1
|
Siddique R, Muhammad F, Faisal MN, Akhtar B, Saleem A, Kousar S, Sharif A, Saeed M, Muhammad S. Gingerol nanoparticles attenuate complete Freund adjuvant-induced arthritis in rats via targeting the RANKL/OPG signaling pathway. Inflammopharmacology 2024; 32:3311-3326. [PMID: 39207636 DOI: 10.1007/s10787-024-01537-5] [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: 04/27/2023] [Accepted: 03/23/2024] [Indexed: 09/04/2024]
Abstract
Rheumatoid arthritis (RA) is characterized by inflammatory joint pathology leading to the degradation of articular bone and cartilage, primarily triggered by synovial inflammation, resulting in joint discomfort. The metacarpophalangeal and proximal interphalangeal joints are predominantly affected. Treatment typically involves a combination of biological and synthetic disease-modifying antirheumatic drugs (DAMARDs) alongside steroid therapy. The application of nanomedicine has been instrumental in enhancing treatment efficacy by facilitating controlled release of pharmacologically active compounds, thus augmenting bioavailability and enabling targeted drug delivery. Gingerol, a constituent of ginger, possesses multifaceted properties. including anti-inflammatory, anti-oxidant, antidiabetic, and antipyretic effects. In this study, gingerol-loaded poly(lactic-co-glycolic acid) (PLGA) nanoparticles (NPs), coated with chitosan, were administered orally to rats over a period of 21 days to address RA induced by complete Freund adjuvant (CFA). The rats were segregated into four experimental groups. Upon completion of the treatment regimen, blood samples were collected for the assessment of cyclooxygenase-2 (COX-2), RA factor, interleukin-6 (IL-6), and tumor necrosis factor-alpha (TNF-α). Subsequent gene expression analysis was conducted to evaluate the levels of interleukin-4 (IL-4), interleukin-17a (IL-17a), IL-6, interferon-gamma (INF-γ), TNF-α, interleukin-1 beta (IL-1β), osteoprotegerin (OPG), and receptor activator of nuclear factor kappa-B ligand (RANKL). Statistical analyses utilizing one-way ANOVA followed by Tukey tests were applied to the data. The gene expression profiling revealed significant disparities in mRNA levels of IL-1β, IL-6, IL-4, IL-17a, RANKL, INF-γ, and TNF-α between the CFA-induced arthritis group and the control group. Consequently, it was inferred that gingerol-loaded PLGA NPs coated with chitosan exhibited heightened therapeutic efficacy in addressing CFA-induced arthritis in rats.
Collapse
Affiliation(s)
- Rida Siddique
- Department of Pharmacology, Faculty of Pharmaceutical Sciences, Government College University, Faisalabad, Pakistan
| | - Faqir Muhammad
- Department of Bio-Sciences, Faculty of Veterinary Sciences, Bahauddin Zakariya University, Multan, Pakistan.
- Institute of Physiology and Pharmacology, University of Agriculture, Faisalabad, Punjab, Pakistan.
| | - Muhammad Naeem Faisal
- Institute of Physiology and Pharmacology, University of Agriculture, Faisalabad, Punjab, Pakistan
| | - Bushra Akhtar
- Department of Pharmacy, University of Agriculture, Faisalabad, 38000, Pakistan
| | - Ammara Saleem
- Department of Pharmacology, Faculty of Pharmaceutical Sciences, Government College University, Faisalabad, Pakistan
| | - Shaneel Kousar
- Department of Pharmacy, The University of Lahore, Lahore, Pakistan
| | - Ali Sharif
- Institute of Pharmacy, Faculty of Pharmaceutical and Allied Health Sciences, Lahore College for Women University, Lahore, 54000, Pakistan
| | - Muhammad Saeed
- DHQ/Allied Hospital-2, Faisalabad Medical University, Faisalabad, Pakistan
| | - Safwan Muhammad
- Sahiwal Medical College, University of Health Sciences, Lahore, Pakistan
| |
Collapse
|
2
|
Xu H, Cui Y, Tian Y, Dou M, Sun S, Wang J, Wu D. Nanoparticle-Based Drug Delivery Systems for Enhancing Bone Regeneration. ACS Biomater Sci Eng 2024; 10:1302-1322. [PMID: 38346448 DOI: 10.1021/acsbiomaterials.3c01643] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/12/2024]
Abstract
The treatment of bone defects has been a long-standing challenge in clinical practice. Among the various bone tissue engineering approaches, there has been substantial progress in the development of drug delivery systems based on functional drugs and appropriate carrier materials owing to technological advances in recent years. A large number of materials based on functional nanocarriers have been developed and applied to improve the complex osteogenic microenvironment, including for promoting osteogenic activity, inhibiting osteoclast activity, and exerting certain antibacterial effects. This Review discusses the physicochemical properties, drug loading mechanisms, advantages and disadvantages of nanoparticles (NPs) used for constructing drug delivery systems. In addition, we provide an overview of the osteogenic microenvironment regulation mechanism of drug delivery systems based on nanoparticle (NP) carriers and the construction strategies of drug delivery systems. Finally, the advantages and disadvantages of NP carriers are summarized along with their prospects and future research trends in bone tissue engineering. This Review thus provides advanced strategies for the design and application of drug delivery systems based on NPs in the treatment of bone defects.
Collapse
Affiliation(s)
- Hang Xu
- Orthopaedic Medical Center, The Second Hospital of Jilin University, Changchun 130041, P. R. China
| | - Yutao Cui
- Orthopaedic Medical Center, The Second Hospital of Jilin University, Changchun 130041, P. R. China
| | - Yuhang Tian
- Orthopaedic Medical Center, The Second Hospital of Jilin University, Changchun 130041, P. R. China
| | - Minghan Dou
- Orthopaedic Medical Center, The Second Hospital of Jilin University, Changchun 130041, P. R. China
| | - Shouye Sun
- Orthopaedic Medical Center, The Second Hospital of Jilin University, Changchun 130041, P. R. China
| | - Jingwei Wang
- Orthopaedic Medical Center, The Second Hospital of Jilin University, Changchun 130041, P. R. China
| | - Dankai Wu
- Orthopaedic Medical Center, The Second Hospital of Jilin University, Changchun 130041, P. R. China
| |
Collapse
|
3
|
Garhwal A, Kendya P, Soni S, Kori S, Soni V, Kashaw SK. Drug Delivery System Approaches for Rheumatoid Arthritis Treatment: A Review. Mini Rev Med Chem 2024; 24:704-720. [PMID: 37711105 DOI: 10.2174/1389557523666230913105803] [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: 04/17/2023] [Revised: 07/22/2023] [Accepted: 07/23/2023] [Indexed: 09/16/2023]
Abstract
Rheumatoid arthritis (RA) is a chronic autoimmune disease that has traditionally been treated using a variety of pharmacological compounds. However, the effectiveness of these treatments is often limited due to challenges associated with their administration. Oral and parenteral routes of drug delivery are often restricted due to issues such as low bioavailability, rapid metabolism, poor absorption, first-pass effect, and severe side effects. In recent years, nanocarrier-based delivery methods have emerged as a promising alternative for overcoming these challenges. Nanocarriers, including nanoparticles, dendrimers, micelles, nanoemulsions, and stimuli-sensitive carriers, possess unique properties that enable efficient drug delivery and targeted therapy. Using nanocarriers makes it possible to circumvent traditional administration routes' limitations. One of the key advantages of nanocarrier- based delivery is the ability to overcome resistance or intolerance to traditional antirheumatic therapies. Moreover, nanocarriers offer improved drug stability, controlled release kinetics, and enhanced solubility, optimizing the therapeutic effect. They can also protect the encapsulated drug, prolonging its circulation time and facilitating sustained release at the target site. This targeted delivery approach ensures a higher concentration of the therapeutic agent at the site of inflammation, leading to improved therapeutic outcomes. This article explores potential developments in nanotherapeutic regimens for RA while providing a comprehensive summary of current approaches based on novel drug delivery systems. In conclusion, nanocarrier-based drug delivery systems have emerged as a promising solution for improving the treatment of rheumatoid arthritis. Further advancements in nanotechnology hold promise for enhancing the efficacy and safety of RA therapies, offering new hope for patients suffering from this debilitating disease.
Collapse
Affiliation(s)
- Anushka Garhwal
- Department of Pharmaceutical Sciences, Dr. Harisingh Gour University (A Central University), Sagar (MP), India
| | - Priyadarshi Kendya
- Department of Pharmaceutical Sciences, Dr. Harisingh Gour University (A Central University), Sagar (MP), India
| | - Sakshi Soni
- Department of Pharmaceutical Sciences, Dr. Harisingh Gour University (A Central University), Sagar (MP), India
| | - Shivam Kori
- Department of Pharmaceutical Sciences, Dr. Harisingh Gour University (A Central University), Sagar (MP), India
| | - Vandana Soni
- Department of Pharmaceutical Sciences, Dr. Harisingh Gour University (A Central University), Sagar (MP), India
| | - Sushil Kumar Kashaw
- Department of Pharmaceutical Sciences, Dr. Harisingh Gour University (A Central University), Sagar (MP), India
| |
Collapse
|
4
|
Vasdev N, Pawar B, Gupta T, Mhatre M, Tekade RK. A Bird's Eye View of Various Cell-Based Biomimetic Nanomedicines for the Treatment of Arthritis. Pharmaceutics 2023; 15:1150. [PMID: 37111636 PMCID: PMC10146206 DOI: 10.3390/pharmaceutics15041150] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Revised: 03/26/2023] [Accepted: 04/02/2023] [Indexed: 04/08/2023] Open
Abstract
Arthritis is the inflammation and tenderness of the joints because of some metabolic, infectious, or constitutional reasons. Existing arthritis treatments help in controlling the arthritic flares, but more advancement is required to cure arthritis meticulously. Biomimetic nanomedicine represents an exceptional biocompatible treatment to cure arthritis by minimizing the toxic effect and eliminating the boundaries of current therapeutics. Various intracellular and extracellular pathways can be targeted by mimicking the surface, shape, or movement of the biological system to form a bioinspired or biomimetic drug delivery system. Different cell-membrane-coated biomimetic systems, and extracellular-vesicle-based and platelets-based biomimetic systems represent an emerging and efficient class of therapeutics to treat arthritis. The cell membrane from various cells such as RBC, platelets, macrophage cells, and NK cells is isolated and utilized to mimic the biological environment. Extracellular vesicles isolated from arthritis patients can be used as diagnostic tools, and plasma or MSCs-derived extracellular vesicles can be used as a therapeutic target for arthritis. Biomimetic systems guide the nanomedicines to the targeted site by hiding them from the surveillance of the immune system. Nanomedicines can be functionalized using targeted ligand and stimuli-responsive systems to reinforce their efficacy and minimize off-target effects. This review expounds on various biomimetic systems and their functionalization for the therapeutic targets of arthritis treatment, and discusses the challenges for the clinical translation of the biomimetic system.
Collapse
Affiliation(s)
| | | | | | | | - Rakesh Kumar Tekade
- National Institute of Pharmaceutical Education and Research (NIPER), Ahmedabad, Department of Pharmaceuticals, Ministry of Chemicals and Fertilizers, Opposite Air Force Station, Palaj, Gandhinagar 382355, Gujarat, India
| |
Collapse
|
5
|
Chi XK, Xu XL, Chen BY, Su J, Du YZ. Combining nanotechnology with monoclonal antibody drugs for rheumatoid arthritis treatments. J Nanobiotechnology 2023; 21:105. [PMID: 36964609 PMCID: PMC10039584 DOI: 10.1186/s12951-023-01857-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Accepted: 03/15/2023] [Indexed: 03/26/2023] Open
Abstract
Rheumatoid arthritis (RA) is a systemic immune disease characterized by synovial inflammation. Patients with RA commonly experience significant damage to their hand and foot joints, which can lead to joint deformities and even disability. Traditional treatments have several clinical drawbacks, including unclear pharmacological mechanisms and serious side effects. However, the emergence of antibody drugs offers a promising approach to overcome these limitations by specifically targeting interleukin-1 (IL-1), interleukin-6 (IL-6), tumor necrosis factor-alpha (TNF-α), and other cytokines that are closely related to the onset of RA. This approach reduces the incidence of adverse effects and contributes to significant therapeutic outcomes. Furthermore, combining these antibody drugs with drug delivery nanosystems (DDSs) can improve their tissue accumulation and bioavailability.Herein, we provide a summary of the pathogenesis of RA, the available antibody drugs and DDSs that improve the efficacy of these drugs. However, several challenges need to be addressed in their clinical applications, including patient compliance, stability, immunogenicity, immunosupression, target and synergistic effects. We propose strategies to overcome these limitations. In summary, we are optimistic about the prospects of treating RA with antibody drugs, given their specific targeting mechanisms and the potential benefits of combining them with DDSs.
