1
|
Ghasemiyeh P, Mohammadi-Samani S. siRNA-based delivery systems: Technologies, carriers, applications, and approved products. Eur J Pharmacol 2025; 996:177441. [PMID: 40023357 DOI: 10.1016/j.ejphar.2025.177441] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2024] [Revised: 02/22/2025] [Accepted: 02/26/2025] [Indexed: 03/04/2025]
Abstract
Ribonucleic acid (RNA) therapeutics are a novel category of therapeutic agents that use different types of RNAs to regulate genes and modulate protein synthesis to treat a wide range of diseases. The main advantages of RNA therapeutics over conventional small molecule drugs would be the potential to target undruggable sites, ease of production and faster development process, and longer duration of action. Various types of RNA therapeutics including antisense oligonucleotides (ASO), RNA interference (RNAi), small interfering RNA (siRNA), microRNA (miRNA), and messenger RNA (mRNA), have been developed and used for various clinical applications, especially for gene and vaccine delivery purposes. This review is focused on various therapeutic applications of RNA-based delivery systems and then siRNA technologies are discussed in more detail. Next, the FDA-approved siRNA therapeutics and those are in clinical trials are listed and summarized. Then, various viral and non-viral vectors used for RNA delivery purposes are discussed. Finally, clinical applications of siRNA therapeutics are reviewed in detail.
Collapse
Affiliation(s)
- Parisa Ghasemiyeh
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Soliman Mohammadi-Samani
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran; Department of Pharmaceutics, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran.
| |
Collapse
|
2
|
Wang X, Liu F, Wang T, He Y, Guo Y. Applications of hydrogels in tissue-engineered repairing of temporomandibular joint diseases. Biomater Sci 2024; 12:2579-2598. [PMID: 38679944 DOI: 10.1039/d3bm01687k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/01/2024]
Abstract
Epidemiological studies reveal that symptoms of temporomandibular joint disorders (TMDs) occur in 60-70% of adults. The inflammatory damage caused by TMDs can easily lead to defects in the articular disc, condylar cartilage, subchondral bone and muscle of the temporomandibular joint (TMJ) and cause pain. Despite the availability of various methods for treating TMDs, few existing treatment schemes can achieve permanent recovery. This necessity drives the search for new approaches. Hydrogels, polymers with high water content, have found widespread use in tissue engineering and regeneration due to their excellent biocompatibility and mechanical properties, which resemble those of human tissues. In the context of TMD therapy, numerous experiments have demonstrated that hydrogels show favorable effects in aspects such as articular disc repair, cartilage regeneration, muscle repair, pain relief, and drug delivery. This review aims to summarize the application of hydrogels in the therapy of TMDs based on recent research findings. It also highlights deficiencies in current hydrogel research related to TMD therapy and outlines the broad potential of hydrogel applications in treating TMJ diseases in the future.
Collapse
Affiliation(s)
- Xuan Wang
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan, China
| | - Fushuang Liu
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan, China
| | - Tianyi Wang
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan, China
| | - Yikai He
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan, China.
| | - Yongwen Guo
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan, China.
| |
Collapse
|
3
|
Lukin I, Erezuma I, Desimone MF, Zhang YS, Dolatshahi-Pirouz A, Orive G. Nanomaterial-based drug delivery of immunomodulatory factors for bone and cartilage tissue engineering. BIOMATERIALS ADVANCES 2023; 154:213637. [PMID: 37778293 DOI: 10.1016/j.bioadv.2023.213637] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Revised: 09/06/2023] [Accepted: 09/20/2023] [Indexed: 10/03/2023]
Abstract
As life expectancy continues to increase, so do disorders related to the musculoskeletal system. Orthopedics-related impairments remain a challenge, with nearly 325 thousand and 120 thousand deaths recorded in 2019. Musculoskeletal system, including bone and cartilage tissue, is a living system in which cells constantly interact with the immune system, which plays a key role in the tissue repair process. An alternative to bridge the gap between these two systems is exploiting nanomaterials, as they have proven to serve as delivery agents of an array of molecules, including immunomodulatory agents (anti-inflammatory drugs, cytokines), as well as having the ability to mimic tissue by their nanoscopic structure and promote tissue repair per se. Therefore, this review outlooks nanomaterials and immunomodulatory factors widely employed in the area of bone and cartilage tissue engineering. Emerging developments in nanomaterials for delivery of immunomodulatory agents for bone and cartilage tissue engineering applications have also been discussed. It can be concluded that latest progress in nanotechnology have enabled to design intricate systems with the ability to deliver biologically active agents, promoting tissue repair and regeneration; thus, nanomaterials studied herein have shown great potential to serve as immunomodulatory agents in the area of tissue engineering.
