue. In this review, we summarized the progress of the application of nanomaterials in the treatment of rheumatoid arthritis and also proposed challenges faced by nanomaterials in the treatment of rheumatoid arthritis.
Collapse
Affiliation(s)
- Miaomiao Zheng
- CAS Key Laboratory for the Biological Effects of Nanomaterials and Nanosafety, Institute of High Energy Physics, Chinese Academy of Sciences Beijing 100049 China
- School of Pharmacy, Hebei University Baoding 071002 China
| | - Huiju Jia
- CAS Key Laboratory for the Biological Effects of Nanomaterials and Nanosafety, Institute of High Energy Physics, Chinese Academy of Sciences Beijing 100049 China
- School of Pharmacy, Hebei University Baoding 071002 China
| | - Huangwei Wang
- CAS Key Laboratory for the Biological Effects of Nanomaterials and Nanosafety, Institute of High Energy Physics, Chinese Academy of Sciences Beijing 100049 China
- School of Pharmacy, Hebei University Baoding 071002 China
| | - Linhong Liu
- CAS Key Laboratory for the Biological Effects of Nanomaterials and Nanosafety, Institute of High Energy Physics, Chinese Academy of Sciences Beijing 100049 China
| | - Zhesheng He
- CAS Key Laboratory for the Biological Effects of Nanomaterials and Nanosafety, Institute of High Energy Physics, Chinese Academy of Sciences Beijing 100049 China
- University of Chinese Academy of Science Beijing 100049 China
| | - Zhiyong Zhang
- CAS Key Laboratory for the Biological Effects of Nanomaterials and Nanosafety, Institute of High Energy Physics, Chinese Academy of Sciences Beijing 100049 China
| | - Wenzhi Yang
- School of Pharmacy, Hebei University Baoding 071002 China
| | - Liang Gao
- Department of Chemistry and Biology, Beijing University of Technology Beijing 100124 China
| | - Xueyun Gao
- Department of Chemistry and Biology, Beijing University of Technology Beijing 100124 China
| | - Fuping Gao
- CAS Key Laboratory for the Biological Effects of Nanomaterials and Nanosafety, Institute of High Energy Physics, Chinese Academy of Sciences Beijing 100049 China
| |
Collapse
|
22
|
Nanoparticle-siRNA: A potential strategy for rheumatoid arthritis therapy? J Control Release 2020; 325:380-393. [PMID: 32653501 DOI: 10.1016/j.jconrel.2020.07.006] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Revised: 07/05/2020] [Accepted: 07/07/2020] [Indexed: 02/07/2023]
Abstract
Rheumatoid arthritis (RA) is a common clinical inflammatory disease of the autoimmune system manifested by persistent synovitis, cartilage damage and even deformities. Despite significant progress in the clinical treatment of RA, long-term administration of anti-rheumatic drugs can cause a series of problems, including infections, gastrointestinal reactions, and abnormal liver and kidney functions. The emergence of RNA interference (RNAi) drugs has brought new hope for the treatment of RA. Designing a reasonable vector for RNAi drugs will greatly expand the application prospects of RNAi. Nanoparticles as a promising drug carrier provide reliable support for RNAi drugs. The review summarizes the pathogenesis of RA as a possible target for small interference RNA (siRNA) design. At the same time, the review also analyzes the nanoparticles used in siRNA carriers in recent years, laying the foundation and prospect for the next step in the development of intelligent nanocarriers.
Collapse
|
23
|
Wei F, Lang Y, Shen Q, Xu L, Cheng N, Chu Y, Lyu H, Chen F. Osteopontin-loaded PLGA nanoparticles enhance the intestinal mucosal barrier and alleviate inflammation via the NF-κB signaling pathway. Colloids Surf B Biointerfaces 2020; 190:110952. [PMID: 32172161 DOI: 10.1016/j.colsurfb.2020.110952] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2019] [Revised: 03/05/2020] [Accepted: 03/06/2020] [Indexed: 01/20/2023]
Abstract
Osteopontin is a multifunctional glycoprotein that is secreted by a variety of tissues or cells, but the role of osteopontin in the epithelial mucosal barrier has not been clearly established. We loaded osteopontin into hyaluronic acid-functionalized polymeric nanoparticles, which were administered by gavage to a colitis mouse model. The disease activity index, weight gain and colon length were calculated to assess the degree of symptoms. Epithelial permeability was measured using fluorescein isothiocyanate-conjugated dextran. The enzymatic activity of myeloperoxidase in the colon and inflammatory cytokines were assayed to assess the levels of inflammation. The histological appearance of the colon was observed by H&E staining. Tight junction proteins and signaling pathway proteins (NF-κB and phospho-NF-κB) were determined by western blotting. The resultant spherical osteopontin-loaded nanoparticles were characterized by the expected particle size (approximately 272.3 nm) and a slightly negative zeta potential (approximately -5.3 mV). Interestingly, we found that the osteopontin-loaded nanoparticles exerted remedial effects on colitis by both enhancing the intestinal barrier and alleviating inflammation in vivo according to the tested parameters. These results suggest that OPN plays a positive role in protecting the epithelial mucosal barrier and may be a therapeutic drug in gut homeostasis.
Collapse
Affiliation(s)
- Feng Wei
- Department of Gastroenterology, Shanghai Fifth People's Hospital, Fudan University, 128 Ruili Road, Shanghai 200240, People's Republic of China
| | - Yuhuang Lang
- Department of Emergency, Shanghai Fifth People's Hospital, Fudan University, 128 Ruili Road, Shanghai 200240, People's Republic of China
| | - Qiang Shen
- Department of Gastroenterology, Shanghai Fifth People's Hospital, Fudan University, 128 Ruili Road, Shanghai 200240, People's Republic of China
| | - Lin Xu
- Department of Gastroenterology, Shanghai Fifth People's Hospital, Fudan University, 128 Ruili Road, Shanghai 200240, People's Republic of China
| | - Nuo Cheng
- Department of Gastroenterology, Shanghai Fifth People's Hospital, Fudan University, 128 Ruili Road, Shanghai 200240, People's Republic of China
| | - Yan Chu
- Department of Gastroenterology, Shanghai Fifth People's Hospital, Fudan University, 128 Ruili Road, Shanghai 200240, People's Republic of China
| | - Hongwei Lyu
- Department of Gastroenterology, Shanghai Fifth People's Hospital, Fudan University, 128 Ruili Road, Shanghai 200240, People's Republic of China
| | - Fengyuan Chen
- Department of Gastroenterology, Shanghai Fifth People's Hospital, Fudan University, 128 Ruili Road, Shanghai 200240, People's Republic of China.
| |
Collapse
|
24
|
Xu XL, Lu KJ, Yao XQ, Ying XY, Du YZ. Stimuli-responsive Drug Delivery Systems as an Emerging Platform for Treatment of Rheumatoid Arthritis. Curr Pharm Des 2020; 25:155-165. [PMID: 30907308 DOI: 10.2174/1381612825666190321104424] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2019] [Accepted: 03/16/2019] [Indexed: 12/21/2022]
Abstract
Rheumatoid Arthritis (RA) is a systemic autoimmune disease accompanied by chronic inflammation. Due to the long-term infiltration in inflammatory sites, joints get steadily deteriorated, eventually resulting in functional incapacitation and disability. Despite the considerable effect, RA sufferers treated with current drug therapeutic efficacy are exposed to severe side effects. Application of Drug Delivery Systems (DDS) has improved these situations while the problem of limited drug exposure remains untackled. Stimuli-responsive DDS that are responsive to a variety of endogenous and exogenous stimuli, such as pH, redox status, and temperature, have emerged as a promising therapeutic strategy to optimize the drug release. Herein, we discussed the therapeutic regimes and serious side effects of current RA therapy, as well as focused on some of the potential stimuliresponsive DDS utilized in RA therapy. Besides, the prospective room in designing DDS for RA treatment has also been discussed.