Collapse
Affiliation(s)
- Xiao-Kai Chi
- College of Pharmacy, Jiamusi University, 258 Xuefu Road, Jiamusi, 154007, China
- Shulan International Medical College, Zhejiang Shuren University), 8 Shuren Street, Hangzhou, 310015, China
- Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, 866 Yu-Hang-Tang Road, Hangzhou, 310058, China
| | - Xiao-Ling Xu
- Shulan International Medical College, Zhejiang Shuren University), 8 Shuren Street, Hangzhou, 310015, China.
| | - Bang-Yao Chen
- Shulan International Medical College, Zhejiang Shuren University), 8 Shuren Street, Hangzhou, 310015, China
| | - Jin Su
- College of Pharmacy, Jiamusi University, 258 Xuefu Road, Jiamusi, 154007, China.
| | - Yong-Zhong Du
- Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, 866 Yu-Hang-Tang Road, Hangzhou, 310058, China.
| |
Collapse
|
6
|
Chitosan-based biomaterials for the treatment of bone disorders. Int J Biol Macromol 2022; 215:346-367. [PMID: 35718150 DOI: 10.1016/j.ijbiomac.2022.06.079] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Revised: 06/06/2022] [Accepted: 06/11/2022] [Indexed: 12/22/2022]
Abstract
Bone is an alive and dynamic organ that is well-differentiated and originated from mesenchymal tissues. Bone undergoes continuous remodeling during the lifetime of an individual. Although knowledge regarding bones and their disorders has been constantly growing, much attention has been devoted to effective treatments that can be used, both from materials and medical performance points of view. Polymers derived from natural sources, for example polysaccharides, are generally biocompatible and are therefore considered excellent candidates for various biomedical applications. This review outlines the development of chitosan-based biomaterials for the treatment of bone disorders including bone fracture, osteoporosis, osteoarthritis, arthritis rheumatoid, and osteosarcoma. Different examples of chitosan-based formulations in the form of gels, micro/nanoparticles, and films are discussed herein. The work also reviews recent patents and important developments related to the use of chitosan in the treatment of bone disorders. Although most of the cited research was accomplished before reaching the clinical application level, this manuscript summarizes the latest achievements within chitosan-based biomaterials used for the treatment of bone disorders and provides perspectives for future scientific activities.
Collapse
|
7
|
Wang J, Xiao L, Wang W, Zhang D, Ma Y, Zhang Y, Wang X. The Auxiliary Role of Heparin in Bone Regeneration and its Application in Bone Substitute Materials. Front Bioeng Biotechnol 2022; 10:837172. [PMID: 35646879 PMCID: PMC9133562 DOI: 10.3389/fbioe.2022.837172] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Accepted: 04/13/2022] [Indexed: 11/18/2022] Open
Abstract
Bone regeneration in large segmental defects depends on the action of osteoblasts and the ingrowth of new blood vessels. Therefore, it is important to promote the release of osteogenic/angiogenic growth factors. Since the discovery of heparin, its anticoagulant, anti-inflammatory, and anticancer functions have been extensively studied for over a century. Although the application of heparin is widely used in the orthopedic field, its auxiliary effect on bone regeneration is yet to be unveiled. Specifically, approximately one-third of the transforming growth factor (TGF) superfamily is bound to heparin and heparan sulfate, among which TGF-β1, TGF-β2, and bone morphogenetic protein (BMP) are the most common growth factors used. In addition, heparin can also improve the delivery and retention of BMP-2 in vivo promoting the healing of large bone defects at hyper physiological doses. In blood vessel formation, heparin still plays an integral part of fracture healing by cooperating with the platelet-derived growth factor (PDGF). Importantly, since heparin binds to growth factors and release components in nanomaterials, it can significantly facilitate the controlled release and retention of growth factors [such as fibroblast growth factor (FGF), BMP, and PDGF] in vivo. Consequently, the knowledge of scaffolds or delivery systems composed of heparin and different biomaterials (including organic, inorganic, metal, and natural polymers) is vital for material-guided bone regeneration research. This study systematically reviews the structural properties and auxiliary functions of heparin, with an emphasis on bone regeneration and its application in biomaterials under physiological conditions.
Collapse
Affiliation(s)
- Jing Wang
- Department of Orthopaedic Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Lan Xiao
- Centre for Biomedical Technologies, School of Mechanical, Medical and Process Engineering, Queensland University of Technology, Brisbane, Australia
- Australia−China Centre for Tissue Engineering and Regenerative Medicine, Brisbane, Australia
| | - Weiqun Wang
- Department of Orthopaedic Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Dingmei Zhang
- Department of Orthopaedic Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Yaping Ma
- Department of Orthopaedic Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Yi Zhang
- Department of Hygiene Toxicology, School of Public Health, Zunyi Medical University, Zunyi, China
| | - Xin Wang
- Department of Orthopaedic Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi, China
- Centre for Biomedical Technologies, School of Mechanical, Medical and Process Engineering, Queensland University of Technology, Brisbane, Australia
- Australia−China Centre for Tissue Engineering and Regenerative Medicine, Brisbane, Australia
| |
Collapse
|
8
|
Schindeler A, Lee LR, O'Donohue AK, Ginn SL, Munns CF. Curative Cell and Gene Therapy for Osteogenesis Imperfecta. J Bone Miner Res 2022; 37:826-836. [PMID: 35306687 PMCID: PMC9324990 DOI: 10.1002/jbmr.4549] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/04/2021] [Revised: 02/03/2022] [Accepted: 02/27/2022] [Indexed: 11/17/2022]
Abstract
Osteogenesis imperfecta (OI) describes a series of genetic bone fragility disorders that can have a substantive impact on patient quality of life. The multidisciplinary approach to management of children and adults with OI primarily involves the administration of antiresorptive medication, allied health (physiotherapy and occupational therapy), and orthopedic surgery. However, advances in gene editing technology and gene therapy vectors bring with them the promise of gene-targeted interventions to provide an enduring or perhaps permanent cure for OI. This review describes emergent technologies for cell- and gene-targeted therapies, major hurdles to their implementation, and the prospects of their future success with a focus on bone disorders. © 2022 The Authors. Journal of Bone and Mineral Research published by Wiley Periodicals LLC on behalf of American Society for Bone and Mineral Research (ASBMR).
Collapse
Affiliation(s)
- Aaron Schindeler
- Bioengineering and Molecular Medicine Laboratorythe Children's Hospital at Westmead and the Westmead Institute for Medical ResearchWestmeadAustralia
- Children's Hospital Westmead Clinical SchoolUniversity of SydneyCamperdownAustralia
| | - Lucinda R Lee
- Bioengineering and Molecular Medicine Laboratorythe Children's Hospital at Westmead and the Westmead Institute for Medical ResearchWestmeadAustralia
- Children's Hospital Westmead Clinical SchoolUniversity of SydneyCamperdownAustralia
| | - Alexandra K O'Donohue
- Bioengineering and Molecular Medicine Laboratorythe Children's Hospital at Westmead and the Westmead Institute for Medical ResearchWestmeadAustralia
- Children's Hospital Westmead Clinical SchoolUniversity of SydneyCamperdownAustralia
| | - Samantha L Ginn
- Gene Therapy Research Unit, Children's Medical Research Institute, Faculty of Medicine and HealthThe University of Sydney and Sydney Children's Hospitals NetworkWestmeadAustralia
| | - Craig F Munns
- Faculty of MedicineThe University of QueenslandBrisbaneQLDAustralia
- Department of Endocrinology and DiabetesQueensland Children's HospitalBrisbaneQLDAustralia
- Child Health Research Centre and Faculty of MedicineThe University of QueenslandBrisbaneQueenslandAustralia
| |
Collapse
|
9
|
Chitosan-Based Nanogels: Synthesis and Toxicity Profile for Drug Delivery to Articular Joints. NANOMATERIALS 2022; 12:nano12081337. [PMID: 35458048 PMCID: PMC9027118 DOI: 10.3390/nano12081337] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Revised: 04/06/2022] [Accepted: 04/09/2022] [Indexed: 12/20/2022]
Abstract
One important challenge in treating avascular-degraded cartilage is the development of new drugs for both pain management and joint preservation. Considerable efforts have been invested in developing nanosystems using biomaterials, such as chitosan, a widely used natural polymer exhibiting numerous advantages, i.e., non-toxic, biocompatible and biodegradable. However, even if chitosan is generally recognized as safe, the safety and biocompatibility of such nanomaterials must be addressed because of potential for greater interactions between nanomaterials and biological systems. Here, we developed chitosan-based nanogels as drug-delivery platforms and established an initial biological risk assessment for osteocartilaginous applications. We investigated the influence of synthesis parameters on the physicochemical characteristics of the resulting nanogels and their potential impact on the biocompatibility on all types of human osteocartilaginous cells. Monodisperse nanogels were synthesized with sizes ranging from 268 to 382 nm according to the acidic solution used (i.e., either citric or acetic acid) with overall positive charge surface. Our results demonstrated that purified chitosan-based nanogels neither affected cell proliferation nor induced nitric oxide production in vitro. However, nanogels were moderately genotoxic in a dose-dependent manner but did not significantly induce acute embryotoxicity in zebrafish embryos, up to 100 µg∙mL−1. These encouraging results hold great promise for the intra-articular delivery of drugs or diagnostic agents for joint pathologies.
Collapse
|
10
|
Brain D, Plant-Hately A, Heaton B, Arshad U, David C, Hedrich C, Owen A, Liptrott NJ. Drug delivery systems as immunomodulators for therapy of infectious disease: Relevance to COVID-19. Adv Drug Deliv Rev 2021; 178:113848. [PMID: 34182016 PMCID: PMC8233062 DOI: 10.1016/j.addr.2021.113848] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Revised: 06/10/2021] [Accepted: 06/22/2021] [Indexed: 02/07/2023]
Abstract
The emergence of SARS-CoV-2, and the ensuing global pandemic, has resulted in an unprecedented response to identify therapies that can limit uncontrolled inflammation observed in patients with moderate to severe COVID-19. The immune pathology behind COVID-19 is complex and involves the activation and interaction of multiple systems including, but not limited to, complement, inflammasomes, endothelial as well as innate and adaptive immune cells to bring about a convoluted profile of inflammation, coagulation and tissue damage. To date, therapeutic approaches have focussed on inhibition of coagulation, untargeted immune suppression and/or cytokine-directed blocking agents. Regardless of recently achieved improvements in individual patient outcomes and survival rates, improved and focussed approaches targeting individual systems involved is needed to further improve prognosis and wellbeing. This review summarizes the current understanding of molecular and cellular systems involved in the pathophysiology of COVID-19, and their contribution to pathogen clearance and damage to then discuss possible therapeutic options involving immunomodulatory drug delivery systems as well as summarising the complex interplay between them.