Collapse
Affiliation(s)
- Izeia Lukin
- NanoBioCel Research Group, School of Pharmacy, University of the Basque Country (UPV/EHU), Vitoria-Gasteiz, Spain; Bioaraba, NanoBioCel Research Group, Vitoria-Gasteiz, Spain
| | - Itsasne Erezuma
- NanoBioCel Research Group, School of Pharmacy, University of the Basque Country (UPV/EHU), Vitoria-Gasteiz, Spain; Bioaraba, NanoBioCel Research Group, Vitoria-Gasteiz, Spain
| | - Martin F Desimone
- Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Instituto de Química y Metabolismo del Fármaco (IQUIMEFA), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| | - Yu Shrike Zhang
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Cambridge, MA 02139, USA
| | | | - Gorka Orive
- NanoBioCel Research Group, School of Pharmacy, University of the Basque Country (UPV/EHU), Vitoria-Gasteiz, Spain; Bioaraba, NanoBioCel Research Group, Vitoria-Gasteiz, Spain; Biomedical Research Networking Centre in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Vitoria-Gasteiz, Spain; University Institute for Regenerative Medicine and Oral Implantology - UIRMI (UPV/EHU-Fundación Eduardo Anitua), Vitoria 01007, Spain; Singapore Eye Research Institute, The Academia, 20 College Road, Discovery Tower, Singapore 169856, Singapore.
| |
Collapse
|
4
|
Huang X, Pan X, Xiong X, Zhao Z, Cen X. Drug delivery systems for treatment of temporomandibular joint osteoarthritis. Front Pharmacol 2022; 13:1054703. [PMID: 36419625 PMCID: PMC9676453 DOI: 10.3389/fphar.2022.1054703] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Accepted: 10/26/2022] [Indexed: 01/27/2025] Open
Abstract
The number of people suffering from temporomandibular joint osteoarthritis (TMJOA) has been increasing. TMJOA cause joint noise, pain on TMJ and/or masticatory muscles, and restricted mandibular movement, which disturb eating, laughing and conversation, and impose serious lifestyle impediments. Chondrocyte apoptosis, extracellular matrix degradation, synovitis, and subchondral bone remodeling are the main pathological features of TMJOA. Various drug delivery systems are developed to controlled release at specific activation sites with high bioactivity and inhibit rapid dilution to enable long-term therapeutic response, which present great potential for the treatment of TMJOA. This review focuses on recently developed drug delivery systems by different administration in the TMJOA treatment, and summarizes their effects, duration, safety, and limitations, which would pave the way for development of TMJOA therapy.
Collapse
Affiliation(s)
- Xinqi Huang
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Xuefeng Pan
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Xiner Xiong
- Hospital of Stomatology, Zunyi Medical University, Zunyi, China
| | - Zhihe Zhao
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Xiao Cen
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| |
Collapse
|
5
|
Bedingfield SK, Colazo JM, Di Francesco M, Yu F, Liu DD, Di Francesco V, Himmel LE, Gupta MK, Cho H, Hasty KA, Decuzzi P, Duvall CL. Top-Down Fabricated microPlates for Prolonged, Intra-articular Matrix Metalloproteinase 13 siRNA Nanocarrier Delivery to Reduce Post-traumatic Osteoarthritis. ACS NANO 2021; 15:14475-14491. [PMID: 34409835 PMCID: PMC9074946 DOI: 10.1021/acsnano.1c04005] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Post-traumatic osteoarthritis (PTOA) associated with joint injury triggers a degenerative cycle of matrix destruction and inflammatory signaling, leading to pain and loss of function. Here, prolonged RNA interference (RNAi) of matrix metalloproteinase 13 (MMP13) is tested as a PTOA disease modifying therapy. MMP13 is upregulated in PTOA and degrades the key cartilage structural protein type II collagen. Short interfering RNA (siRNA) loaded nanoparticles (siNPs) were encapsulated in shape-defined poly(lactic-co-glycolic acid) (PLGA) based microPlates (μPLs) to formulate siNP-μPLs that maintained siNPs in the joint significantly longer than delivery of free siNPs. Treatment with siNP-μPLs against MMP13 (siMMP13-μPLs) in a mechanical load-induced mouse model of PTOA maintained potent (65-75%) MMP13 gene expression knockdown and reduced MMP13 protein production in joint tissues throughout a 28-day study. MMP13 silencing reduced PTOA articular cartilage degradation/fibrillation, meniscal deterioration, synovial hyperplasia, osteophytes, and pro-inflammatory gene expression, supporting the therapeutic potential of long-lasting siMMP13-μPL therapy for PTOA.