Collapse
Affiliation(s)
- Xiao-Ling Xu
- Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Kong-Jun Lu
- Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Xiao-Qin Yao
- School of Medicine, Zhejiang University City College, Hangzhou 310058, China
| | - Xiao-Ying Ying
- Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Yong-Zhong Du
- Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| |
Collapse
|
25
|
Leng Q, Chen L, Lv Y. RNA-based scaffolds for bone regeneration: application and mechanisms of mRNA, miRNA and siRNA. Am J Cancer Res 2020; 10:3190-3205. [PMID: 32194862 PMCID: PMC7053199 DOI: 10.7150/thno.42640] [Citation(s) in RCA: 82] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2019] [Accepted: 01/16/2020] [Indexed: 02/07/2023] Open
Abstract
Globally, more than 1.5 million patients undergo bone graft surgeries annually, and the development of biomaterial scaffolds that mimic natural bone for bone grafting remains a tremendous challenge. In recent decades, due to the improved understanding of the mechanisms of bone remodeling and the rapid development of gene therapy, RNA (including messenger RNA (mRNA), microRNA (miRNA), and short interfering RNA (siRNA)) has attracted increased attention as a new tool for bone tissue engineering due to its unique nature and great potential to cure bone defects. Different types of RNA play roles via a variety of mechanisms in bone-related cells in vivo as well as after synthesis in vitro. In addition, RNAs are delivered to injured sites by loading into scaffolds or systemic administration after combination with vectors for bone tissue engineering. However, the challenge of effectively and stably delivering RNA into local tissue remains to be solved. This review describes the mechanisms of the three types of RNAs and the application of the relevant types of RNA delivery vectors and scaffolds in bone regeneration. The improvements in their development are also discussed.
Collapse
|
26
|
Jiang S, Prozeller D, Pereira J, Simon J, Han S, Wirsching S, Fichter M, Mottola M, Lieberwirth I, Morsbach S, Mailänder V, Gehring S, Crespy D, Landfester K. Controlling protein interactions in blood for effective liver immunosuppressive therapy by silica nanocapsules. NANOSCALE 2020; 12:2626-2637. [PMID: 31939969 DOI: 10.1039/c9nr09879h] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/21/2023]
Abstract
Immunosuppression with glucocorticoids is a common treatment for autoimmune liver diseases and after liver transplant, which is however associated with severe side-effects. Targeted delivery of glucocorticoids to inflammatory cells, e.g. liver macrophages and Kupffer cells, is a promising approach for minimizing side effects. Herein, we prepare core-shell silica nanocapsules (SiO2 NCs) via a sol-gel process confined in nanodroplets for targeted delivery of dexamethasone (DXM) for liver immunosuppressive therapy. DXM with concentrations up to 100 mg mL-1 in olive oil are encapsulated while encapsulation efficiency remains over 95% after 15 days. Internalization of NCs by non-parenchymal murine liver cells significantly reduces the release of inflammatory cytokines, indicating an effective suppression of inflammatory response of liver macrophages. Fluorescent and magnetic labeling of the NCs allows for monitoring their intracellular trafficking and biodegradation. Controlled interaction with blood proteins and good colloidal stability in blood plasma are achieved via PEGylation of the NCs. Specific proteins responsible for stealth effect, such as apolipoprotein A-I, apolipoprotein A-IV, and clusterin, are present in large amounts on the PEGylated NCs. In vivo biodistribution investigations prove an efficient accumulation of NCs in the liver, underlining the suitability of the SiO2 NCs as a dexamethasone carrier for treating inflammatory liver diseases.
Collapse
Affiliation(s)
- Shuai Jiang
- Max Planck Institute for Polymer Research, Ackermannweg 10, 55128 Mainz, Germany.
| | - Domenik Prozeller
- Max Planck Institute for Polymer Research, Ackermannweg 10, 55128 Mainz, Germany.
| | - Jorge Pereira
- Max Planck Institute for Polymer Research, Ackermannweg 10, 55128 Mainz, Germany.
| | - Johanna Simon
- Max Planck Institute for Polymer Research, Ackermannweg 10, 55128 Mainz, Germany. and Dermatology Clinic, University Medical Center of the Johannes Gutenberg-University, Langenbeckstr. 1, 55131 Mainz, Germany
| | - Shen Han
- Max Planck Institute for Polymer Research, Ackermannweg 10, 55128 Mainz, Germany.
| | - Sebastian Wirsching
- Children's Hospital, University Medical Center of the Johannes-Gutenberg University, Mainz, Germany
| | - Michael Fichter
- Children's Hospital, University Medical Center of the Johannes-Gutenberg University, Mainz, Germany
| | - Milagro Mottola
- Max Planck Institute for Polymer Research, Ackermannweg 10, 55128 Mainz, Germany.
| | - Ingo Lieberwirth
- Max Planck Institute for Polymer Research, Ackermannweg 10, 55128 Mainz, Germany.
| | - Svenja Morsbach
- Max Planck Institute for Polymer Research, Ackermannweg 10, 55128 Mainz, Germany.
| | - Volker Mailänder
- Max Planck Institute for Polymer Research, Ackermannweg 10, 55128 Mainz, Germany. and Dermatology Clinic, University Medical Center of the Johannes Gutenberg-University, Langenbeckstr. 1, 55131 Mainz, Germany
| | - Stephan Gehring
- Children's Hospital, University Medical Center of the Johannes-Gutenberg University, Mainz, Germany
| | - Daniel Crespy
- Max Planck Institute for Polymer Research, Ackermannweg 10, 55128 Mainz, Germany. and Department of Materials Science and Engineering, School of Molecular Science and Engineering, Vidyasirimedhi Institute of Science and Technology (VISTEC), Rayong 21210, Thailand
| | - Katharina Landfester
- Max Planck Institute for Polymer Research, Ackermannweg 10, 55128 Mainz, Germany.
| |
Collapse
|
27
|
Nanomaterials for direct and indirect immunomodulation: A review of applications. Eur J Pharm Sci 2020; 142:105139. [DOI: 10.1016/j.ejps.2019.105139] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2019] [Revised: 10/14/2019] [Accepted: 11/03/2019] [Indexed: 01/03/2023]
|
28
|
Li X, Qu J, Zhang T, He X, Jiang Y, Chen J. Nuclear Factor kappa B (NF-κB) Targeted Self-Assembled Nanoparticles Loaded with Methotrexate for Treatment of Rheumatoid Arthritis. Med Sci Monit 2019; 25:8204-8212. [PMID: 31674342 PMCID: PMC6849372 DOI: 10.12659/msm.917396] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Background Nanotechnology is one of the most productive approaches for specifically delivering drug payloads to the region of interest to decrease nonspecific distribution and unwanted toxicities. Material/Methods We prepared glycol chitosan stearate self-assembled nanoparticles loaded with methotrexate (MTX) for NF-κB targeting in treatment of rheumatoid arthritis (RA). The nanoparticles were prepared using hydrophobic modification of glycol chitosan (GC) with steric acid (SA) and was characterized using IR. The efficiency of nanoparticles after their physiochemical characterization was measured in vitro and by in vivo studies in mice. Results The nanoparticles thus prepared were spherical in shape, 235 nm in diameter, and had negative zeta potential. The entrapment efficiency of MTX-GC-SA was more than 70%. The in vitro higher uptake of MTX-GC-SA in murine macrophage cells (RAW 264.7) was confirmed using confocal microscopy and FACS analysis. Systemic administration of MTX-GC-SA into collagen-induced arthritis (CIA) mice resulted in high accumulation in inflamed joints. The MTX-GS-SA revealed significantly better therapeutic efficacy against CIA mice compared to free MTX. Conclusions These findings highlight the potential of using this MTX-GC-SA nanoparticle formulation in suppressing inflammatory arthritis for effective treatment of RA.