Collapse
Affiliation(s)
- Danielle Brain
- Immunocompatibility Group, Department of Pharmacology and Therapeutics, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool, UK; Centre of Excellence for Long-acting Therapeutics (CELT), Department of Pharmacology and Therapeutics, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool, UK
| | - Alex Plant-Hately
- Immunocompatibility Group, Department of Pharmacology and Therapeutics, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool, UK; Centre of Excellence for Long-acting Therapeutics (CELT), Department of Pharmacology and Therapeutics, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool, UK
| | - Bethany Heaton
- Immunocompatibility Group, Department of Pharmacology and Therapeutics, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool, UK; Centre of Excellence for Long-acting Therapeutics (CELT), Department of Pharmacology and Therapeutics, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool, UK
| | - Usman Arshad
- Centre of Excellence for Long-acting Therapeutics (CELT), Department of Pharmacology and Therapeutics, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool, UK
| | - Christopher David
- Immunocompatibility Group, Department of Pharmacology and Therapeutics, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool, UK; Centre of Excellence for Long-acting Therapeutics (CELT), Department of Pharmacology and Therapeutics, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool, UK
| | - Christian Hedrich
- Department of Women's & Children's Health, Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool, UK; Department of Rheumatology, Alder Hey Children's NHS Foundation Trust, Liverpool, UK
| | - Andrew Owen
- Centre of Excellence for Long-acting Therapeutics (CELT), Department of Pharmacology and Therapeutics, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool, UK
| | - Neill J Liptrott
- Immunocompatibility Group, Department of Pharmacology and Therapeutics, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool, UK; Centre of Excellence for Long-acting Therapeutics (CELT), Department of Pharmacology and Therapeutics, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool, UK.
| |
Collapse
|
11
|
Nanoparticles to Target and Treat Macrophages: The Ockham's Concept? Pharmaceutics 2021; 13:pharmaceutics13091340. [PMID: 34575416 PMCID: PMC8469871 DOI: 10.3390/pharmaceutics13091340] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Revised: 08/15/2021] [Accepted: 08/19/2021] [Indexed: 12/19/2022] Open
Abstract
Nanoparticles are nanomaterials with three external nanoscale dimensions and an average size ranging from 1 to 1000 nm. Nanoparticles have gained notoriety in technological advances due to their tunable physical, chemical, and biological characteristics. However, the administration of functionalized nanoparticles to living beings is still challenging due to the rapid detection and blood and tissue clearance by the mononuclear phagocytic system. The major exponent of this system is the macrophage. Regardless the nanomaterial composition, macrophages can detect and incorporate foreign bodies by phagocytosis. Therefore, the simplest explanation is that any injected nanoparticle will be probably taken up by macrophages. This explains, in part, the natural accumulation of most nanoparticles in the spleen, lymph nodes, and liver (the main organs of the mononuclear phagocytic system). For this reason, recent investigations are devoted to design nanoparticles for specific macrophage targeting in diseased tissues. The aim of this review is to describe current strategies for the design of nanoparticles to target macrophages and to modulate their immunological function involved in different diseases with special emphasis on chronic inflammation, tissue regeneration, and cancer.
Collapse
|
12
|
The pathophysiology of immunoporosis: innovative therapeutic targets. Inflamm Res 2021; 70:859-875. [PMID: 34272579 DOI: 10.1007/s00011-021-01484-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2020] [Revised: 04/14/2021] [Accepted: 06/08/2021] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND The physiological balance between bone resorption and bone formation is now known to be mediated by a cascade of events parallel to the classic osteoblast-osteoclast interaction. Thus, osteoimmunology now encompasses the role played by other cell types, such as cytokines, lymphocytes and chemokines, in immunological responses and how they help modulate bone metabolism. All these factors have an impact on the RANK/RANKL/OPG pathway, which is the major pathway for the maturation and resorption activity of osteoclast precursor cells, responsible for osteoporosis development. Recently, immunoporosis has emerged as a new research area in osteoimmunology dedicated to the immune system's role in osteoporosis. METHODS The first part of this review presents theoretical concepts on the factors involved in the skeletal system and osteoimmunology. Secondly, existing treatments and novel therapeutic approaches to treat osteoporosis are summarized. These were selected from to the most recent studies published on PubMed containing the term osteoporosis. All data relate to the results of in vitro and in vivo studies on the osteoimmunological system of humans, mice and rats. FINDINGS Treatments for osteoporosis can be classified into two categories. They either target osteoclastogenesis inhibition (denosumab, bisphosphonates), or they aim to restore the number and function of osteoblasts (romozumab, abaloparatide). Even novel therapies, such as resolvins, gene therapy, and mesenchymal stem cell transplantation, fall within this classification system. CONCLUSION This review presents alternative pathways in the pathophysiology of osteoporosis, along with some recent therapeutic breakthroughs to restore bone homeostasis.
Collapse
|
13
|
Zhang G, Ma L, Bai L, Li M, Guo T, Tian B, He Z, Fu Q. Inflammatory microenvironment-targeted nanotherapies. J Control Release 2021; 334:114-126. [PMID: 33887284 DOI: 10.1016/j.jconrel.2021.04.018] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 04/16/2021] [Accepted: 04/17/2021] [Indexed: 12/14/2022]
Abstract
Inflammatory microenvironments (IMEs) are common pathological characteristics and drive the development of multiple chronic diseases. Thus, IME-targeted therapies exhibit potential for the treatment of inflammatory diseases. Nanoplatforms have significant advantages in improving the efficiency of anti-inflammatory treatments. Owing to their improved therapeutic effects and reduced side effects, IME-targeted nanotherapies have recently drawn interest from the research community. This review introduces IMEs and discusses the application of IME-targeted nanotherapies for inflammatory diseases. The development of rational targeting strategies tailored to IMEs in damaged tissues can help promote therapies for chronic diseases.
Collapse
Affiliation(s)
- Guangshuai Zhang
- Wuya College of Innovation, Shenyang Pharmaceutical University, No. 103, Wenhua Road, Shenyang 110016, China.
| | - Lixue Ma
- Wuya College of Innovation, Shenyang Pharmaceutical University, No. 103, Wenhua Road, Shenyang 110016, China
| | - Lijun Bai
- Wuya College of Innovation, Shenyang Pharmaceutical University, No. 103, Wenhua Road, Shenyang 110016, China
| | - Mo Li
- Liaoning Institute for Drug Control, No. 7 Chongshan West Road, Shenyang 110016, China
| | - Tiange Guo
- Laboratory Animal Department, General Hospital of Northern Theater Command, No. 83, Wenhua Road, Shenyang 110016, China
| | - Baocheng Tian
- School of Pharmacy, Binzhou Medical University, No. 346, Guanhai Road, Yantai 264003, China
| | - Zhonggui He
- Wuya College of Innovation, Shenyang Pharmaceutical University, No. 103, Wenhua Road, Shenyang 110016, China
| | - Qiang Fu
- Wuya College of Innovation, Shenyang Pharmaceutical University, No. 103, Wenhua Road, Shenyang 110016, China.
| |
Collapse
|
14
|
Zhao J, Chen X, Ho KH, Cai C, Li CW, Yang M, Yi C. Nanotechnology for diagnosis and therapy of rheumatoid arthritis: Evolution towards theranostic approaches. CHINESE CHEM LETT 2021. [DOI: 10.1016/j.cclet.2020.11.048] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
|
15
|
Liu Y, Cao F, Sun B, Bellanti JA, Zheng SG. Magnetic nanoparticles: A new diagnostic and treatment platform for rheumatoid arthritis. J Leukoc Biol 2020; 109:415-424. [PMID: 32967052 DOI: 10.1002/jlb.5mr0420-008rr] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2020] [Revised: 04/19/2020] [Accepted: 04/21/2020] [Indexed: 12/30/2022] Open
Abstract
Rheumatoid arthritis (RA) is a chronic inflammatory condition characterized by articular synovitis that eventually leads to the destruction of cartilage and bone in the joints with resulting pain and disability. The current therapies for RA are divided into 4 categories: non-steroidal anti-inflammatory drugs (NSAIDs), glucocorticoids, nonbiological disease-modifying anti-rheumatic drugs (DMARDs), and biological DMARDs. Each drug grouping is beset with significant setbacks that not only include limited drug bioavailability and high clearance, but also varying degrees of drug toxicity to normal tissues. Recently, nanotechnology has provided a promising tool for the development of novel therapeutic and diagnostic systems in the area of malignant and inflammatory diseases. Among these, magnetic nanoparticles (MNPs) have provided an attractive carrier option for delivery of therapeutic agents. Armed with an extra magnetic probe, MNPs are capable of more accurately targeting the local lesion with avoidance of unpleasant systemic side effects. This review aims to provide an introduction to the applications of magnetic nanoparticles in RA, focusing on the latest advances, challenges, and opportunities for future development.
Collapse
Affiliation(s)
- Yan Liu
- Institute of Clinical Immunology Center, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Fenglin Cao
- Department of Internal Medicine, the First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Baoqing Sun
- Department of Allergy and Clinical Immunology, Guangzhou Institute of Respiratory Health, State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Medical University, First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Joseph A Bellanti
- Department of Pediatrics and Microbiology-Immunology, Georgetown University Medical Center, Washington, District of Columbia, United States
| | - Song Guo Zheng
- Department of Internal Medicine, Ohio State University College of Medicine and Wexner Medical Center, Columbus, Ohio, United States
| |
Collapse
|
16
|
Molupe N, Babu B, Oluwole DO, Prinsloo E, Gai L, Shen Z, Mack J, Nyokong T. Photodynamic activity of 2,6-diiodo-3,5-dithienylvinyleneBODIPYs and their folate-functionalized chitosan-coated Pluronic® F-127 micelles on MCF-7 breast cancer cells. J PORPHYR PHTHALOCYA 2020. [DOI: 10.1142/s1088424619501773] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
A 2,6-diiodo-3,5-dithienylvinyleneBODIPY dye was prepared and encapsulated with folate-chitosan capped Pluronic[Formula: see text] F-127 to provide drug delivery systems for photodynamic therapy (PDT). Moderately enhanced singlet oxygen quantum yields were observed for the dye encapsulation complexes in water. The in vitro dark cytotoxicity and photodynamic activity were investigated on the human breast adenocarcinoma (MCF-7) cell line. Minimal dark cytotoxicity was observed for the BODIPY dyes in 5% DMSO and when encapsulated in folate-functionalized chitosan-coated Pluronic[Formula: see text] F-127 micelles, since the cell viability values are consistently greater than 80% over the 0-40 [Formula: see text] concentration range. Upon irradiation of the samples, significant cytocidal activity was observed for the encapsulation complex of a 2,6-diiodo-8-dimethylaminophenyl-3,5-dithienylvinyleneBODIPY dye with less than 50% viable cells observed at concentrations [Formula: see text].
Collapse
Affiliation(s)
- Nthabeleng Molupe
- Institute for Nanotechnology Innovation, Department of Chemistry, Rhodes University, Makhanda 6140, South Africa
| | - Balaji Babu
- Institute for Nanotechnology Innovation, Department of Chemistry, Rhodes University, Makhanda 6140, South Africa
| | - David O. Oluwole
- Institute for Nanotechnology Innovation, Department of Chemistry, Rhodes University, Makhanda 6140, South Africa
| | - Earl Prinsloo
- Biotechnology Innovation Centre, Rhodes University, Makhanda 6140, South Africa
| | - Lizhi Gai
- State Key Laboratory of Coordination Chemistry, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210023, P. R. China
| | - Zhen Shen
- State Key Laboratory of Coordination Chemistry, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210023, P. R. China
| | - John Mack
- Institute for Nanotechnology Innovation, Department of Chemistry, Rhodes University, Makhanda 6140, South Africa
| | - Tebello Nyokong
- Institute for Nanotechnology Innovation, Department of Chemistry, Rhodes University, Makhanda 6140, South Africa
| |
Collapse
|
17
|
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: 1.3] [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
|
18
|
Qindeel M, Ahmed N, Khan GM, Rehman AU. Ligand decorated chitosan as an advanced nanocarrier for targeted delivery: a critical review. Nanomedicine (Lond) 2019; 14:1623-1642. [PMID: 31166147 DOI: 10.2217/nnm-2018-0490] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Nontargeted delivery systems present nonspecific delivery, low transfection efficiency and high toxicity. Ligand-conjugated chitosan (CS) nanocarriers have emerged as an outstanding option for achieving active delivery specifically and preferentially to the target sites by exploiting receptors mediated endocytosis. Mannosylated CS nanocarriers have brought tremendous breakthrough in gene therapy and have proven to be an excellent choice for treatment of infectious and inflammatory diseases. Similarly, folate and antibodies-conjugated CS play a significant role in diagnosis and treatment of various cancers. Current evidences obviously propose ligand-decorated CS as an attractive option for diagnosis and treatment of dreadful conditions. In order to bring huge revolution in the field of targeted delivery, challenges associated with these nanocarriers needs to be addressed.