Collapse
Affiliation(s)
- Sean K Bedingfield
- Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee 37232, United States
| | - Juan M. Colazo
- Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee 37232, United States; Vanderbilt University School of Medicine, Vanderbilt University, Nashville, Tennessee 37232, United States; Medical Scientist Training Program, Vanderbilt University School of Medicine, Nashville, Tennessee 37232, United States
| | - Martina Di Francesco
- Laboratory of Nanotechnology for Precision Medicine, Istituto Italiano di Tecnologia, Genoa 16163, Italy
| | - Fang Yu
- Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee 37232, United States
| | - Danielle D. Liu
- Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee 37232, United States; Vanderbilt University School of Medicine, Vanderbilt University, Nashville, Tennessee 37232, United States; Medical Scientist Training Program, Vanderbilt University School of Medicine, Nashville, Tennessee 37232, United States
| | - Valentina Di Francesco
- Laboratory of Nanotechnology for Precision Medicine, Istituto Italiano di Tecnologia, Genoa 16163, Italy
| | - Lauren E. Himmel
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee 37232, United States
| | - Mukesh K. Gupta
- Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee 37232, United States
| | - Hongsik Cho
- Department of Orthopaedic Surgery and Biomedical Engineering, University of Tennessee Health Science Center-Campbell Clinic, Memphis, Tennessee 38104, United States; Research 151, VA Medical Center, Memphis, Tennessee 38104, United States
| | - Karen A. Hasty
- Department of Orthopaedic Surgery and Biomedical Engineering, University of Tennessee Health Science Center-Campbell Clinic, Memphis, Tennessee 38104, United States; Research 151, VA Medical Center, Memphis, Tennessee 38104, United States
| | - Paolo Decuzzi
- Laboratory of Nanotechnology for Precision Medicine, Istituto Italiano di Tecnologia, Genoa 16163, Italy
| | - Craig L. Duvall
- Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee 37232, United States
| |
Collapse
|
6
|
Systematic review of studies on drug-delivery systems for management of temporomandibular-joint osteoarthritis. JOURNAL OF STOMATOLOGY, ORAL AND MAXILLOFACIAL SURGERY 2021; 123:e336-e341. [PMID: 34400376 DOI: 10.1016/j.jormas.2021.08.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Revised: 08/01/2021] [Accepted: 08/11/2021] [Indexed: 02/06/2023]
Abstract
INTRODUCTION Temporomandibular-joint osteoarthritis (TMJOA) management is a major challenge. Minimally invasive therapies (based mainly on injections) have been developed to increase local efficacy and limit adverse systemic effects. However, the requirement for repeat injections due to a short duration of action and expensive healthcare costs have pushed researchers to develop, via tissue engineering, drug-delivery systems (DDSs). In this literature systematic review, we aim to provide an overview of studies that tested DDSs on a TMJOA model. MATERIAL AND METHODS We searched on PubMed for articles published from November 1965 to March 2021 on DDSs using a TMJOA model. We highlighted the different DDSs and the active molecule employed. Route of drug administration, model type, test duration, and efficacy duration were assessed. To evaluate the quality of each study, a protocol bias was tested using QUADAS-2™. RESULTS Of the 10 studies that were full text-screened, four used a poly(lactic-co-glycolic acid)-based delivery system. The other DDSs employed chitosan-based hydrogels, microneedles patches, nanostructured lipid carriers, or poloxamer micelles. Hyaluronic acid, nonsteroidal anti-inflammatory drugs, and analgesics were used as active molecules in five studies. The main way to administer DDSs was intra-articular injection and the most used model was the rat. DISCUSSION Various DDSs and active molecules have been studied on a TMJOA model that could aid TMJOA management. Further works using longer test durations are necessary to validate these advances.