Collapse
Affiliation(s)
- Xiong Li
- Department of Sports Medicine, Xiangya Hospital Central South University, Changsha, Hunan, China (mainland)
| | - Jin Qu
- Department of Sports Medicine, Xiangya Hospital Central South University, Changsha, Hunan, China (mainland)
| | - Tao Zhang
- Department of Sports Medicine, Xiangya Hospital Central South University, Changsha, Hunan, China (mainland)
| | - Xi He
- Department of Sports Medicine, Xiangya Hospital Central South University, Changsha, Hunan, China (mainland)
| | - Ying Jiang
- Department of Rheumatology, Xiangya Hospital Central South University, Changsha, Hunan, China (mainland)
| | - Jiangyan Chen
- Department of Rheumatology, Xiangya Hospital Central South University, Changsha, Hunan, China (mainland)
| |
Collapse
|
29
|
Chen X, Zhu X, Ma L, Lin A, Gong Y, Yuan G, Liu J. A core-shell structure QRu-PLGA-RES-DS NP nanocomposite with photothermal response-induced M2 macrophage polarization for rheumatoid arthritis therapy. NANOSCALE 2019; 11:18209-18223. [PMID: 31560010 DOI: 10.1039/c9nr05922a] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/23/2023]
Abstract
Rheumatoid arthritis (RA) is a degenerative joint disease caused by autoimmunity; for the effective treatment of RA while avoiding the side effects of conventional drugs, we have proposed a new therapeutic strategy to eliminate the inflammatory response in RA by regulating the immune system that promotes the transformation of M1-type macrophages to M2-type macrophages. Herein, we designed and synthesized a core-shell nanocomposite (QRu-PLGA-RES-DS NPs), which showed an effective therapeutic effect on RA by accurately inducing the polarization of M2 macrophages. In this system, the quadrilateral ruthenium nanoparticles (QRuNPs) with a photothermal effect were utilized as a core and the thermosensitive molecular poly (lactic-co-glycolic acid) (PLGA) modified with the targeted molecule dextran sulfate (DS) was employed as a shell. Then, the nanocarrier QRu-PLGA-DS NPs effectively improved the water solubility and targeting of resveratrol (RES) through self-assembly. Therefore, the QRu-PLGA-RES-DS NPs significantly enhanced the ability of RES to reverse the M1 type macrophages to the M2 type macrophages through an accurate release. In vivo experiments further demonstrated that the QRu-PLGA-RES-DS NPs could effectively accumulate in the lesion area with an exogenous stimulus, and this significantly enhanced the transformation of the M2 type macrophages and decreased the recruitment of the M1 type macrophages. Furthermore, the QRu-PLGA-RES-DS NPs effectively treated RA by eliminating the inflammatory response; in addition, photoacoustic imaging (PA) of the QRu NPs provided image guidance for the distribution and analysis of nanomedicine in inflammatory tissues. Hence, this therapeutic strategy promotes the biological applications of Ru-based nanoparticles in disease treatment.
Collapse
Affiliation(s)
- Xu Chen
- College of Chemistry and Materials Science, Jinan University, 510632, P. R. China.
| | | | | | | | | | | | | |
Collapse
|
30
|
García-Couce J, Almirall A, Fuentes G, Kaijzel E, Chan A, Cruz LJ. Targeting Polymeric Nanobiomaterials as a Platform for Cartilage Tissue Engineering. Curr Pharm Des 2019; 25:1915-1932. [DOI: 10.2174/1381612825666190708184745] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2019] [Accepted: 06/26/2019] [Indexed: 01/05/2023]
Abstract
Articular cartilage is a connective tissue structure that is found in anatomical areas that are important for the movement of the human body. Osteoarthritis is the ailment that most often affects the articular cartilage. Due to its poor intrinsic healing capacity, damage to the articular cartilage is highly detrimental and at present the reconstructive options for its repair are limited. Tissue engineering and the science of nanobiomaterials are two lines of research that together can contribute to the restoration of damaged tissue. The science of nanobiomaterials focuses on the development of different nanoscale structures that can be used as carriers of drugs / cells to treat and repair damaged tissues such as articular cartilage. This review article is an overview of the composition of articular cartilage, the causes and treatments of osteoarthritis, with a special emphasis on nanomaterials as carriers of drugs and cells, which reduce inflammation, promote the activation of biochemical factors and ultimately contribute to the total restoration of articular cartilage.
Collapse
Affiliation(s)
- Jomarien García-Couce
- Translational Nanobiomaterials and Imaging (TNI) group, Radiology department, Leiden University Medical Centrum, Leiden, Netherlands
| | - Amisel Almirall
- Translational Nanobiomaterials and Imaging (TNI) group, Radiology department, Leiden University Medical Centrum, Leiden, Netherlands
| | - Gastón Fuentes
- Translational Nanobiomaterials and Imaging (TNI) group, Radiology department, Leiden University Medical Centrum, Leiden, Netherlands
| | - Eric Kaijzel
- Translational Nanobiomaterials and Imaging (TNI) group, Radiology department, Leiden University Medical Centrum, Leiden, Netherlands
| | - Alan Chan
- Percuros B.V., Zernikedreef 8, 2333 CL Leiden, Netherlands
| | - Luis J. Cruz
- Translational Nanobiomaterials and Imaging (TNI) group, Radiology department, Leiden University Medical Centrum, Leiden, Netherlands
| |
Collapse
|
31
|
Ansari S, Karkhaneh A, Bonakdar S, Haghighipour N. Simultaneous effects of hydrostatic pressure and dexamethasone release from electrospun fibers on inflammation-induced chondrocytes. Eur Polym J 2019; 118:244-253. [DOI: 10.1016/j.eurpolymj.2019.06.003] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
|
32
|
Hao F, Lee RJ, Zhong L, Dong S, Yang C, Teng L, Meng Q, Lu J, Xie J, Teng L. Hybrid micelles containing methotrexate-conjugated polymer and co-loaded with microRNA-124 for rheumatoid arthritis therapy. Theranostics 2019; 9:5282-5297. [PMID: 31410215 PMCID: PMC6691571 DOI: 10.7150/thno.32268] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2018] [Accepted: 05/29/2019] [Indexed: 12/13/2022] Open
Abstract
Purpose: Methotrexate (MTX) is a first-line drug for rheumatoid arthritis (RA)therapy. However, MTX monotherapy often results in irreversible joint damage due to its slow onset of action and long duration. microRNA-124 (miR-124) has shown direct bone protection activity against RA. A co-delivery system for MTX and microRNA combination may provide therapeutic synergy. Methods: Methotrexate-conjugated polymer hybrid micelles (M-PHMs) were prepared by self-assembly of two functional amphiphilic polymers (MTX-PEI-LA and mPEG-LA) at an optimized weight ratio. Incorporation of microRNA was achieved through electrostatic interactions between microRNA and cationic polymer MTX-PEI-LA. Cellular uptake, endosome escape, biodistribution, and therapeutic efficacy of M-PHMs/miR-124 complexes were investigated and evaluated in RAW264.7 cells and a rat adjuvant-induced arthritis (AIA) model. Results: M-PHMs/miR-124 complexes exhibited folate receptor-mediated uptake in activated RAW264.7 cells. miR-124 was able to escape from the endosome and down-regulate nuclear factor of activated T cells cytoplasmic1 (NFATc1). M-PHMs/miR-124 complexes accumulated in inflamed joints of AIA rats and showed superior therapeutic efficacy through both anti-inflammatory effect and direct bone protective effect. Combination of miR-124 and MTX in these micelles induced disease remission. Conclusions: M-PHMs/miR-124 was highly effective against RA through therapeutic synergy. Additional studies are warranted to further investigate its therapeutic potential and delineate its mechanisms of action.