Collapse
Affiliation(s)
- Maimoona Qindeel
- Department of Pharmacy, Quaid-i-Azam University, Islamabad 45320, Pakistan
| | - Naveed Ahmed
- Department of Pharmacy, Quaid-i-Azam University, Islamabad 45320, Pakistan
| | - Gul Majid Khan
- Department of Pharmacy, Quaid-i-Azam University, Islamabad 45320, Pakistan
| | - Asim Ur Rehman
- Department of Pharmacy, Quaid-i-Azam University, Islamabad 45320, Pakistan
| |
Collapse
|
19
|
Wang X, Shao J, Abd El Raouf M, Xie H, Huang H, Wang H, Chu PK, Yu XF, Yang Y, AbdEl-Aal AM, Mekkawy NH, Miron RJ, Zhang Y. Near-infrared light-triggered drug delivery system based on black phosphorus for in vivo bone regeneration. Biomaterials 2018; 179:164-174. [DOI: 10.1016/j.biomaterials.2018.06.039] [Citation(s) in RCA: 70] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2018] [Revised: 06/07/2018] [Accepted: 06/28/2018] [Indexed: 10/28/2022]
|
20
|
Pirmardvand Chegini S, Varshosaz J, Taymouri S. Recent approaches for targeted drug delivery in rheumatoid arthritis diagnosis and treatment. ARTIFICIAL CELLS NANOMEDICINE AND BIOTECHNOLOGY 2018; 46:502-514. [PMID: 29661045 DOI: 10.1080/21691401.2018.1460373] [Citation(s) in RCA: 61] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Rheumatoid arthritis (RA) is a chronic inflammatory disease with complex pathology characterized by inflammation of joints, devastation of the synovium, pannus formation, bones and cartilage destruction and often is associated with persistent arthritic pain, swelling, stiffness and work disability. In conventional RA therapy, because of short biological half-life, poor bioavailability, high and frequent dosing is required. Thereby, these anti-RA medications, which unable to selectively target affected zone, may cause severe side effects in extra-articular tissues. Today, nanotechnology has emerged as promising tool in the development of novel drug delivery systems for the treatment and diagnosis of intractable diseases such as RA. Active targeting in RA nanomedicine has also been introduced a successful way for facilitating specific uptake of therapeutic agents by the disease cells. In this review, it is attempted to describe various targeted drug delivery systems (localized and receptor-based) used for RA diagnosis and therapy. Then, we highlight recent developments related to various non-viral gene delivery systems for RA gene therapy.
Collapse
Affiliation(s)
- Sana Pirmardvand Chegini
- a Department of Pharmaceutics, School of Pharmacy and Novel Drug Delivery Systems Research Centre , Isfahan University of Medical Sciences , Isfahan , Iran
| | - Jaleh Varshosaz
- a Department of Pharmaceutics, School of Pharmacy and Novel Drug Delivery Systems Research Centre , Isfahan University of Medical Sciences , Isfahan , Iran
| | - Somayeh Taymouri
- a Department of Pharmaceutics, School of Pharmacy and Novel Drug Delivery Systems Research Centre , Isfahan University of Medical Sciences , Isfahan , Iran
| |
Collapse
|
21
|
Chuang SY, Lin CH, Huang TH, Fang JY. Lipid-Based Nanoparticles as a Potential Delivery Approach in the Treatment of Rheumatoid Arthritis. NANOMATERIALS 2018; 8:nano8010042. [PMID: 29342965 PMCID: PMC5791129 DOI: 10.3390/nano8010042] [Citation(s) in RCA: 75] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/24/2017] [Revised: 01/10/2018] [Accepted: 01/12/2018] [Indexed: 12/23/2022]
Abstract
Rheumatoid arthritis (RA), a chronic and joint-related autoimmune disease, results in immune dysfunction and destruction of joints and cartilages. Small molecules and biological therapies have been applied in a wide variety of inflammatory disorders, but their utility as a therapeutic agent is limited by poor absorption, rapid metabolism, and serious side effects. To improve these limitations, nanoparticles, which are capable of encapsulating and protecting drugs from degradation before they reach the target site in vivo, may serve as drug delivery systems. The present research proposes a platform for different lipid nanoparticle approaches for RA therapy, taking advantage of the newly emerging field of lipid nanoparticles to develop a targeted theranostic system for application in the treatment of RA. This review aims to present the recent major application of lipid nanoparticles that provide a biocompatible and biodegradable delivery system to effectively improve RA targeting over free drugs via the presentation of tissue-specific targeting of ligand-controlled drug release by modulating nanoparticle composition.
Collapse
Affiliation(s)
- Shih-Yi Chuang
- Research Center for Food and Cosmetic Safety and Research Center for Chinese Herbal Medicine, Chang Gung University of Science and Technology, Kweishan, Taoyuan 333, Taiwan.
| | - Chih-Hung Lin
- Center for General Education, Chang Gung University of Science and Technology, Kweishan, Taoyuan 333, Taiwan.
| | - Tse-Hung Huang
- Department of Traditional Chinese Medicine, Chang Gung Memorial Hospital, Keelung 204, Taiwan.
- School of Traditional Chinese Medicine, Chang Gung University, Taoyuan 333, Taiwan.
- School of Nursing, National Taipei University of Nursing and Health Sciences, Taipei 112, Taiwan.
| | - Jia-You Fang
- Research Center for Food and Cosmetic Safety and Research Center for Chinese Herbal Medicine, Chang Gung University of Science and Technology, Kweishan, Taoyuan 333, Taiwan.
- Pharmaceutics Laboratory, Graduate Institute of Natural Products, Chang Gung University, Kweishan, Taoyuan 333, Taiwan.
- Chinese Herbal Medicine Research Team, Healthy Aging Research Center, Chang Gung University, Kweishan, Taoyuan 333, Taiwan.
- Department of Anesthesiology, Chang Gung Memorial Hospital, Linkou, Taoyuan 333, Taiwan.
| |
Collapse
|
22
|
Shi Q, Rondon-Cavanzo EP, Dalla Picola IP, Tiera MJ, Zhang X, Dai K, Benabdoune HA, Benderdour M, Fernandes JC. In vivo therapeutic efficacy of TNFα silencing by folate-PEG-chitosan-DEAE/siRNA nanoparticles in arthritic mice. Int J Nanomedicine 2018; 13:387-402. [PMID: 29391796 PMCID: PMC5769564 DOI: 10.2147/ijn.s146942] [Citation(s) in RCA: 54] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND Tumor necrosis factor-alpha (TNFα), a pro-inflammatory cytokine, has been shown to play a role in the pathophysiology of rheumatoid arthritis. Silencing TNFα expression with small interfering RNA (siRNA) is a promising approach to treatment of the condition. METHODS Towards this end, our team has developed a modified chitosan (CH) nanocarrier, deploying folic acid, diethylethylamine (DEAE) and polyethylene glycol (PEG) (folate-PEG-CH-DEAE15). The gene carrier protects siRNA against nuclease destruction, its ligands facilitate siRNA uptake via cell surface receptors, and it provides improved solubility at neutral pH with transport of its load into target cells. In the present study, nanoparticles were prepared with siRNA-TNFα, DEAE, and folic acid-CH derivative. Nanoparticle size and zeta potential were verified by dynamic light scattering. Their TNFα-knockdown effects were tested in a murine collagen antibody-induced arthritis model. TNFα expression was examined along with measurements of various cartilage and bone turnover markers by performing histology and microcomputed tomography analysis. RESULTS We demonstrated that folate-PEG-CH-DEAE15/siRNA nanoparticles did not alter cell viability, and significantly decreased inflammation, as demonstrated by improved clinical scores and lower TNFα protein concentrations in target tissues. This siRNA nanocarrier also decreased articular cartilage destruction and bone loss. CONCLUSION The results indicate that folate-PEG-CH-DEAE15 nanoparticles are a safe and effective platform for nonviral gene delivery of siRNA, and their potential clinical applications warrant further investigation.
Collapse
Affiliation(s)
- Qin Shi
- Orthopedic Research Laboratory, Hôpital du Sacré-Coeur de Montréal, Université de Montréal, Montréal, QC, Canada
| | - Elsa-Patricia Rondon-Cavanzo
- Orthopedic Research Laboratory, Hôpital du Sacré-Coeur de Montréal, Université de Montréal, Montréal, QC, Canada
| | - Isadora Pfeifer Dalla Picola
- Orthopedic Research Laboratory, Hôpital du Sacré-Coeur de Montréal, Université de Montréal, Montréal, QC, Canada
- Department of Chemistry and Environmental Sciences, UNESP-São Paulo State University, São José do Rio Preto, Brazil
| | - Marcio José Tiera
- Department of Chemistry and Environmental Sciences, UNESP-São Paulo State University, São José do Rio Preto, Brazil
| | - Xiaoling Zhang
- Orthopedic Cellular and Molecular Biology Laboratories, Institute of Health Sciences, Chinese Academy of Sciences and Shanghai Jiao Tong University School of Medicine, Shanghai
| | - Kerong Dai
- Department of Orthopedics, Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Houda Abir Benabdoune
- Orthopedic Research Laboratory, Hôpital du Sacré-Coeur de Montréal, Université de Montréal, Montréal, QC, Canada
| | - Mohamed Benderdour
- Orthopedic Research Laboratory, Hôpital du Sacré-Coeur de Montréal, Université de Montréal, Montréal, QC, Canada
| | - Julio Cesar Fernandes
- Orthopedic Research Laboratory, Hôpital du Sacré-Coeur de Montréal, Université de Montréal, Montréal, QC, Canada
| |
Collapse
|
23
|
Durymanov M, Kamaletdinova T, Lehmann SE, Reineke J. Exploiting passive nanomedicine accumulation at sites of enhanced vascular permeability for non-cancerous applications. J Control Release 2017. [DOI: 10.1016/j.jconrel.2017.06.013] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
24
|
Liang H, He L, Zhou B, Li B, Li J. Folate-functionalized assembly of low density lipoprotein/sodium carboxymethyl cellulose nanoparticles for targeted delivery. Colloids Surf B Biointerfaces 2017; 156:19-28. [PMID: 28499201 DOI: 10.1016/j.colsurfb.2017.05.004] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2017] [Revised: 04/25/2017] [Accepted: 05/01/2017] [Indexed: 02/06/2023]
Abstract
In this study, well-defined folate (FA)-functionalized low density lipoproteins (LDL)/sodium carboxymethyl cellulose (CMC) nanoparticles (NP) were first formulated, utilized in tumor targeting and pH-triggered drug release. CMC was modified with FA before the preparation of NP. A model anti-tumor drug, doxorubicin (DOX), was effectively loaded into the LDL/CMC-FA NP by ionic bonding and hydrophobic interactions. To enhance non-covalent encapsulation stability, self-assembly of DOX-loaded LDL/CMC-FA NP (NP-DOX) was cross-linked by multivalent cations such as Ca2+ (Ca2+-NP-DOX). The active targeting efficiency of NP-DOX and Ca2+-NP-DOX was tested against KB cells (FA-receptor over-expressing cells, FR+) and A549 cells (FA-receptor negative-expressing cells, FR-), using FA non-modified DOX-loaded LDL/CMC NP (NG-DOX) as control. The competition assay proved that free FA molecules prevented the cellular uptake of the NP by competitive binding to the FA receptors on the surface of KB cells. This new pH-responsive and FA-targeted nanocarrier may be a promising efficient drug delivery system for potential cancer therapy.