Collapse
|
7
|
Zhang B, Su Y, Zhou J, Zheng Y, Zhu D. Toward a Better Regeneration through Implant-Mediated Immunomodulation: Harnessing the Immune Responses. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2021; 8:e2100446. [PMID: 34117732 PMCID: PMC8373114 DOI: 10.1002/advs.202100446] [Citation(s) in RCA: 69] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Revised: 03/08/2021] [Indexed: 05/06/2023]
Abstract
Tissue repair/regeneration, after implantation or injury, involves comprehensive physiological processes wherein immune responses play a crucial role to enable tissue restoration, amidst the immune cells early-stage response to tissue damages. These cells break down extracellular matrix, clear debris, and secret cytokines to orchestrate regeneration. However, the immune response can also lead to abnormal tissue healing or scar formation if not well directed. This review first introduces the general immune response post injury, with focus on the major immune cells including neutrophils, macrophages, and T cells. Next, a variety of implant-mediated immunomodulation strategies to regulate immune response through physical, chemical, and biological cues are discussed. At last, various scaffold-facilitated regenerations of different tissue types, such as, bone, cartilage, blood vessel, and nerve system, by harnessing the immunomodulation are presented. Therefore, the most recent data in biomaterials and immunomodulation is presented here in a bid to shape expert perspectives, inspire researchers to go in new directions, and drive development of future strategies focusing on targeted, sequential, and dynamic immunomodulation elicited by implants.
Collapse
Affiliation(s)
- Ben Zhang
- Department of Biomedical EngineeringStony Brook UniversityStony BrookNew York11794USA
| | - Yingchao Su
- Department of Biomedical EngineeringStony Brook UniversityStony BrookNew York11794USA
| | - Juncen Zhou
- Department of Biomedical EngineeringStony Brook UniversityStony BrookNew York11794USA
| | - Yufeng Zheng
- Department of Materials Science and EngineeringCollege of EngineeringPeking UniversityBeijing100871China
| | - Donghui Zhu
- Department of Biomedical EngineeringStony Brook UniversityStony BrookNew York11794USA
| |
Collapse
|
8
|
Zhao M, Zhu T, Chen J, Cui Y, Zhang X, Lee RJ, Sun F, Li Y, Teng L. PLGA/PCADK composite microspheres containing hyaluronic acid-chitosan siRNA nanoparticles: A rational design for rheumatoid arthritis therapy. Int J Pharm 2021; 596:120204. [PMID: 33493604 DOI: 10.1016/j.ijpharm.2021.120204] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2020] [Revised: 12/14/2020] [Accepted: 01/01/2021] [Indexed: 12/27/2022]
Abstract
Myeloid cell leukemia-1 (Mcl-1), a member of the Bcl-2 anti-apoptotic family, is overexpressed in the synovial macrophages of patients with rheumatoid arthritis (RA). Small interfering RNA (siRNA) Mcl-1 can induce macrophage apoptosis in the joints and is a potential therapeutic target of RA. Nevertheless, the application of siRNA is limited owing to its instability and susceptibility to degradation in vivo. To address these shortcomings, we developed composite microspheres (MPs) loaded with hyaluronic acid (HA)-chitosan (CS) nanoparticles (NPs). First, we synthesized HA-CS/siRNA NPs (HCNPs) using ionotropic gelation process. Then, HCNPs, as an internal aqueous phase, were loaded into poly (D, L-lactide-co-glycolide) (PLGA) and poly (cyclohexane-1,4-diyl acetone dimethylene ketal) (PCADK) MPs using the double emulsion method. The NPs-in-MPs (NiMPs) composite system provided sustained release of NPs, protected siRNA against nuclease degradation in the serum, and could readily cross the cellular membrane. In addition, we evaluated the advantages of NiMPs in an adjuvant-induced arthritis rat model. Our experimental results demonstrate that NiMPs have greater pharmacodynamic effects than common MPs. Meanwhile, compared with HCNPs, NiMPs reduced the frequency of drug administration. Therefore, NiMPs are a promising and novel siRNA delivery vehicle for RA therapy.