Collapse
Affiliation(s)
- Fei Hao
- School of Life Sciences, Jilin University, No.2699, Qianjin Street, Changchun130012, P.R. China
| | - Robert J Lee
- School of Life Sciences, Jilin University, No.2699, Qianjin Street, Changchun130012, P.R. China
- College of Pharmacy, The Ohio State University, Columbus, 500 W 12th Ave, Columbus, OH 43210, USA
| | - Lihuang Zhong
- School of Life Sciences, Jilin University, No.2699, Qianjin Street, Changchun130012, P.R. China
| | - Shiyan Dong
- School of Life Sciences, Jilin University, No.2699, Qianjin Street, Changchun130012, P.R. China
| | - Chunmiao Yang
- School of Life Sciences, Jilin University, No.2699, Qianjin Street, Changchun130012, P.R. China
| | - Lirong Teng
- School of Life Sciences, Jilin University, No.2699, Qianjin Street, Changchun130012, P.R. China
| | - Qingfan Meng
- School of Life Sciences, Jilin University, No.2699, Qianjin Street, Changchun130012, P.R. China
| | - Jiahui Lu
- School of Life Sciences, Jilin University, No.2699, Qianjin Street, Changchun130012, P.R. China
| | - Jing Xie
- School of Life Sciences, Jilin University, No.2699, Qianjin Street, Changchun130012, P.R. China
| | - Lesheng Teng
- School of Life Sciences, Jilin University, No.2699, Qianjin Street, Changchun130012, P.R. China
| |
Collapse
|
33
|
Song P, Yang C, Thomsen JS, Dagnæs-Hansen F, Jakobsen M, Brüel A, Deleuran B, Kjems J. Lipidoid-siRNA Nanoparticle-Mediated IL-1β Gene Silencing for Systemic Arthritis Therapy in a Mouse Model. Mol Ther 2019; 27:1424-1435. [PMID: 31153827 DOI: 10.1016/j.ymthe.2019.05.002] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2018] [Revised: 04/29/2019] [Accepted: 05/01/2019] [Indexed: 11/18/2022] Open
Abstract
Interleukin-1 beta (IL-1β) plays a central role in the induction of rheumatoid arthritis (RA). In the present study, we demonstrated that lipidoid-polymer hybrid nanoparticle (FS14-NP) can efficiently deliver siRNA against IL-1β (siIL-1β) to macrophages and effectively suppress the pathogenesis of experimental arthritis induced by collagen antibody (CAIA mice). FS14-NP/siIL-1β achieved approximately 70% and 90% gene-silencing efficiency in the RAW 264.7 cell line and intraperitoneal macrophages, respectively. Intravenous administration of FS14-NP/siRNA led to rapid accumulation of siRNA in macrophages within the arthritic joints. Furthermore, FS14-NP/siIL-1β treatment lowered the expression of pro-inflammatory cytokines in arthritic joints and dramatically attenuated ankle swelling, bone erosion, and cartilage destruction. These results demonstrate that FS14-NP/siIL-1β may represent an effective therapy for systemic arthritis and other inflammatory disorders.
Collapse
Affiliation(s)
- Ping Song
- Interdisciplinary Nanoscience Center (iNANO), Aarhus University, 8000 Aarhus C, Denmark; Department of Molecular Biology and Genetics, Aarhus University, 8000 Aarhus C, Denmark
| | - Chuanxu Yang
- Interdisciplinary Nanoscience Center (iNANO), Aarhus University, 8000 Aarhus C, Denmark; Department of Molecular Biology and Genetics, Aarhus University, 8000 Aarhus C, Denmark.
| | | | | | - Maria Jakobsen
- Department of Molecular Biology and Genetics, Aarhus University, 8000 Aarhus C, Denmark
| | - Annemarie Brüel
- Department of Biomedicine, Aarhus University, 8000 Aarhus C, Denmark
| | - Bent Deleuran
- Department of Biomedicine, Aarhus University, 8000 Aarhus C, Denmark; Department of Rheumatology, Aarhus University Hospital, 8000 Aarhus C, Denmark
| | - Jørgen Kjems
- Interdisciplinary Nanoscience Center (iNANO), Aarhus University, 8000 Aarhus C, Denmark; Department of Molecular Biology and Genetics, Aarhus University, 8000 Aarhus C, Denmark.
| |
Collapse
|
34
|
Janakiraman K, Krishnaswami V, Rajendran V, Natesan S, Kandasamy R. Novel nano therapeutic materials for the effective treatment of rheumatoid arthritis-recent insights. MATERIALS TODAY. COMMUNICATIONS 2018; 17:200-213. [PMID: 32289062 PMCID: PMC7104012 DOI: 10.1016/j.mtcomm.2018.09.011] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/26/2018] [Revised: 09/11/2018] [Accepted: 09/13/2018] [Indexed: 05/02/2023]
Abstract
Rheumatoid arthritis (RA) is the most common complex multifactorial joint related autoimmune inflammatory disease with unknown etiology accomplished with increased cardiovascular risks. RA is characterized by the clinical findings of synovial inflammation, autoantibody production, and cartilage/bone destruction, cardiovascular, pulmonary and skeletal disorders. Pro-inflammatory cytokines such as IL-1, IL-6, IL-8, and IL-10 were responsible for the induction of inflammation in RA patients. Drawbacks such as poor efficacy, higher doses, frequent administration, low responsiveness, and higher cost and serious side effects were associated with the conventional dosage forms for RA treatment. Nanomedicines were recently gaining more interest towards the treatment of RA, and researchers were also focusing towards the development of various anti-inflammatory drug loaded nanoformulations with an aid to both actively/passively targeting the inflamed site to afford an effective treatment regimen for RA. Alterations in the surface area and nanoscale size of the nanoformulations elicit beneficial physical and chemical properties for better pharmacological activities. These drug loaded nanoformulations may enhances the solubility of poorly water soluble drugs, improves the bioavailability, affords targetability and may improve the therapeutic activity. In this regimen, the present review focus towards the novel nanoparticulate formulations (nanoparticles, nanoemulsions, solid lipid nanoparticles, nanomicelles, and nanocapsules) utilized for the treatment of RA. The recent advancements such as siRNA, peptide and targeted based nanoparticulate systems for RA treatment were also discussed. Special emphasis was provided regarding the pathophysiology, prevalence and symptoms towards the development of RA.