Collapse
Affiliation(s)
- Hongshan Liang
- College of Food Science and Technology, Huazhong Agricultural University, Wuhan 430070, China; Key Laboratory of Environment Correlative Dietology (Huazhong Agricultural University), Ministry of Education, China; Functional Food Enginnering & Technology Research Center of Hubei Province, Wuhan, China
| | - Lei He
- College of Food Science and Technology, Huazhong Agricultural University, Wuhan 430070, China; Key Laboratory of Environment Correlative Dietology (Huazhong Agricultural University), Ministry of Education, China; Functional Food Enginnering & Technology Research Center of Hubei Province, Wuhan, China
| | - Bin Zhou
- College of Food Science and Technology, Shanghai Ocean University, LinGang New City, Shanghai, 201306, China
| | - Bin Li
- College of Food Science and Technology, Huazhong Agricultural University, Wuhan 430070, China; Key Laboratory of Environment Correlative Dietology (Huazhong Agricultural University), Ministry of Education, China; Functional Food Enginnering & Technology Research Center of Hubei Province, Wuhan, China
| | - Jing Li
- College of Food Science and Technology, Huazhong Agricultural University, Wuhan 430070, China; Key Laboratory of Environment Correlative Dietology (Huazhong Agricultural University), Ministry of Education, China; Functional Food Enginnering & Technology Research Center of Hubei Province, Wuhan, China.
| |
Collapse
|
25
|
Lin YY, Jean YH, Lee HP, Lin SC, Pan CY, Chen WF, Wu SF, Su JH, Tsui KH, Sheu JH, Sung PJ, Wen ZH. Excavatolide B Attenuates Rheumatoid Arthritis through the Inhibition of Osteoclastogenesis. Mar Drugs 2017; 15:md15010009. [PMID: 28067799 PMCID: PMC5295229 DOI: 10.3390/md15010009] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2016] [Revised: 12/17/2016] [Accepted: 12/26/2016] [Indexed: 02/08/2023] Open
Abstract
Osteoclasts are multinucleated giant cells of macrophage/monocyte lineage, and cell differentiation with the upregulation of osteoclast-related proteins is believed to play a major role in the destruction of the joints in the course of rheumatoid arthritis (RA). Pro-inflammatory cytokines, such as interleukin-17A (IL-17A) and macrophage colony-stimulating factor (M-CSF), can be overexpressed in RA and lead to osteoclastogenesis. In a previous study, we found that cultured-type soft coral-derived excavatolide B (Exc-B) exhibited anti-inflammatory properties. In the present study, we thus aimed to evaluate the anti-arthritic activity of Exc-B in in vitro and in vivo models. The results demonstrated that Exc-B inhibits LPS-induced multinucleated cell and actin ring formation, as well as TRAP, MMP-9, and cathepsin K expression. Additionally, Exc-B significantly attenuated the characteristics of RA in adjuvant (AIA) and type II collagen-induced arthritis (CIA) in rats. Moreover, Exc-B improved histopathological features, and reduced the number of TRAP-positive multinucleated cells in the in vivo AIA and CIA models. Immunohistochemical analysis showed that Exc-B attenuated the protein expression of cathepsin K, MMP-2, MMP-9, CD11b, and NFATc1 in ankle tissues of AIA and CIA rats. Level of interleukin-17A and macrophage colony-stimulating factor were also decreased by Exc-B. These findings strongly suggest that Exc-B could be of potential use as a therapeutic agent by inhibiting osteoclast differentiation in arthritis. Moreover, this study also illustrates the use of the anti-inflammatory marine compound, Exc-B, as a potential therapeutic strategy for RA.
Collapse
Affiliation(s)
- Yen-You Lin
- Department of Marine Biotechnology and Resources, National Sun Yat-sen University, No.70, Lianhai Road, Gushan District, Kaohsiung 80424, Taiwan.
| | - Yen-Hsuan Jean
- Department of Orthopaedic Surgery, Ping-Tung Christian Hospital, No.60, Dalian Road, Pingtung 90059, Taiwan.
| | - Hsin-Pai Lee
- Department of Orthopaedic Surgery, Ping-Tung Christian Hospital, No.60, Dalian Road, Pingtung 90059, Taiwan.
| | - Sung-Chun Lin
- Department of Orthopaedic Surgery, Ping-Tung Christian Hospital, No.60, Dalian Road, Pingtung 90059, Taiwan.
| | - Chieh-Yu Pan
- Department and Graduate Institute of Aquaculture, National Kaohsiung Marine University, No.142, Haizhuan Road, Nanzi District, Kaohsiung 81157, Taiwan.
| | - Wu-Fu Chen
- Department of Marine Biotechnology and Resources, National Sun Yat-sen University, No.70, Lianhai Road, Gushan District, Kaohsiung 80424, Taiwan.
- Department of Neurosurgery, Chang Gung Memorial Hospital-Kaohsiung Medical Center and Chang Gung University College of Medicine, No.123, Dapi Road, Niaosong District, Kaohsiung 83301, Taiwan.
- Department of Neurosurgery, Xiamen Chang Gung Memorial Hospital, No.123, Xiafei Road, Haicang District, Xiamen 361000, China.
| | - Shu-Fen Wu
- Department of Life Science, Institute of Molecular Biology, National Chung-Cheng University, No.168, Sec. 1, University Road, Min-Hsiung, Chia-yi 62102, Taiwan.
| | - Jui-Hsin Su
- Taiwan Coral Research Center, National Museum of Marine Biology & Aquarium, No.2 Houwan Road, Checheng, Pingtung 94450, Taiwan.
- Graduate Institute of Marine Biotechnology, National Dong Hwa University, No.2 Houwan Road, Checheng, Pingtung 94450, Taiwan.
| | - Kuan-Hao Tsui
- Department of Obstetrics and Gynecology, Kaohsiung Veterans General Hospital, No.386, Dazhong 1st Road, Zuoying District, Kaohsiung 81362, Taiwan.
- Department of Obstetrics and Gynecology and Institute of Clinical Medicine, National Yang-Ming University, No.155, Sec. 2, Linong Street, Taipei 11221, Taiwan.
- Department of Pharmacy and Graduate Institute of Pharmaceutical Technology, Tajen University, No.20, Weixin Road, Yanpu, Pingtung 90741, Taiwan.
| | - Jyh-Horng Sheu
- Department of Marine Biotechnology and Resources, National Sun Yat-sen University, No.70, Lianhai Road, Gushan District, Kaohsiung 80424, Taiwan.
- Doctoral Degree Program in Marine Biotechnology, National Sun Yat-sen University and Academia Sinica, No.70, Lianhai Road, Gushan District, Kaohsiung 80424, Taiwan.
| | - Ping-Jyun Sung
- Taiwan Coral Research Center, National Museum of Marine Biology & Aquarium, No.2 Houwan Road, Checheng, Pingtung 94450, Taiwan.
- Graduate Institute of Marine Biotechnology, National Dong Hwa University, No.2 Houwan Road, Checheng, Pingtung 94450, Taiwan.
| | - Zhi-Hong Wen
- Department of Marine Biotechnology and Resources, National Sun Yat-sen University, No.70, Lianhai Road, Gushan District, Kaohsiung 80424, Taiwan.
- Doctoral Degree Program in Marine Biotechnology, National Sun Yat-sen University and Academia Sinica, No.70, Lianhai Road, Gushan District, Kaohsiung 80424, Taiwan.
| |
Collapse
|
26
|
Utilization of nanoparticle technology in rheumatoid arthritis treatment. Biomed Pharmacother 2016; 80:30-41. [PMID: 27133037 DOI: 10.1016/j.biopha.2016.03.004] [Citation(s) in RCA: 114] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2016] [Revised: 03/03/2016] [Accepted: 03/03/2016] [Indexed: 02/06/2023] Open
Abstract
Rheumatoid arthritis (RA) is one of the common and severe autoimmune diseases related to joints. This chronic autoimmune inflammatory disease, leads to functional limitation and reduced quality of life, since as there is bone and cartilage destruction, joint swelling and pain. Current advances and new treatment approaches have considerably postponed disease progression and improved the quality of life for many patients. In spite of major advances in therapeutic options, restrictions on the routes of administration and the necessity for frequent and long-term dosing often result in systemic adverse effects and patient non-compliance. Unlike usual drugs, nanoparticle systems are planned to deliver therapeutic agents especially to inflamed synovium, so avoiding systemic and unpleasant effects. The present review discusses about some of the most successful drugs in RA therapy and their side effects and also focuses on key design parameters of RA-targeted nanotechnology-based strategies for improving RA therapies.
Collapse
|
27
|
Abstract
Plasmids are currently an indispensable molecular tool in life science research and a central asset for the modern biotechnology industry, supporting its mission to produce pharmaceutical proteins, antibodies, vaccines, industrial enzymes, and molecular diagnostics, to name a few key products. Furthermore, plasmids have gradually stepped up in the past 20 years as useful biopharmaceuticals in the context of gene therapy and DNA vaccination interventions. This review provides a concise coverage of the scientific progress that has been made since the emergence of what are called today plasmid biopharmaceuticals. The most relevant topics are discussed to provide researchers with an updated overview of the field. A brief outline of the initial breakthroughs and innovations is followed by a discussion of the motivation behind the medical uses of plasmids in the context of therapeutic and prophylactic interventions. The molecular characteristics and rationale underlying the design of plasmid vectors as gene transfer agents are described and a description of the most important methods used to deliver plasmid biopharmaceuticals in vivo (gene gun, electroporation, cationic lipids and polymers, and micro- and nanoparticles) is provided. The major safety issues (integration and autoimmunity) surrounding the use of plasmid biopharmaceuticals is discussed next. Aspects related to the large-scale manufacturing are also covered, and reference is made to the plasmid products that have received marketing authorization as of today.
Collapse
|
28
|
Talat Z, Tursun X, Cheng L, Mijiti A, Aisa HA. Anti-Arthritic and Antiinflammatory Effects of the Traditional Uighur Formula Kursi Caper In Vivo. Phytother Res 2015; 29:1901-9. [PMID: 26434647 DOI: 10.1002/ptr.5479] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2015] [Revised: 08/18/2015] [Accepted: 08/29/2015] [Indexed: 11/11/2022]
Abstract
Kursi Caper (KC) is a Uighur medicine based on caper which is widely used to treat arthritis and rheumatism, and preliminary studies in our laboratory showed that this traditional formula may possess potent antiinflammatory effects. This study confirms the antiinflammatory effect of KC in the adjuvant induced arthritis (AIA) model, the carrageenan and cotton-pellet induced granuloma rat models, and further investigates in vivo the mechanism of action by measuring relevant indicators of anti-arthritic activity. KC showed significant and dose-dependent anti-arthritic and antiinflammatory effects, demonstrated by reduced paw edema and arthritic scores in all animal models. Histopathological examination showed that KC reduced levels of synovial inflammatory factors in AIA rats. The overproduction of TNF-α and IL-1β was attenuated, and CAT, MDA and SOD levels were restored to normal in KC-treated rats. KC also significantly reduced LPS-induced proliferation of B lymphocytes and ConA induced proliferation of T lymphocytes in a dose-dependent manner. Flow cytometry showed that the high dose KC-treated group had a significantly decreased frequency of Th17 cells. This study indicates that KC can significantly attenuate arthritis and inflammation in rats by decreasing the levels of inflammatory cytokines, regulating oxidative stress, reducing lymphocyte proliferation and decreasing Th17. This supports the traditional use of KC as a potential modern therapeutic agent for the treatment of arthritis and related conditions.