Collapse
Affiliation(s)
- Menghui Zhao
- School of Life Sciences, Jilin University, Changchun, Jilin, China
| | - Tianyu Zhu
- School of Life Sciences, Jilin University, Changchun, Jilin, China
| | - Jicong Chen
- School of Life Sciences, Jilin University, Changchun, Jilin, China
| | - Yaxin Cui
- School of Life Sciences, Jilin University, Changchun, Jilin, China
| | - Xueyan Zhang
- School of Life Sciences, Jilin University, Changchun, Jilin, China
| | - Robert J Lee
- School of Life Sciences, Jilin University, Changchun, Jilin, China; College of Pharmacy, the Ohio State University, Columbus, OH, USA
| | - Fengying Sun
- School of Life Sciences, Jilin University, Changchun, Jilin, China
| | - Youxin Li
- School of Life Sciences, Jilin University, Changchun, Jilin, China.
| | - Lesheng Teng
- School of Life Sciences, Jilin University, Changchun, Jilin, China.
| |
Collapse
|
9
|
Nicot R, Barry F, Chijcheapaza-Flores H, Garcia-Fernandez MJ, Raoul G, Blanchemain N, Chai F. A Systematic Review of Rat Models With Temporomandibular Osteoarthritis Suitable for the Study of Emerging Prolonged Intra-Articular Drug Delivery Systems. J Oral Maxillofac Surg 2021; 79:1650-1671. [PMID: 33775650 DOI: 10.1016/j.joms.2021.02.034] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Revised: 02/20/2021] [Accepted: 02/22/2021] [Indexed: 11/29/2022]
Abstract
PURPOSE Development of minimally invasive therapies for temporomandibular joint osteoarthritis (TMJOA) has focused on drug intra-articular injections to avoid the systemic adverse effects experienced when these substances are administered orally. Therefore, we performed a systematic review to answer the question "Which method of induction of a TMJOA-related pain model in rats leads to prolonged painful symptoms, allowing the best assessment of a sustained drug delivery system?" MATERIALS AND METHODS Following the PRISMA guidelines, we searched MEDLINE for papers published from 1994 to July 2020 on a TMJ arthritis model using rats. We identified the means of pain induction and of nociception assessment. We assessed protocol bias using an adaptation of the QUADAS-2 tool. Animal selection, the reference standard method of pain assessment, applicability of a statistical assessment, and flow and timing were assessed. RESULTS Of the 59 full papers we reviewed, 41 performed no pain assessment after the first 7 days following induction of the TMJ-related pain model. We eventually identified 18 long-term TMJOA-related pain models. Pain was induced by injection of toxic substances, most commonly Freund's complete adjuvant (50 μg per 50 μl), formalin at various concentrations, or monosodium iodoacetate (0,5 mg per 50 μl), into the TMJ, or by physical methods. Few studies reported data on pain after 21 days of follow-up. Heterogeneity of induction methods, pain assessment methods, and flow and timing biases precluded a meta-analysis. CONCLUSIONS Given that pain is 1 of the main symptoms of TMJOA, experimental study protocols should include long-term pain assessment.
Collapse
Affiliation(s)
- Romain Nicot
- Associate Professor, Univ. Lille, CHU Lille, INSERM, Service de Chirurgie Maxillo-Faciale et Stomatologie, U1008 - Controlled Drug Delivery Systems and Biomaterials, Lille, France.