Collapse
Key Words
- A-SLN, actarit loaded solid lipid nanoparticles
- ACF-SLN, aceclofenac loaded solid lipid nanoparticles
- AIA, antigen-induced arthritis
- ALP, alkaline phosphate
- ALT, alanine aminotransferase
- AST, aspartate aminotransferase
- C-SLN, curcumin loaded solid lipid nanoparticles
- CEL-TS-LN, celecoxib loaded tristearin based lipidic nanoparticles
- CFA, complete freund’s adjuvant
- CHNP, chitosan nanoparticle
- CLSM, confocal laser scanning microscopy
- COX- 1, cyclooxygenase - 1
- COX- 2, cyclooxygenase - 2
- DEX, dexamethasone
- DEX-PMs, dexamethasone-loaded polymeric micelles
- DMARD, disease modifying antirheumatic drugs
- FA, folic acid
- FR-β, folate receptor-beta
- GC, glucocorticoid
- HA- AuNP/TCZ, hyaluronate gold nanoparticle/Tocilizumab
- HEKcells, human embryonic kidney cells
- HSA-NCs, human serum albumin nanocapsules
- HUVEC, human umbilical vein cells
- IL, interleukin
- IND-NMs, indomethacin loaded polymeric micelles
- Ig, immunoglobulin
- Ind-NCs, indomethacin-loaded nanocapsules
- Inflammation
- LDE, lipidic nanoemulsion
- LX-NMs, larnoxicam loaded nanomicelles
- MTX-LCNCs, methotrexate-loaded lipidic core nanocapsules
- NSAIDs, non steroidal anti-inflammatory drugs
- Nanoformulation
- Nanoparticles
- P-SLN, piperine loaded solid lipid nanoparticle
- PCL, polycaprolactone
- PCL-PEG, poly (ethylene glycol)-block-poly (ε-caprolactone)
- PSA, polysialic acid
- PSA-PCL-CyA-NMs, polysialic acid- polycaprolactone cyclosporine A nanomicelles
- Pir-SLN, piroxicam solid lipid nanoparticles
- RA, rheumatoid arthritis
- RGD, arginine-glycine aspartic acid
- RNAi, RNA interference
- Rheumatoid arthritis
- SLN, solid lipid nanoparticles
- TAC-HSA-NPs, tacrolimus human serum albumin nanoparticle
- TAC-LCNCs, tacrolimus loaded lipidic core nanocapsules
- TNF-α, tumour necrosis factor
- VCAM-1, vascular cell adhesion molecule-1
- VEGF, vascular endothelial growth factor
- VIP, vasoactive intestinal peptide
- mRNA, messenger RNA
- shRNA, short hairpin RNA
- siRNA, small interfering RNA
Collapse
Affiliation(s)
- Kumar Janakiraman
- National Facility for Drug Development for Academia, Pharmaceutical and Allied Industries (NFDD), Centre for Excellence in Nanobio Translational REsearch (CENTRE), Department of Pharmaceutical Technology, University College of Engineering, Anna University, BIT Campus, Tiruchirappalli 620 024, Tamil Nadu, India
| | - Venkateshwaran Krishnaswami
- National Facility for Drug Development for Academia, Pharmaceutical and Allied Industries (NFDD), Centre for Excellence in Nanobio Translational REsearch (CENTRE), Department of Pharmaceutical Technology, University College of Engineering, Anna University, BIT Campus, Tiruchirappalli 620 024, Tamil Nadu, India
| | - Vijaya Rajendran
- National Facility for Drug Development for Academia, Pharmaceutical and Allied Industries (NFDD), Centre for Excellence in Nanobio Translational REsearch (CENTRE), Department of Pharmaceutical Technology, University College of Engineering, Anna University, BIT Campus, Tiruchirappalli 620 024, Tamil Nadu, India
| | - Subramanian Natesan
- National Facility for Drug Development for Academia, Pharmaceutical and Allied Industries (NFDD), Centre for Excellence in Nanobio Translational REsearch (CENTRE), Department of Pharmaceutical Technology, University College of Engineering, Anna University, BIT Campus, Tiruchirappalli 620 024, Tamil Nadu, India
| | - Ruckmani Kandasamy
- National Facility for Drug Development for Academia, Pharmaceutical and Allied Industries (NFDD), Centre for Excellence in Nanobio Translational REsearch (CENTRE), Department of Pharmaceutical Technology, University College of Engineering, Anna University, BIT Campus, Tiruchirappalli 620 024, Tamil Nadu, India
| |
Collapse
|
35
|
Chiesa E, Pisani S, Colzani B, Dorati R, Conti B, Modena T, Braekmans K, Genta I. Intra-Articular Formulation of GE11-PLGA Conjugate-Based NPs for Dexamethasone Selective Targeting-In Vitro Evaluation. Int J Mol Sci 2018; 19:E2304. [PMID: 30082640 PMCID: PMC6121689 DOI: 10.3390/ijms19082304] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2018] [Revised: 07/31/2018] [Accepted: 08/03/2018] [Indexed: 12/16/2022] Open
Abstract
Selectively targeted nanoscale drug delivery systems have recently emerged as promising intravenously therapeutic option for most chronic joint diseases. Here, a newly synthetized dodecapeptide (GE11)-polylactide-co-glycolide (PLGA)-based conjugate was used to prepare smart nanoparticles (NPs) intended for intra-articular administration and for selectively targeting Epidermal Growth Factor Receptor (EGFR). GE11-PLGA conjugate-based NPs are specifically uptaken by EGFR-overexpressed fibroblast; such as synoviocytes; which are the primarily cellular component involved in the development of destructive joint inflammation. The selective uptake could help to tune drug effectiveness in joints and to decrease local and systemic side effects. Dexamethasone (DXM) is a glucorticoid drug commonly used in joint disease treatment for both systemic and local administration route. In the present research; DXM was efficiently loaded into GE11-PLGA conjugate-based NPs through an eco-friendly nanoprecipitation method set up for this purpose. DXM loaded GE11-PLGA conjugate-based NPs revealed satisfactory ex vivo cytocompatibility; with proper size (≤150 nm) and good dimensional stability in synovial fluid. Intra-articular formulation was developed embedding DXM loaded GE11-PLGA conjugate-based NPs into thermosetting chitosan-based hydrogel; forming a biocompatible composite hydrogel able to quickly turn from liquid state into gel state at physiological temperature; within 15 min. Moreover; the use of thermosetting chitosan-based hydrogel extends the local release of active agent; DXM.
Collapse
Affiliation(s)
- Enrica Chiesa
- Department of Drug Sciences, University of Pavia, V.le Taramelli 12, 27100 Pavia (PV), Italy.
| | - Silvia Pisani
- Department of Drug Sciences, University of Pavia, V.le Taramelli 12, 27100 Pavia (PV), Italy.
| | - Barbara Colzani
- Department of Drug Sciences, University of Pavia, V.le Taramelli 12, 27100 Pavia (PV), Italy.
| | - Rossella Dorati
- Department of Drug Sciences, University of Pavia, V.le Taramelli 12, 27100 Pavia (PV), Italy.
| | - Bice Conti
- Department of Drug Sciences, University of Pavia, V.le Taramelli 12, 27100 Pavia (PV), Italy.
| | - Tiziana Modena
- Department of Drug Sciences, University of Pavia, V.le Taramelli 12, 27100 Pavia (PV), Italy.
| | - Kevin Braekmans
- Laboratory of General Biochemistry and Physical Pharmacy, Ghent University, Ottergemsesteenweg 460, 9000 Gent, Belgium.
| | - Ida Genta
- Department of Drug Sciences, University of Pavia, V.le Taramelli 12, 27100 Pavia (PV), Italy.
| |
Collapse
|
36
|
Liu Y, Hardie J, Zhang X, Rotello VM. Effects of engineered nanoparticles on the innate immune system. Semin Immunol 2017; 34:25-32. [PMID: 28985993 PMCID: PMC5705289 DOI: 10.1016/j.smim.2017.09.011] [Citation(s) in RCA: 175] [Impact Index Per Article: 21.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/11/2017] [Revised: 09/26/2017] [Accepted: 09/27/2017] [Indexed: 02/04/2023]
Abstract
Engineered nanoparticles (NPs) have broad applications in industry and nanomedicine. When NPs enter the body, interactions with the immune system are unavoidable. The innate immune system, a non-specific first line of defense against potential threats to the host, immediately interacts with introduced NPs and generates complicated immune responses. Depending on their physicochemical properties, NPs can interact with cells and proteins to stimulate or suppress the innate immune response, and similarly activate or avoid the complement system. NPs size, shape, hydrophobicity and surface modification are the main factors that influence the interactions between NPs and the innate immune system. In this review, we will focus on recent reports about the relationship between the physicochemical properties of NPs and their innate immune response, and their applications in immunotherapy.
Collapse
Affiliation(s)
- Yuanchang Liu
- Department of Chemistry, University of Massachusetts Amherst, 710 North Pleasant Street, Amherst, MA, 01003, USA
| | - Joseph Hardie
- Department of Chemistry, University of Massachusetts Amherst, 710 North Pleasant Street, Amherst, MA, 01003, USA
| | - Xianzhi Zhang
- Department of Chemistry, University of Massachusetts Amherst, 710 North Pleasant Street, Amherst, MA, 01003, USA
| | - Vincent M Rotello
- Department of Chemistry, University of Massachusetts Amherst, 710 North Pleasant Street, Amherst, MA, 01003, USA.