Collapse
Affiliation(s)
- Zulfiye Talat
- The Key Laboratory of Plant Resources and Chemistry of Arid Zone, Chinese Academy of Sciences, Urumqi, China.,State Key Laboratory Basis of Xinjiang Indigenous Medicinal Plants Resource Utilization, Xinjiang Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Urumqi, 830011, China
| | - Xirali Tursun
- The Key Laboratory of Plant Resources and Chemistry of Arid Zone, Chinese Academy of Sciences, Urumqi, China.,State Key Laboratory Basis of Xinjiang Indigenous Medicinal Plants Resource Utilization, Xinjiang Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Urumqi, 830011, China
| | - Lufeng Cheng
- Xinjiang Medical University, Urumqi, 830011, China
| | | | - Haji Akber Aisa
- The Key Laboratory of Plant Resources and Chemistry of Arid Zone, Chinese Academy of Sciences, Urumqi, China.,State Key Laboratory Basis of Xinjiang Indigenous Medicinal Plants Resource Utilization, Xinjiang Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Urumqi, 830011, China
| |
Collapse
|
29
|
Prasad LK, O’Mary H, Cui Z. Nanomedicine delivers promising treatments for rheumatoid arthritis. Nanomedicine (Lond) 2015; 10:2063-74. [PMID: 26084368 PMCID: PMC4552357 DOI: 10.2217/nnm.15.45] [Citation(s) in RCA: 86] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
An increased understanding in the pathophysiology of chronic inflammatory diseases, such as rheumatoid arthritis, reveals that the diseased tissue and the increased presence of macrophages and other overexpressed molecules within the tissue can be exploited to enhance the delivery of nanomedicine. Nanomedicine can passively accumulate into chronic inflammatory tissues via the enhanced permeability and retention phenomenon, or be surface conjugated with a ligand to actively bind to receptors overexpressed by cells within chronic inflammatory tissues, leading to increased efficacy and reduced systemic side-effects. This review highlights the research conducted over the past decade on using nanomedicine for potential treatment of rheumatoid arthritis and summarizes some of the major findings and promising opportunities on using nanomedicine to treat this prevalent and chronic disease.
Collapse
Affiliation(s)
- Leena Kumari Prasad
- Pharmaceutics Division, College of Pharmacy, The University of Texas at Austin, Austin, TX 78712, USA
| | - Hannah O’Mary
- Pharmaceutics Division, College of Pharmacy, The University of Texas at Austin, Austin, TX 78712, USA
| | - Zhengrong Cui
- Pharmaceutics Division, College of Pharmacy, The University of Texas at Austin, Austin, TX 78712, USA
- Inner Mongolia Key Laboratory of Molecular Biology, Inner Mongolia Medical University, Hohhot, Inner Mongolia, China
| |
Collapse
|
30
|
Chen X, Zhao Y, Geng S, Miron RJ, Zhang Q, Wu C, Zhang Y. In vivo experimental study on bone regeneration in critical bone defects using PIB nanogels/boron-containing mesoporous bioactive glass composite scaffold. Int J Nanomedicine 2015; 10:839-46. [PMID: 25653525 PMCID: PMC4309792 DOI: 10.2147/ijn.s69001] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023] Open
Abstract
PURPOSE In the present study, the fabrication of novel p(N-isopropylacrylamide-co-butyl methylacrylate) (PIB) nanogels was combined with boron-containing mesoporous bioactive glass (B-MBG) scaffolds in order to improve the mechanical properties of PIB nanogels alone. Scaffolds were tested for mechanical strength and the ability to promote new bone formation in vivo. PATIENTS AND METHODS To evaluate the potential of each scaffold in bone regeneration, ovariectomized rats were chosen as a study model to determine the ability of PIB nanogels to stimulate bone formation in a complicated anatomical bone defect. PIB nanogels and PIB nanogels/B-MBG composites were respectively implanted into ovariectomized rats with critical-sized femur defects following treatment periods of 2, 4, and 8 weeks post-implantation. RESULTS Results from the present study demonstrate that PIB nanogels/B-MBG composites showed greater improvement in mechanical strength when compared to PIB nanogels alone. In vivo, hematoxylin and eosin staining revealed significantly more newly formed bone in defects containing PIB nanogels/B-MBG composite scaffolds when compared to PIB nanogels alone. Tartrate-resistant acid phosphatase-positive staining demonstrated that both scaffolds were degraded over time and bone remodeling occurred in the surrounding bone defect as early as 4 weeks post-implantation. CONCLUSION The results from the present study indicate that PIB nanogels are a potential bone tissue engineering biomaterial able to treat defects of irregular shapes and deformities as an injectable, thermoresponsive, biocompatible hydrogel which undergoes rapid thermal gelation once body temperature is reached. Furthermore, its combination with B-MBG scaffolds improves the mechanical properties and ability to promote new bone formation when compared to PIB nanogels alone.
Collapse
Affiliation(s)
- Xiaohui Chen
- 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, People's Republic of China ; Department of Dental Implantology, School and Hospital of Stomatology, Wuhan University, People's Republic of China
| | - Yanbing Zhao
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Shinan Geng
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Richard J Miron
- 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, People's Republic of China
| | - Qiao Zhang
- 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, People's Republic of China
| | - Chengtie Wu
- State Key Laboratory of High Performance Ceramics and Superfine Microstructure, Shanghai Institute of Ceramics, Chinese Academy of Sciences, Shanghai, People's Republic of China
| | - Yufeng Zhang
- 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, People's Republic of China ; Department of Dental Implantology, School and Hospital of Stomatology, Wuhan University, People's Republic of China
| |
Collapse
|
31
|
Tran TH, Amiji MM. Targeted delivery systems for biological therapies of inflammatory diseases. Expert Opin Drug Deliv 2014; 12:393-414. [DOI: 10.1517/17425247.2015.972931] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
|
32
|
Chen H, Chen Y, Yang H, Xu W, Zhang M, Ma Y, Achilefu S, Gu Y. A dual-targeting nanocarrier based on modified chitosan micelles for tumor imaging and therapy. Polym Chem 2014. [DOI: 10.1039/c4py00495g] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
|
33
|
Singh A, Talekar M, Raikar A, Amiji M. Macrophage-targeted delivery systems for nucleic acid therapy of inflammatory diseases. J Control Release 2014; 190:515-30. [PMID: 24747762 DOI: 10.1016/j.jconrel.2014.04.021] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2014] [Accepted: 03/21/2014] [Indexed: 01/10/2023]
Abstract
Inflammation is an immune response that marks several pathophysiological conditions in our body. Though adaptive immune cells play a major role in the progression of the disease, components of innate immune system, mainly monocytes and macrophages play the central role in onset of inflammation. Tissue-associated macrophages are widely distributed in the body showing tremendous anatomical and functional diversity and are actively involved in maintaining the homeostasis. They exhibit different phenotypes depending on their residing tissue microenvironment and the two major functional phenotypes are classically activated M1 phenotype showing pro-inflammatory characteristics and alternatively activated M2 phenotype demonstrating anti-inflammatory nature. Several cytokines, chemokines and other regulatory mediators delicately govern the balance of the two phenotypes in a tissue. This balance, however, is subverted during infection, injury or autoimmune response leading to increased population of M1 phenotype and subsequent chronic inflammatory disease states. This review underlines the role of macrophages in inflammatory diseases with an insight into potential molecular targets for nucleic acid therapy. Finally, some recent nanotechnology-based approaches to devise macrophage-specific targeted therapy have been highlighted.
Collapse
Affiliation(s)
- Amit Singh
- Department of Pharmaceutical Sciences, School of Pharmacy, Northeastern University, Boston 02115, USA
| | - Meghna Talekar
- Department of Pharmaceutical Sciences, School of Pharmacy, Northeastern University, Boston 02115, USA
| | - Ankita Raikar
- Department of Pharmaceutical Sciences, School of Pharmacy, Northeastern University, Boston 02115, USA
| | - Mansoor Amiji
- Department of Pharmaceutical Sciences, School of Pharmacy, Northeastern University, Boston 02115, USA.
| |
Collapse
|
34
|
Cheng N, Wang Y, Zhang Y, Shi B. The osteogenic potential of mesoporous bioglasses/silk and non-mesoporous bioglasses/silk scaffolds in ovariectomized rats: in vitro and in vivo evaluation. PLoS One 2013; 8:e81014. [PMID: 24265840 PMCID: PMC3827187 DOI: 10.1371/journal.pone.0081014] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2013] [Accepted: 10/14/2013] [Indexed: 02/02/2023] Open
Abstract
Silk-based scaffolds have been introduced to bone tissue regeneration for years, however, their local therapeutic efficency in bone metabolic disease condition has been seldom reported. According to our previous report, mesoporous bioactive glass (MBG)/silk scaffolds exhibits superior in vitro bioactivity and in vivo osteogenic properties compared to non-mesoporous bioactive glass (BG)/silk scaffolds, but no information could be found about their efficiency in osteoporotic (OVX) environment. This study investigated a biomaterial-based approach for improving MSCs behavior in vitro, and accelerating OVX defect healing by using 3D BG/silk and MBG/silk scaffolds, and pure silk scaffolds as control. The results of SEM, CCK-8 assay and quantitative ALP activity showed that MBG/silk scaffolds can improve attachment, proliferation and osteogenic differentiation of both O-MSCs and sham control. In vivo therapeutic efficiency was evaluated by μCT analysis, hematoxylin and eosin staining, safranin O staining and tartrate-resistant acid phosphatase, indicating accelerated bone formation with compatible scaffold degradation and reduced osteoclastic response of defect healing in OVX rats after 2 and 4 weeks treatment, with a rank order of MBG/silk > BG/silk > silk group. Immunohistochemical markers of COL I, OPN, BSP and OCN also revealed that MBG/silk scaffolds can better induce accelerated collagen and non-collagen matrix production. The findings of this study suggest that MBG/silk scaffolds provide a better environment for cell attachment, proliferation and differentiation, and act as potential substitute for treating local osteoporotic defects.
Collapse
Affiliation(s)
- Ning Cheng
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School & Hospital of Stomatology, Wuhan University, Wuhan, People’s Republic of China.
| | - Yuanqin Wang
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School & Hospital of Stomatology, Wuhan University, Wuhan, People’s Republic of China.
| | - Yufeng Zhang
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School & Hospital of Stomatology, Wuhan University, Wuhan, People’s Republic of China.
| | - Bin Shi
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School & Hospital of Stomatology, Wuhan University, Wuhan, People’s Republic of China.
- * E-mail:
| |
Collapse
|
35
|
Abstract
Cancer is one of the major causes of mortality worldwide and advanced techniques for therapy are urgently needed. The development of novel nanomaterials and nanocarriers has allowed a major drive to improve drug delivery in cancer. The major aim of most nanocarrier applications has been to protect the drug from rapid degradation after systemic delivery and allowing it to reach tumor site at therapeutic concentrations, meanwhile avoiding drug delivery to normal sites as much as possible to reduce adverse effects. These nanocarriers are formulated to deliver drugs either by passive targeting, taking advantage of leaky tumor vasculature or by active targeting using ligands that increase tumoral uptake potentially resulting in enhanced antitumor efficacy, thus achieving a net improvement in therapeutic index. The rational design of nanoparticles plays a critical role since structural and physical characteristics, such as size, charge, shape, and surface characteristics determine the biodistribution, pharmacokinetics, internalization and safety of the drugs. In this review, we focus on several novel and improved strategies in nanocarrier design for cancer therapy.