| | - Florent Barry
- Resident, Univ. Lille, CHU Lille, INSERM, Service de Chirurgie Maxillo-Faciale et Stomatologie, U1008 - Controlled Drug Delivery Systems and Biomaterials, Lille, France
| | - Henry Chijcheapaza-Flores
- Research Assistant, Univ. Lille, INSERM, CHU Lille, U1008 - Controlled Drug Delivery Systems and Biomaterials, Lille, France
| | - Maria José Garcia-Fernandez
- Associate Professor, Univ. Lille, INSERM, CHU Lille, U1008 - Controlled Drug Delivery Systems and Biomaterials, Lille, France
| | - Gwénaël Raoul
- Professor, Univ. Lille, CHU Lille, INSERM, Service de Chirurgie Maxillo-Faciale et Stomatologie, U1008 - Controlled Drug Delivery Systems and Biomaterials, Lille, France
| | - Nicolas Blanchemain
- Professor, Department Head, Univ. Lille, INSERM, CHU Lille, U1008 - Controlled Drug Delivery Systems and Biomaterials, Lille, France
| | - Feng Chai
- Research engineer, Univ. Lille, INSERM, CHU Lille, U1008 - Controlled Drug Delivery Systems and Biomaterials, Lille, France
| |
Collapse
|
10
|
Wu M, Cai J, Yu Y, Hu S, Wang Y, Wu M. Therapeutic Agents for the Treatment of Temporomandibular Joint Disorders: Progress and Perspective. Front Pharmacol 2021; 11:596099. [PMID: 33584275 PMCID: PMC7878564 DOI: 10.3389/fphar.2020.596099] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Accepted: 11/19/2020] [Indexed: 02/05/2023] Open
Abstract
Temporomandibular joint disorders (TMD) are a common health condition caused by the structural or functional disorders of masticatory muscles and the temporomandibular joint (TMJ). Abnormal mandibular movement in TMD patients may cause pain, chronic inflammation, and other discomfort, which could be relieved by a variety of drugs through various delivery systems. In this study, we summarized commonly used therapeutic agents in the management of TMD as well as novel bioactive molecules in preclinical stage and clinical trials. The emerging therapy strategies such as novel intra-TMJ delivery systems and implants based on tissue engineering are also discussed. This comprehensive review will strengthen our understanding of pharmacological approaches for TMD therapy.
Collapse
Affiliation(s)
- Mengjie Wu
- The Affiliated Hospital of Stomatology, School of Stomatology, Zhejiang University School of Medicine, and Key Laboratory of Oral Biomedical Research of Zhejiang Province, Hangzhou, China
| | - Jingyi Cai
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases and Department of Orthodontics, West China Hospital of Stomato-logy, Sichuan University, Chengdu, China
| | - Yeke Yu
- Shanghai Key Laboratory of Stomatology and Shanghai Research Institute of Stomatology, National Clinical Research Center of Stomatology, Department of Oral Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Sihui Hu
- The Affiliated Hospital of Stomatology, School of Stomatology, Zhejiang University School of Medicine, and Key Laboratory of Oral Biomedical Research of Zhejiang Province, Hangzhou, China
| | - Yingnan Wang
- The Affiliated Hospital of Stomatology, School of Stomatology, Zhejiang University School of Medicine, and Key Laboratory of Oral Biomedical Research of Zhejiang Province, Hangzhou, China
| | - Mengrui Wu
- College of Life Sciences, Zhejiang University, Zhejiang, China
| |
Collapse
|
11
|
Macrophages Are Key Regulators of Stem Cells during Skeletal Muscle Regeneration and Diseases. Stem Cells Int 2019; 2019:4761427. [PMID: 31396285 PMCID: PMC6664695 DOI: 10.1155/2019/4761427] [Citation(s) in RCA: 107] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2019] [Accepted: 06/09/2019] [Indexed: 12/31/2022] Open
Abstract
Muscle regeneration is a closely regulated process that involves a variety of cell types such as satellite cells, myofibers, fibroadipogenic progenitors, endothelial cells, and inflammatory cells. Among these different cell types, macrophages emerged as a central actor coordinating the different cellular interactions and biological processes. Particularly, the transition of macrophages from their proinflammatory to their anti-inflammatory phenotype was shown to regulate inflammation, myogenesis, fibrosis, vascularization, and return to homeostasis. On the other hand, deregulation of macrophage accumulation or polarization in chronic degenerative muscle disorders was shown to impair muscle regeneration. Considering the key roles of macrophages in skeletal muscle, they represent an attractive target for new therapeutic approaches aiming at mitigating various muscle disorders. This review aims at summarizing the novel insights into macrophage heterogeneity, plasticity, and functions in skeletal muscle homeostasis, regeneration, and disease.