| |
Collapse
|
37
|
Chen X, Kuang N, Zeng X, Zhang Z, Li Y, Liu W, Fu Y. Effects of daphnetin combined with Bcl2‑siRNA on antiapoptotic genes in synovial fibroblasts of rats with collagen‑induced arthritis. Mol Med Rep 2017; 17:884-890. [PMID: 29115552 DOI: 10.3892/mmr.2017.8008] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2016] [Accepted: 07/21/2017] [Indexed: 11/06/2022] Open
Abstract
The aim of the present study was to investigate the effects of daphnetin combined with B cell lymphoma 2 (Bcl2)‑targeted small interfering (si)RNA (si‑Bcl2) on antiapoptotic genes in fibroblast‑like synoviocytes (FLS) in rats with collagen II‑induced arthritis (CIA). The roles of si‑Bcl2 and daphnetin were determined by measuring the expression levels of Bcl2. Protein and mRNA expression levels of Bcl2 in FLS were determined by flow cytometry and reverse transcription‑quantitative polymerase chain reaction. Apoptosis of FLS was also determined by flow cytometry. It was revealed that treatment with si‑Bcl2 or daphnetin alone resulted in downregulation of Bcl2 mRNA and protein expression. In addition, the mRNA expression levels of the signal transducer and activator of transcription 3 (STAT3), which transcriptionally regulates the activity of mitochondria, were reduced. The combination of si‑Bcl2 and daphnetin exhibited an enhanced effect on rheumatoid arthritis FLS (RAFLS), in which the apoptotic rate was significantly higher than either treatment alone. The results suggested that si‑Bcl combined with daphnetin may have an enhanced effect in promoting apoptosis of RAFLS derived from CIA rats, and a possible underlying molecular mechanism may function through the downregulation of Bcl2 expression and STAT3, which is located upstream of Bcl2 in the mitochondrial apoptotic pathway. The results of the present study are expected to provide theoretical and experimental basis for the treatment of RA and the medicinal development of daphnetin combined siRNA.
Collapse
Affiliation(s)
- Xiaoying Chen
- Department of Immunology, Medical College of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Nanzhen Kuang
- Department of Immunology, Medical College of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Xiaoping Zeng
- Department of Immunology, Medical College of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Zhiqin Zhang
- Department of Immunology, Medical College of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Yanyan Li
- Department of Immunology, Medical College of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Wei Liu
- Department of Immunology, Medical College of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Yingyuan Fu
- Department of Immunology, Medical College of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| |
Collapse
|
38
|
Yang C, Vu-Quang H, Husum DMU, Tingskov SJ, Vinding MS, Nielsen T, Song P, Nielsen NC, Nørregaard R, Kjems J. Theranostic poly(lactic-co-glycolic acid) nanoparticle for magnetic resonance/infrared fluorescence bimodal imaging and efficient siRNA delivery to macrophages and its evaluation in a kidney injury model. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2017; 13:2451-2462. [DOI: 10.1016/j.nano.2017.08.007] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/08/2017] [Revised: 07/25/2017] [Accepted: 08/12/2017] [Indexed: 01/23/2023]
|
39
|
Wang Y, Wu M, Gu L, Li X, He J, Zhou L, Tong A, Shi J, Zhu H, Xu J, Guo G. Effective improvement of the neuroprotective activity after spinal cord injury by synergistic effect of glucocorticoid with biodegradable amphipathic nanomicelles. Drug Deliv 2017; 24:391-401. [PMID: 28165815 PMCID: PMC8241193 DOI: 10.1080/10717544.2016.1256003] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2016] [Revised: 10/27/2016] [Accepted: 10/30/2016] [Indexed: 02/05/2023] Open
Abstract
Dexamethasone acetate (DA) produces neuroprotective effects by inhibiting lipid peroxidation and inflammation by reducing cytokine release and expression. However, its clinical application is limited by its hydrophobicity, low biocompatibility and numerous side effects when using large dosage. Therefore, improving DA's water solubility, biocompatibility and reducing its side effects are important goals that will improve its clinical utility. The objective of this study is to use a biodegradable polymer as the delivery vehicle for DA to achieve the synergism between inhibiting lipid peroxidation and inflammation effects of the hydrophobic-loaded drugs and the amphipathic delivery vehicle. We successfully prepared DA-loaded polymeric micelles (DA/MPEG-PCL micelles) with monodispersed and approximately 25 nm in diameter, and released DA over an extended period in vitro. Additionally, in the hemisection spinal cord injury (SCI) model, DA micelles were more effective in promoting hindlimb functional recover, reducing glial scar and cyst formation in injured site, decreasing neuron lose and promoting axon regeneration. Therefore, our data suggest that DA/MPEG-PCL micelles have the potential to be applied clinically in SCI therapy.
Collapse
Affiliation(s)
- YueLong Wang
- State Key Laboratory of Biotherapy and Cancer Center and Department of Neurosurgery, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, PR China
| | - Min Wu
- Department of Radiology, Huaxi MR Research Center (HMRRC), West China Hospital, Sichuan University, Chengdu, PR China
| | - Lei Gu
- Department of Radiology, Huaxi MR Research Center (HMRRC), West China Hospital, Sichuan University, Chengdu, PR China
| | - XiaoLing Li
- State Key Laboratory of Biotherapy and Cancer Center and Department of Neurosurgery, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, PR China
| | - Jun He
- State Key Laboratory of Biotherapy and Cancer Center and Department of Neurosurgery, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, PR China
| | - LiangXue Zhou
- State Key Laboratory of Biotherapy and Cancer Center and Department of Neurosurgery, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, PR China
| | - Aiping Tong
- State Key Laboratory of Biotherapy and Cancer Center and Department of Neurosurgery, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, PR China
| | - Juan Shi
- National Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, PR China, and
| | - HongYan Zhu
- Laboratory of Stem Cell Biology, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, PR China
| | - JianGuo Xu
- State Key Laboratory of Biotherapy and Cancer Center and Department of Neurosurgery, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, PR China
| | - Gang Guo
- State Key Laboratory of Biotherapy and Cancer Center and Department of Neurosurgery, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, PR China
| |
Collapse
|
40
|
Silva AL, Peres C, Conniot J, Matos AI, Moura L, Carreira B, Sainz V, Scomparin A, Satchi-Fainaro R, Préat V, Florindo HF. Nanoparticle impact on innate immune cell pattern-recognition receptors and inflammasomes activation. Semin Immunol 2017; 34:3-24. [PMID: 28941640 DOI: 10.1016/j.smim.2017.09.003] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/03/2017] [Revised: 09/10/2017] [Accepted: 09/11/2017] [Indexed: 12/19/2022]
Abstract
Nanotechnology-based strategies can dramatically impact the treatment, prevention and diagnosis of a wide range of diseases. Despite the unprecedented success achieved with the use of nanomaterials to address unmet biomedical needs and their particular suitability for the effective application of a personalized medicine, the clinical translation of those nanoparticulate systems has still been impaired by the limited understanding on their interaction with complex biological systems. As a result, unexpected effects due to unpredicted interactions at biomaterial and biological interfaces have been underlying the biosafety concerns raised by the use of nanomaterials. This review explores the current knowledge on how nanoparticle (NP) physicochemical and surface properties determine their interactions with innate immune cells, with particular attention on the activation of pattern-recognition receptors and inflammasome. A critical perspective will additionally address the impact of biological systems on the effect of NP on immune cell activity at the molecular level. We will discuss how the understanding of the NP-innate immune cell interactions can significantly add into the clinical translation by guiding the design of nanomedicines with particular effect on targeted cells, thus improving their clinical efficacy while minimizing undesired but predictable toxicological effects.