Collapse
|
36
|
Cheng N, Dai J, Cheng X, Li S, Miron RJ, Wu T, Chen W, Zhang Y, Shi B. Porous CaP/silk composite scaffolds to repair femur defects in an osteoporotic model. JOURNAL OF MATERIALS SCIENCE. MATERIALS IN MEDICINE 2013; 24:1963-1975. [PMID: 23674058 PMCID: PMC5995474 DOI: 10.1007/s10856-013-4945-y] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/12/2012] [Accepted: 04/29/2013] [Indexed: 06/02/2023]
Abstract
The most common complication for patients with postmenopausal osteoporosis is bone-related defects and fractures. While routine medication has a high probability of undesirable side effects, new approaches have aimed to develop regeneration procedures that stimulate new bone formation while reversing bone loss. Recently, we have synthesized a new hybrid CaP/silk scaffold with a CaP-phase distribution and pore architecture better suited to facilitate cell differentiation and bone formation. The aim of the present study was to compare the involved remodeling process and therapeutic effect of porous CaP/silk composite scaffolds upon local implantation into osteoporotic defects. Wistar rats were used to induce postmenopausal osteoporotic model by bilateral ovariectomy. The pure silk and hybrid CaP/silk scaffolds were implanted into critical sized defects created in distal femoral epiphysis. After 14 and 28 days, the in vivo osteogenetic efficiency was evaluated by μCT analysis, hematoxylin and eosin staining, Safranin O staining, tartrate-resistant acid phosphatase staining, and immunohistochemical assessment. Animals with or without critical-sized defects were used as drill or blank controls, respectively. The osteoporotic defect model was well established with significantly decreased μCT parameters of BV/TV, Tb.N and increased Tb.Sp, porosity, combined with changes in histological observations. During the healing process, the critical-sized drill control defects failed to regenerate appreciable bone tissue, while more significantly increased bone formation and mineralization with dynamic scaffold degradation and decreased osteoclastic bone resorption could be detected within defects with hybrid CaP/silk scaffolds compared to pure silk scaffolds.
Collapse
Affiliation(s)
- Ning Cheng
- State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School & Hospital of Stomatology, Wuhan University, 237 Luoyu Road, Wuhan, 430079, People's Republic of China
| | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Shi Q, Abusarah J, Baroudi G, Fernandes JC, Fahmi H, Benderdour M. Response to 'Ramipril attenuates lipid peroxidation and cardiac fibrosis in an experimental model of rheumatoid arthritis' - authors' reply. Arthritis Res Ther 2013; 15:406. [PMID: 23566352 PMCID: PMC3672737 DOI: 10.1186/ar4196] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
|
38
|
de Paula Pansani Oliveira F, Dalla Picola IP, Shi Q, Barbosa HFG, Tiera VADO, Fernandes JC, Tiera MJ. Synthesis and evaluation of diethylethylamine-chitosan for gene delivery: composition effects on the in vitro transfection efficiency. NANOTECHNOLOGY 2013; 24:055101. [PMID: 23306549 DOI: 10.1088/0957-4484/24/5/055101] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/01/2023]
Abstract
Chitosan has been indicated as a safe and promising polycation vector for gene delivery. However its low transfection efficiency has been a challenging obstacle for its application. To address this limitation, we synthesized chitosan derivatives which had increasing amounts of diethylethylamine groups (DEAE) attached to the chitosan main chain. The plasmid DNA VR1412 (pDNA), encoding the ß-galactosidase (ß-gal) reporter gene was used to prepare nanoparticles with the chitosan derivatives, and the transfection studies were performed with HeLa cells. By means of dynamic light scattering and zeta potential measurements, it was shown that diethylethylamine-chitosan derivatives (DEAE(x)-CH) were able to condense DNA into small particles having a surface charge depending on the polymer/DNA ratio (N/P ratio). Nanoparticles prepared with derivatives containing 15 and 25% of DEAE groups (DEAE(15)-CH and DEAE(25)-CH) exhibited transfection efficiencies ten times higher than that observed with deacetylated chitosan (CH). For derivatives with higher degrees of substitution (DS), transfection efficiency decreased. The most effective carriers showed low cytotoxicity and good transfection activities at low charge ratios (N/P). Vectors with low DS were easily degraded in the presence of lysozyme at physiological conditions in vitro and the nontoxicity displayed by these vectors opens up new opportunities in the design of DEAE-chitosan-based nanoparticles for gene delivery.
Collapse
|
39
|
Nicolas J, Mura S, Brambilla D, Mackiewicz N, Couvreur P. Design, functionalization strategies and biomedical applications of targeted biodegradable/biocompatible polymer-based nanocarriers for drug delivery. Chem Soc Rev 2013; 42:1147-235. [DOI: 10.1039/c2cs35265f] [Citation(s) in RCA: 977] [Impact Index Per Article: 88.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
|
40
|
Fernandes JC, Qiu X, Winnik FM, Benderdour M, Zhang X, Dai K, Shi Q. Low molecular weight chitosan conjugated with folate for siRNA delivery in vitro: optimization studies. Int J Nanomedicine 2012; 7:5833-45. [PMID: 23209368 PMCID: PMC3511191 DOI: 10.2147/ijn.s35567] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2012] [Indexed: 12/04/2022] Open
Abstract
The low transfection efficiency of chitosan is one of its drawbacks as a gene delivery carrier. Low molecular weight chitosan may help to form small-sized polymer-DNA or small interfering RNA (siRNA) complexes. Folate conjugation may improve gene transfection efficiency because of the promoted uptake of folate receptor-bearing cells. In the present study, chitosan was conjugated with folate and investigated for its efficacy as a delivery vector for siRNA in vitro. We demonstrate that the molecular weight of chitosan has a major influence on its biological and physicochemical properties, and very low molecular weight chitosan (below 10 kDa) has difficulty in forming stable complexes with siRNA. In this study, chitosan 25 kDa and 50 kDa completely absorbed siRNA and formed nanoparticles (≤220 nm) at a chitosan to siRNA weight ratio of 50:1. The introduction of a folate ligand onto chitosan decreased nanoparticle toxicity. Compared with chitosan-siRNA, folate-chitosan-siRNA nanoparticles improved gene silencing transfection efficiency. Therefore, folate-chitosan shows potential as a viable candidate vector for safe and efficient siRNA delivery.
Collapse
Affiliation(s)
- Julio C Fernandes
- Orthopaedics Research Laboratory, Research Centre, Sacré-Coeur Hospital, Montreal, Quebec, Canada
| | | | | | | | | | | | | |
Collapse
|
41
|
Shi Q, Abusarah J, Baroudi G, Fernandes JC, Fahmi H, Benderdour M. Ramipril attenuates lipid peroxidation and cardiac fibrosis in an experimental model of rheumatoid arthritis. Arthritis Res Ther 2012; 14:R223. [PMID: 23079082 PMCID: PMC3580534 DOI: 10.1186/ar4062] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2011] [Accepted: 08/31/2012] [Indexed: 12/13/2022] Open
Abstract
INTRODUCTION Recent studies revealed that co-morbidity and mortality due to cardiovascular disease are increased in patients with rheumatoid arthritis (RA) but little is known about factors involved in these manifestations. This study aimed at characterizing the impact of arthritis on oxidative stress status and tissue fibrosis in the heart of rats with adjuvant-induced arthritis (AIA). METHODS AIA was induced with complete Freund's adjuvant in female Lewis rats. Animals were treated by oral administration of vehicle or angiotensin-converting enzyme inhibitor ramipril (10 mg/kg/day) for 28 days, beginning 1 day after arthritis induction. Isolated adult cardiomyocytes were exposed to 10 μM 4-hydroxynonenal (HNE) for 24 hours in the presence or absence of 10 μM ramipril. RESULTS Compared to controls, AIA rats showed significant 55 and 30% increase of 4-HNE/protein adducts in serum and left ventricular (LV) tissues, respectively. Cardiac mitochondrial NADP+-isocitrate dehydrogenase (mNADP-ICDH) activity decreased by 25% in AIA rats without any changes in its protein and mRNA expression. The loss of mNADP-ICDH activity was correlated with enhanced accumulation of HNE/mNADP-ICDH adducts as well as with decrease of glutathione and NADPH. Angiotensin II type 1 receptor (AT1R) expression and tissue fibrosis were induced in LV tissues from AIA rats. In isolated cardiomyocytes, HNE significantly decreased mNADP-ICDH activity and enhanced type I collagen and connective tissue growth factor expression. The oral administration of ramipril significantly reduced HNE and AT1R levels and restored mNADP-ICDH activity and redox status in LV tissues of AIA rats. The protective effects of this drug were also evident from the decrease in arthritis scoring and inflammatory markers. CONCLUSION Collectively, our findings disclosed that AIA induced oxidative stress and fibrosis in the heart. The fact that ramipril attenuates inflammation, oxidative stress and tissue fibrosis may provide a novel strategy to prevent heart diseases in RA.
Collapse
|
42
|
Tautzenberger A, Kovtun A, Ignatius A. Nanoparticles and their potential for application in bone. Int J Nanomedicine 2012; 7:4545-57. [PMID: 22923992 PMCID: PMC3423651 DOI: 10.2147/ijn.s34127] [Citation(s) in RCA: 117] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Biomaterials are commonly applied in regenerative therapy and tissue engineering in bone, and have been substantially refined in recent years. Thereby, research approaches focus more and more on nanoparticles, which have great potential for a variety of applications. Generally, nanoparticles interact distinctively with bone cells and tissue, depending on their composition, size, and shape. Therefore, detailed analyses of nanoparticle effects on cellular functions have been performed to select the most suitable candidates for supporting bone regeneration. This review will highlight potential nanoparticle applications in bone, focusing on cell labeling as well as drug and gene delivery. Labeling, eg, of mesenchymal stem cells, which display exceptional regenerative potential, makes monitoring and evaluation of cell therapy approaches possible. By including bioactive molecules in nanoparticles, locally and temporally controlled support of tissue regeneration is feasible, eg, to directly influence osteoblast differentiation or excessive osteoclast behavior. In addition, the delivery of genetic material with nanoparticulate carriers offers the possibility of overcoming certain disadvantages of standard protein delivery approaches, such as aggregation in the bloodstream during systemic therapy. Moreover, nanoparticles are already clinically applied in cancer treatment. Thus, corresponding efforts could lead to new therapeutic strategies to improve bone regeneration or to treat bone disorders.
Collapse
Affiliation(s)
- Andrea Tautzenberger
- Institute of Orthopedic Research and Biomechanics, Centre of Musculoskeletal Research, Ulm University, Ulm, Germany.
| | | | | |
Collapse
|
43
|
Zhang Y, Cheng N, Miron R, Shi B, Cheng X. Delivery of PDGF-B and BMP-7 by mesoporous bioglass/silk fibrin scaffolds for the repair of osteoporotic defects. Biomaterials 2012; 33:6698-708. [PMID: 22763224 DOI: 10.1016/j.biomaterials.2012.06.021] [Citation(s) in RCA: 135] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2012] [Accepted: 06/15/2012] [Indexed: 01/23/2023]
Abstract
Osteoporosis is a chronic disease affecting millions of people worldwide caused by an imbalance between bone-forming osteoblasts and bone-resorbing osteoclasts. Despite recent developments in pharmacological agents to prevent osteoporotic-related fractures, much less attention has been placed on the repair of bone defects following fracture. Critical to this process is the recruitment of mesenchymal stem cells (MSCs) to defect sites by growth factors. One method which has been effective for the sustained release of growth factors is that of gene therapy. The aim of the present study was to investigate newly developed mesoporous bioglass/silk fibrin scaffolds containing adPDGF-b and adBMP-7 into osteoporotic critical-sized femur defects in ovariectomised rats following treatment periods of 2 and 4 weeks. In vivo osteogenetic efficiency evaluated by μ-CT analysis, hematoxylin and eosin staining, and immunohistochemical (type I collagen, osteopontin and BSP) revealed significantly new bone formation in defects containing adenovirus for both PDGF-b and BMP-7 when compared to scaffolds alone and scaffolds containing BMP-7. TRAP-positive staining also demonstrated the ability for these scaffolds to be degraded over time and initiate bone turnover/remodeling. Although the use of gene therapy for clinical applications is still in its infancy, results from the present study demonstrate their potent ability to recruit mesenchymal progenitor cells through sustained release of PDGF-b and BMP-7 which may be beneficial for patients suffering from osteoporotic-related fractures.
Collapse
Affiliation(s)
- Yufeng Zhang
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School & Hospital of Stomatology, Wuhan University, 237 Luoyu Road, Wuhan 430079, PR China.
| | | | | | | | | |
Collapse
|
44
|
Nagai T, Kyo A, Hasui K, Takao S, Matsuyama T. Efficacy of an immunotoxin to folate receptor beta in the intra-articular treatment of antigen-induced arthritis. Arthritis Res Ther 2012; 14:R106. [PMID: 22551402 PMCID: PMC3446483 DOI: 10.1186/ar3831] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2011] [Revised: 04/02/2012] [Accepted: 05/02/2012] [Indexed: 12/13/2022] Open
Abstract
Introduction We previously demonstrated that synovial sublining macrophages express folate receptor beta (FRβ). The aim of this study was to evaluate the efficacy of intra-articular administration of a recombinant immunotoxin to FRβ for treating rat antigen-induced arthritis. Methods A monoclonal antibody (mAb) to rat FRβ was produced by immunizing mice with B300-19 cells (murine pre-B cells) transfected with the rat FRβ gene. Recombinant immunotoxin was prepared by conjugating the Fv portion of the anti-rat FRβ mAb heavy chain with a truncated Pseudomonas exotoxin A and the Fv portion of the anti-rat FRβ mAb light chain. Antigen-induced arthritis was induced through intra-articular injection of methylated bovine serum albumin (mBSA) after two subcutaneous injections of mBSA and complete Freund's adjuvant. Immunotoxin was intra-articularly injected into the arthritis joint every other day for seven days after arthritis onset. Joint swelling was measured and histological scores of inflammation, synovial thickness, cartilage, and bone destruction were determined. Immunohistochemistry was performed to detect osteoclast and osteoclast precursor FRβ-expressing macrophages and cathepsin K-positive cells on day 21. Results Intra-articular administration of the immunotoxin attenuated joint swelling (61% suppression; P < 0.01 compared to the control on day 21) and improved histological findings, particularly cartilage and bone destruction (scores of rats treated with control versus the immunotoxin: 2.2 versus 0.5; P < 0.01), by reducing the number of FRβ-expressing macrophages and cathepsin K-positive cells. Conclusions Intra-articular administration of an immunotoxin to FRβ is effective for improving rat antigen-induced arthritis.
Collapse
Affiliation(s)
- Taku Nagai
- Department of Immunology, Graduate school of Medical and Dental Sciences, Kagoshima University, Kagoshima 890-8544, Japan
| | | | | | | | | |
Collapse
|
45
|
Jain S, Amiji M. Tuftsin-modified alginate nanoparticles as a noncondensing macrophage-targeted DNA delivery system. Biomacromolecules 2012; 13:1074-85. [PMID: 22385328 DOI: 10.1021/bm2017993] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
The main objective of this study was to evaluate macrophage-targeted alginate nanoparticles as a noncondensing gene delivery system for potential anti-inflammatory therapy. An external gelation method was employed to form plasmid DNA-encapsulated alginate nanoparticles. The nanoparticle surface was modified with a peptide sequence containing tuftsin (TKPR), and transfection efficiency was determined in J774A.1 macrophages. The effect of transfected mIL-10 in blocking expression of tumor necrosis factor-alpha (TNF-α) was evaluated in lipopolysaccharide (LPS)-stimulated cells. Scrambled peptide- and tuftsin-modified cross-linked alginate nanoparticles efficiently encapsulated plasmid DNA and protected against DNase I degradation. The transgene expression efficiencies, measured using GFP and mIL-10 expressing plasmid DNA, were highest with tuftsin-modified nanoparticles. Levels of TNF-α were significantly lower (p < 0.0001) in LPS-stimulated cells that were transfected with mIL-10 using alginate nanoparticles. The results of the study show that noncondensing alginate nanoparticles can efficiently deliver plasmid DNA, leading to sustained in vitro gene expression in macrophages.
Collapse
Affiliation(s)
- Shardool Jain
- Department of Pharmaceutical Sciences, School of Pharmacy, Northeastern University, Boston, Massachusetts 02115, United States
| | | |
Collapse
|
46
|
Jreyssaty C, Shi Q, Wang H, Qiu X, Winnik FM, Zhang X, Dai K, Benderdour M, Fernandes JC. Efficient Nonviral Gene Therapy Using Folate-Targeted Chitosan-DNA Nanoparticles In Vitro. ISRN PHARMACEUTICS 2012; 2012:369270. [PMID: 22474605 PMCID: PMC3317051 DOI: 10.5402/2012/369270] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/15/2011] [Accepted: 12/12/2011] [Indexed: 11/30/2022]
Abstract
Nonviral cationic polymers like chitosan can be combined with DNA to protect it from degradation. The chitosan is a biocompatible, biodegradable, nontoxic, and cheap polycationic polymer with low immunogenicity. The objective of this study was to synthesize and then assess different chitosan-DNA nanoparticles and to select the best ones for selective in vitro transfection in human epidermoid carcinoma (KB) cell lines. It revealed that different combinations of molecular weight, the presence or absence of folic acid ligand, and different plasmid DNA sizes can lead to nanoparticles with various diameters and diverse transfection efficiencies. The intracellular trafficking, nuclear uptake, and localization are also studied by confocal microscopy, which confirmed that DNA was delivered to cell nuclei to be expressed.
Collapse
Affiliation(s)
- Christian Jreyssaty
- Orthopaedics Research Laboratory, Research Center, Sacré-Coeur Hospital, University of Montreal, 5400 West Gouin Boulevard, Montreal, QC, Canada H4J 1C5
| | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Chitosan-Grafted Copolymers and Chitosan-Ligand Conjugates as Matrices for Pulmonary Drug Delivery. ACTA ACUST UNITED AC 2011. [DOI: 10.1155/2011/865704] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Recently, much attention has been given to pulmonary drug delivery by means of nanosized systems to treat both local and systemic diseases. Among the different materials used for the production of nanocarriers, chitosan enjoys high popularity due to its inherent characteristics such as biocompatibility, biodegradability, and mucoadhesion, among others. Through the modification of chitosan chemical structure, either by the addition of new chemical groups or by the functionalization with ligands, it is possible to obtain derivatives with advantageous and specific characteristics for pulmonary administration. In this paper, we discuss the advantages of using chitosan for nanotechnology-based pulmonary delivery of drugs and summarize the most recent and promising modifications performed to the chitosan molecule in order to improve its characteristics.
Collapse
|
48
|
Sukedai M, Ariyoshi W, Okinaga T, Iwanaga K, Habu M, Yoshioka I, Tominaga K, Nishihara T. Inhibition of adjuvant arthritis in rats by electroporation with interleukin-1 receptor antagonist. J Interferon Cytokine Res 2011; 31:839-46. [PMID: 21859345 DOI: 10.1089/jir.2011.0024] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
To assess the protective effects of the cytokine inhibitor interleukin-1 receptor antagonist (IL-1ra) on gene induction, an electroporation technique to treat adjuvant-induced arthritis (AIA) in rats was established, and its advantage was estimated in the present study. Electroporation with human IL-1ra was performed in Lewis rats before and after induction of AIA. Local inflammation was evaluated by monitoring hind paw swelling, whereas histological evaluations were performed using paraffin embedded sections of hind paw specimens stained with hematoxylin and eosin. In addition, serum IL-1? levels were analyzed using an enzyme-linked immunosorbent assay. Induction of IL-1ra by our electroporation method inhibited systematic body weight loss and enhancement of local inflammation after intradermal injection of heat-killed Mycobacterium tuberculosis. Notably, IL-1ra electroporation reduced paw swelling, inflammation, and bone erosion scores in embedded sections and serum IL-1? levels induced in AIA rats. The IL-1ra gene induction using the present electroporation technique inhibited local and systematic inflammation in AIA rats. These results indicate that this method may represent a novel pharmacotherapy strategy for arthritis.
Collapse
Affiliation(s)
- Miho Sukedai
- Division of Infections and Molecular Biology, Department of Health Promotion, Kyushu Dental College, Kitakyushu, Japan
| | | | | | | | | | | | | | | |
Collapse
|
49
|
Pham CTN. Nanotherapeutic approaches for the treatment of rheumatoid arthritis. WILEY INTERDISCIPLINARY REVIEWS-NANOMEDICINE AND NANOBIOTECHNOLOGY 2011; 3:607-19. [PMID: 21837725 DOI: 10.1002/wnan.157] [Citation(s) in RCA: 66] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Rheumatoid arthritis (RA) is a common inflammatory disease characterized by progressive bone and cartilage destruction, resulting in severe functional limitations, shortened lifespan, and increased mortality rates. Recent advances and new treatment approaches have significantly delayed disease progression and improved the quality of life for many patients. Yet few patients attain or can be maintained in disease remission without continuous immunosuppressive therapy. In addition, a sizable portion of patients also fails to respond or eventually develops tolerance to current therapies. Thus there is a continued need for the development of new therapeutic strategies for the treatment of RA. Unlike conventional drugs, nanosystems are designed to deliver therapeutic agents specifically to the site of inflammation, therefore avoiding potential systemic and off-target unwanted effects. They allow investigators to consider or reconsider therapeutic agents that were previously deemed too toxic to deliver through a systemic route. This article reviews recent nanotechnology-based strategies that are being developed for the treatment of inflammatory arthritis.
Collapse
Affiliation(s)
- Christine T N Pham
- Division of Rheumatology, Washington University School of Medicine, Saint Louis, MO, USA.
| |
Collapse
|
50
|
Duceppe N, Tabrizian M. Advances in using chitosan-based nanoparticles for in vitro and in vivo drug and gene delivery. Expert Opin Drug Deliv 2011; 7:1191-207. [PMID: 20836623 DOI: 10.1517/17425247.2010.514604] [Citation(s) in RCA: 155] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
IMPORTANCE OF THE FIELD This review aims to provide an overview of state-of-the-art chitosan-based nanosized carriers for the delivery of therapeutic agents. Chitosan nanocarriers are smart delivery systems owing to the possibility of their property alterations with various approaches, which would confer them with the possibility of spatiotemporal delivery features. AREAS COVERED IN THIS REVIEW The focus of this review is principally on those aspects that have not often been addressed in other reviews. These include the influence of physicochemical properties of chitosan on delivery mechanisms and chitosan modification with a variety of ligand moieties specific for cell surface receptors to increase recognition and uptake of nanocarriers into cells through receptor-mediated endocytosis. Multiple examples that demonstrate the advantages of chitosan-based nanocarriers over other delivery systems of therapeutic agents are highlighted. Particular emphasis is given to the alteration of material properties by functionalization or combination with other polymers for their specific applications. Finally, structural and experimental parameters influencing transfection efficiency of chitosan-based nanocarriers are presented for both in vitro and in vivo gene delivery. WHAT THE READER WILL GAIN The readers will acquire knowledge of parameters influencing the properties of the chitosan-based nanocarriers for delivery of therapeutic agents (genetic material or drugs) in vitro and in vivo. They will get a better idea of the strategies to be adapted to tune the characteristics of chitosan and chitosan derivatives for specific delivery applications. TAKE HOME MESSAGE Chitosan is prone to chemical and physical modifications, and is very responsive to environmental stimuli such as temperature and pH. These features make chitosan a smart material with great potential for developing multifunctional nanocarrier systems to deliver large varieties of therapeutic agents administrated in multiple ways with reduced side effects.
Collapse
Affiliation(s)
- Nicolas Duceppe
- Department of Biomedical Engineering, Faculty of Medicine, McGill University, Duff Medical Science Building, 3775 University Street, Montréal, Québec, Canada
| | | |
Collapse
|