Collapse
Affiliation(s)
- Ana Luísa Silva
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003 Lisbon, Portugal
| | - Carina Peres
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003 Lisbon, Portugal; Université Catholique de Louvain, Louvain Drug Research Institute, Advanced Drug Delivery and Biomaterials, 1200 Brussels, Belgium
| | - João Conniot
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003 Lisbon, Portugal
| | - Ana I Matos
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003 Lisbon, Portugal
| | - Liane Moura
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003 Lisbon, Portugal
| | - Bárbara Carreira
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003 Lisbon, Portugal
| | - Vanessa Sainz
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003 Lisbon, Portugal
| | - Anna Scomparin
- Department of Physiology and Pharmacology, Sackler School of Medicine, Tel Aviv University, Tel Aviv 69978, Israel and dSagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel
| | - Ronit Satchi-Fainaro
- Department of Physiology and Pharmacology, Sackler School of Medicine, Tel Aviv University, Tel Aviv 69978, Israel and dSagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel; Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel
| | - Véronique Préat
- Université Catholique de Louvain, Louvain Drug Research Institute, Advanced Drug Delivery and Biomaterials, 1200 Brussels, Belgium.
| | - Helena F Florindo
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003 Lisbon, Portugal.
| |
Collapse
|
41
|
Wang Q, Jiang H, Li Y, Chen W, Li H, Peng K, Zhang Z, Sun X. Targeting NF-kB signaling with polymeric hybrid micelles that co-deliver siRNA and dexamethasone for arthritis therapy. Biomaterials 2017; 122:10-22. [DOI: 10.1016/j.biomaterials.2017.01.008] [Citation(s) in RCA: 120] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2016] [Revised: 01/04/2017] [Accepted: 01/07/2017] [Indexed: 12/17/2022]
|
42
|
Scott EA, Karabin NB, Augsornworawat P. Overcoming Immune Dysregulation with Immunoengineered Nanobiomaterials. Annu Rev Biomed Eng 2017; 19:57-84. [PMID: 28226216 DOI: 10.1146/annurev-bioeng-071516-044603] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
The immune system is governed by an immensely complex network of cells and both intracellular and extracellular molecular factors. It must respond to an ever-growing number of biochemical and biophysical inputs by eliciting appropriate and specific responses in order to maintain homeostasis. But as with any complex system, a plethora of false positives and false negatives can occur to generate dysregulated responses. Dysregulated immune responses are essential components of diverse inflammation-driven pathologies, including cancer, heart disease, and autoimmune disorders. Nanoscale biomaterials (i.e., nanobiomaterials) have emerged as highly customizable platforms that can be engineered to interact with and direct immune responses, holding potential for the design of novel and targeted approaches to redirect or inhibit inflammation. Here, we present recent developments of nanobiomaterials that were rationally designed to target and modulate inflammatory cells and biochemical pathways for the treatment of immune dysregulation.
Collapse
Affiliation(s)
- Evan A Scott
- Department of Biomedical Engineering, Northwestern University, Evanston, Illinois 60208
| | - Nicholas B Karabin
- Department of Biomedical Engineering, Northwestern University, Evanston, Illinois 60208
| | - Punn Augsornworawat
- Department of Biomedical Engineering, Northwestern University, Evanston, Illinois 60208
| |
Collapse
|
43
|
Roozbahani M, Kharaziha M, Emadi R. pH sensitive dexamethasone encapsulated laponite nanoplatelets: Release mechanism and cytotoxicity. Int J Pharm 2017; 518:312-319. [PMID: 28062364 DOI: 10.1016/j.ijpharm.2017.01.001] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2016] [Revised: 01/01/2017] [Accepted: 01/02/2017] [Indexed: 12/16/2022]
|
44
|
Abstract
Schematic illustration of inflammatory microenvironment in inflamed joints and events occurring in rheumatoid arthritis.
Collapse
Affiliation(s)
- Qin Wang
- Key Laboratory of Drug Targeting and Drug Delivery Systems
- Ministry of Education
- West China School of Pharmacy
- Sichuan University
- Chengdu
| | - Xun Sun
- Key Laboratory of Drug Targeting and Drug Delivery Systems
- Ministry of Education
- West China School of Pharmacy
- Sichuan University
- Chengdu
| |
Collapse
|
45
|
Xiao B, Zhang Z, Viennois E, Kang Y, Zhang M, Han MK, Chen J, Merlin D. Combination Therapy for Ulcerative Colitis: Orally Targeted Nanoparticles Prevent Mucosal Damage and Relieve Inflammation. Am J Cancer Res 2016; 6:2250-2266. [PMID: 27924161 PMCID: PMC5135446 DOI: 10.7150/thno.15710] [Citation(s) in RCA: 164] [Impact Index Per Article: 18.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2016] [Accepted: 08/12/2016] [Indexed: 02/06/2023] Open
Abstract
Combination therapy is an emerging strategy that is under intensive preclinical investigation for the treatment of various diseases. CD98 is highly overexpressed on the surfaces of epithelial cells and macrophages in the colon tissue with ulcerative colitis (UC), which is usually associated with mucosal damage and inflammation. We previously proved that CD98 siRNA (siCD98)-induced down-regulation of CD98 in colitis tissue decreased the severity of UC to a certain extent. In an effort to further improve the therapeutic efficacy, we aim to simultaneously deliver siCD98 in combination with a potent anti-inflammatory agent, curcumin (CUR), using hyaluronic acid (HA)-functionalized polymeric nanoparticles (NPs). The resultant spherical HA-siCD98/CUR-NPs are featured by a desirable particle size (∼246 nm) and slightly negative zeta potential (∼-14 mV). The NPs functionalized with HA are able to guide the co-delivery of drugs to the targeted cells related to UC therapy (colonic epithelial cells and macrophages). Compared to either siCD98- or CUR-based monotherapy, co-delivery of siCD98 and CUR by HA-functionalized NPs can exert combinational effects against UC by protecting the mucosal layer and alleviating inflammation both in vitro and in vivo. This study shows the promising capability of the co-delivered siCD98 and CUR for boosting the conventional monotherapy via this novel nanotherapeutic agent, which offers a structurally simple platform for orally administered delivery of drugs to target cells in UC therapy.
Collapse
|
46
|
Song J, Chen Y, Jiang S, Yang K, Li X, Zhao X, Ouyang Y, Fan C, Yuan W. Efficient and Non-Toxic Biological Response Carrier Delivering TNF-α shRNA for Gene Silencing in a Murine Model of Rheumatoid Arthritis. Front Immunol 2016; 7:305. [PMID: 27594856 PMCID: PMC4990551 DOI: 10.3389/fimmu.2016.00305] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2016] [Accepted: 07/27/2016] [Indexed: 12/29/2022] Open
Abstract
Small interfering RNA (siRNA) is an effective and specific method for silencing genes. However, an efficient and non-toxic carrier is needed to deliver the siRNA into the target cells. Tumor necrosis factor α (TNF-α) plays a central role in the occurrence and progression of rheumatoid arthritis (RA). In this study, we pre-synthetized a degradable cationic polymer (PDAPEI) from 2,6-pyridinedicarboxaldehyde and low-molecular-weight polyethyleneimine (PEI, Mw = 1.8 kDa) as a gene vector for the delivery of TNF-α shRNA. The PDAPEI/pDNA complex showed a suitable particle size and stable zeta potential for transfection. In vitro study of the PDAPEI/pDNA complex revealed a lower cytotoxicity and higher transfection efficiency when transfecting TNF-α shRNA to macrophages by significantly down-regulating the expression of TNF-α. Moreover, the complex was extremely efficient in decreasing the severity of arthritis in mice with collagen-induced arthritis. PDAPEI delivered TNF-α shRNA has great potential in the treatment of RA.
Collapse
Affiliation(s)
- Jialin Song
- Shanghai Jiao Tong University Affiliated Sixth People's Hospital , Shanghai , China
| | - Yinghui Chen
- Department of Neurology, Jinshan Hospital, Fudan University , Shanghai , China
| | - Shichao Jiang
- Department of Orthopedics, Shandong Provincial Hospital Affiliated to Shandong University , Jinan, Shandong , China
| | - Kejia Yang
- School of Pharmacy, Shanghai JiaoTong University , Shanghai , China
| | - Xiaoming Li
- School of Pharmacy, Shanghai JiaoTong University , Shanghai , China
| | - Xiaotian Zhao
- School of Pharmacy, Shanghai JiaoTong University , Shanghai , China
| | - Yuanming Ouyang
- Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China; Shanghai Sixth People's Hospital East Campus, Shanghai University of Medicine and Health, Shanghai, China
| | - Cunyi Fan
- Shanghai Jiao Tong University Affiliated Sixth People's Hospital , Shanghai , China
| | - Weien Yuan
- School of Pharmacy, Shanghai JiaoTong University , Shanghai , China
| |
Collapse
|
47
|
Lin JB, Poh S, Panitch A. Controlled release of anti-inflammatory peptides from reducible thermosensitive nanoparticles suppresses cartilage inflammation. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2016; 12:2095-2100. [PMID: 27241526 DOI: 10.1016/j.nano.2016.05.010] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/23/2016] [Revised: 04/28/2016] [Accepted: 05/14/2016] [Indexed: 01/25/2023]
Abstract
Characterized by pain, cartilage degradation, and inflammation, osteoarthritis is often treated with anti-inflammatory therapies that provide short-term relief but can have adverse side effects; intra-articular drug delivery systems with controlled release of anti-inflammatory peptides using degradable poly(N-isopropylacrylamide) (pNIPAM) nanoparticles could prolong relief and minimize these side effects. Nanoparticles provide a biocompatible drug carrier that can protect encapsulated therapeutics from enzymatic degradation and increase payload delivery upon encountering a degradation stimulus. Here we demonstrate passive targeting of inflamed cartilage ex vivo by uptake of PEGylated pNIPAM nanoparticles with degradable disulfide crosslinks (abbreviated as NGPEGSS) into chondrocytes and subsequent intracellular release of an anti-inflammatory peptide KAFAKLAARLYRKALARQLGVAA (KAFAK). The KAFAK-loaded NGPEGSS treatment reduced ex vivo inflammation to a greater extent compared to its non-degradable counterparts. This study highlights a nanoparticle system that delivers therapeutics intracellularly with improved efficacy by triggered degradation and suppresses inflammation in multiple cell types within an inflamed joint.
Collapse
Affiliation(s)
- Jenny B Lin
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN, USA
| | - Scott Poh
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN, USA
| | - Alyssa Panitch
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN, USA.
| |
Collapse
|
48
|
Dobrovolskaia MA, Shurin M, Shvedova AA. Current understanding of interactions between nanoparticles and the immune system. Toxicol Appl Pharmacol 2016; 299:78-89. [PMID: 26739622 PMCID: PMC4811709 DOI: 10.1016/j.taap.2015.12.022] [Citation(s) in RCA: 202] [Impact Index Per Article: 22.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2015] [Revised: 12/24/2015] [Accepted: 12/26/2015] [Indexed: 10/22/2022]
Abstract
The delivery of drugs, antigens, and imaging agents benefits from using nanotechnology-based carriers. The successful translation of nanoformulations to the clinic involves thorough assessment of their safety profiles, which, among other end-points, includes evaluation of immunotoxicity. The past decade of research focusing on nanoparticle interaction with the immune system has been fruitful in terms of understanding the basics of nanoparticle immunocompatibility, developing a bioanalytical infrastructure to screen for nanoparticle-mediated immune reactions, beginning to uncover the mechanisms of nanoparticle immunotoxicity, and utilizing current knowledge about the structure-activity relationship between nanoparticles' physicochemical properties and their effects on the immune system to guide safe drug delivery. In the present review, we focus on the most prominent pieces of the nanoparticle-immune system puzzle and discuss the achievements, disappointments, and lessons learned over the past 15years of research on the immunotoxicity of engineered nanomaterials.
Collapse
Affiliation(s)
- Marina A Dobrovolskaia
- Nanotechnology Characterization Laboratory, Cancer Research Technology Program, Leidos Biomedical Research Inc., Frederick National Laboratory for Cancer Research, NCI at Frederick, Frederick, MD 21702, USA.
| | - Michael Shurin
- Department of Pathology, University of Pittsburgh Medical Center, Pittsburgh, PA 15213, USA; Department of Immunology, University of Pittsburgh Medical Center, Pittsburgh, PA 15213, USA
| | - Anna A Shvedova
- Health Effects Laboratory Division, National Institute of Occupational Safety and Health, Centers for Disease Control and Prevention, Morgantown, WV 26505, USA; Department of Physiology and Pharmacology, West Virginia University, Morgantown, WV 26506, USA.
| |
Collapse
|
49
|
Liu Q, Yuan W, Tong D, Liu G, Lan W, Zhang D, Xiao H, Zhang Y, Huang Z, Yang J, Zhang J, Jiang J. Metformin represses bladder cancer progression by inhibiting stem cell repopulation via COX2/PGE2/STAT3 axis. Oncotarget 2016; 7:28235-46. [PMID: 27058422 PMCID: PMC5053723 DOI: 10.18632/oncotarget.8595] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2016] [Accepted: 03/28/2016] [Indexed: 01/05/2023] Open
Abstract
Cancer stem cells (CSCs) are a sub-population of tumor cells playing essential roles in initiation, differentiation, recurrence, metastasis and development of drug resistance of various cancers, including bladder cancer. Although multiple lines of evidence suggest that metformin is capable of repressing CSC repopulation in different cancers, the effect of metformin on bladder cancer CSCs remains largely unknown. Using the N-methyl-N-nitrosourea (MNU)-induced rat orthotropic bladder cancer model, we demonstrated that metformin is capable of repressing bladder cancer progression from both mild to moderate/severe dysplasia lesions and from carcinoma in situ (CIS) to invasive lesions. Metformin also can arrest bladder cancer cells in G1/S phases, which subsequently leads to apoptosis. And also metformin represses bladder cancer CSC repopulation evidenced by reducing cytokeratin 14 (CK14+) and octamer-binding transcription factor 3/4 (OCT3/4+) cells in both animal and cellular models. More importantly, we found that metformin exerts these anticancer effects by inhibiting COX2, subsequently PGE2 as well as the activation of STAT3. In conclusion, we are the first to systemically demonstrate in both animal and cell models that metformin inhibits bladder cancer progression by inhibiting stem cell repopulation through the COX2/PGE2/STAT3 axis.
Collapse
Affiliation(s)
- Qiuli Liu
- Department of Urology, Institute of Surgery Research, Daping Hospital, Third Military Medical University, Chongqing, 400042, PR China
| | - Wenqiang Yuan
- Department of Urology, Institute of Surgery Research, Daping Hospital, Third Military Medical University, Chongqing, 400042, PR China
| | - Dali Tong
- Department of Urology, Institute of Surgery Research, Daping Hospital, Third Military Medical University, Chongqing, 400042, PR China
| | - Gaolei Liu
- Department of Urology, Institute of Surgery Research, Daping Hospital, Third Military Medical University, Chongqing, 400042, PR China
| | - Weihua Lan
- Department of Urology, Institute of Surgery Research, Daping Hospital, Third Military Medical University, Chongqing, 400042, PR China
| | - Dianzheng Zhang
- Department of Bio-Medical Sciences, Philadelphia College of Osteopathic Medicine, Philadelphia, PA, 19131, USA
| | - Hualiang Xiao
- Department of Pathology, Institute of Surgery Research, Daping Hospital, Third Military Medical University, Chongqing, 400042, PR China
| | - Yao Zhang
- Department of Urology, Institute of Surgery Research, Daping Hospital, Third Military Medical University, Chongqing, 400042, PR China
| | - Zaoming Huang
- Department of Urology, Institute of Surgery Research, Daping Hospital, Third Military Medical University, Chongqing, 400042, PR China
| | - Junjie Yang
- Department of Urology, Institute of Surgery Research, Daping Hospital, Third Military Medical University, Chongqing, 400042, PR China
| | - Jun Zhang
- Department of Urology, Institute of Surgery Research, Daping Hospital, Third Military Medical University, Chongqing, 400042, PR China
| | - Jun Jiang
- Department of Urology, Institute of Surgery Research, Daping Hospital, Third Military Medical University, Chongqing, 400042, PR China
| |
Collapse
|
50
|
Double emulsion solvent evaporation techniques used for drug encapsulation. Int J Pharm 2015; 496:173-90. [DOI: 10.1016/j.ijpharm.2015.10.057] [Citation(s) in RCA: 256] [Impact Index Per Article: 25.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2015] [Revised: 10/21/2015] [Accepted: 10/22/2015] [Indexed: 12/11/2022]
|