Collapse
|
12
|
Biomaterials: Foreign Bodies or Tuners for the Immune Response? Int J Mol Sci 2019; 20:ijms20030636. [PMID: 30717232 PMCID: PMC6386828 DOI: 10.3390/ijms20030636] [Citation(s) in RCA: 363] [Impact Index Per Article: 60.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2018] [Revised: 01/22/2019] [Accepted: 01/28/2019] [Indexed: 12/11/2022] Open
Abstract
The perspectives of regenerative medicine are still severely hampered by the host response to biomaterial implantation, despite the robustness of technologies that hold the promise to recover the functionality of damaged organs and tissues. In this scenario, the cellular and molecular events that decide on implant success and tissue regeneration are played at the interface between the foreign body and the host inflammation, determined by innate and adaptive immune responses. To avoid adverse events, rather than the use of inert scaffolds, current state of the art points to the use of immunomodulatory biomaterials and their knowledge-based use to reduce neutrophil activation, and optimize M1 to M2 macrophage polarization, Th1 to Th2 lymphocyte switch, and Treg induction. Despite the fact that the field is still evolving and much remains to be accomplished, recent research breakthroughs have provided a broader insight on the correct choice of biomaterial physicochemical modifications to tune the reaction of the host immune system to implanted biomaterial and to favor integration and healing.
Collapse
|
13
|
Dashnyam K, Lee JH, Mandakhbayar N, Jin GZ, Lee HH, Kim HW. Intra-articular biomaterials-assisted delivery to treat temporomandibular joint disorders. J Tissue Eng 2018; 9:2041731418776514. [PMID: 29785258 PMCID: PMC5954570 DOI: 10.1177/2041731418776514] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2018] [Accepted: 04/23/2018] [Indexed: 12/19/2022] Open
Abstract
The temporomandibular joint disorder, also known as myofascial pain syndrome, is considered one of the prevalent chronic pain diseases caused by muscle inflammation and cartilage degradation in head and neck, and thus influences even biopsychosocial conditions in a lifetime. There are several current treatment methodologies relieving inflammation and preventing degradation of the joint complex. One of the promising non-surgical treatment methods is an intra-articular injection of drugs such as corticosteroids, analgesics, and anti-depressants. However, the side effects of drugs due to frequent injections and over-doses, including dizziness, dry mouth, and possible drug dependency are considered limitations. Thus, the delivery of therapeutic molecules through the use of nano/microparticles is currently considered as a promising strategy primarily due to the controlled release. This review highlights the nano/microparticle systems for effective intra-articular therapeutics delivery to prevent cartilage degradation and protect subchondral bone in a temporomandibular joint.
Collapse
Affiliation(s)
- Khandmaa Dashnyam
- Institute of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan, South Korea.,Department of Nanobiomedical Science & BK21 PLUS Global Research Center for Regenerative Medicine, Dankook University, Cheonan, South Korea
| | - Jung-Hwan Lee
- Institute of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan, South Korea.,Department of Biomaterials Science, College of Dentistry, Dankook University, Cheonan, South Korea
| | - Nandin Mandakhbayar
- Institute of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan, South Korea.,Department of Nanobiomedical Science & BK21 PLUS Global Research Center for Regenerative Medicine, Dankook University, Cheonan, South Korea
| | - Guang-Zhen Jin
- Institute of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan, South Korea.,Department of Biomaterials Science, College of Dentistry, Dankook University, Cheonan, South Korea
| | - Hae-Hyoung Lee
- Institute of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan, South Korea.,Department of Biomaterials Science, College of Dentistry, Dankook University, Cheonan, South Korea
| | - Hae-Won Kim
- Institute of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan, South Korea.,Department of Nanobiomedical Science & BK21 PLUS Global Research Center for Regenerative Medicine, Dankook University, Cheonan, South Korea.,Department of Biomaterials Science, College of Dentistry, Dankook University, Cheonan, South Korea
| |
Collapse
|
14
|
Recent development of synthetic nonviral systems for sustained gene delivery. Drug Discov Today 2017; 22:1318-1335. [PMID: 28428056 DOI: 10.1016/j.drudis.2017.04.001] [Citation(s) in RCA: 84] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2016] [Revised: 03/02/2017] [Accepted: 04/05/2017] [Indexed: 12/22/2022]
Abstract
Sustained gene delivery is of particular importance today because it circumvents the need for repeated therapeutic administration and provides spatial and temporal control of the release profile. Better understanding of the genetic basis of diseases and advances in gene therapy have propelled significant research on biocompatible gene carriers for therapeutic purposes. Varied biodegradable polymer-based architectures have been used to create new compositions with unique properties suitable for sustained gene delivery. This review presents the most recent advances in various polymeric systems: hydrogels, microspheres, nanospheres and scaffolds, having complex architectures to encapsulate and deliver functional genes. Through the recombination of different existing polymer systems, the multicomplex systems can be further endowed with new properties for better-targeted biomedical applications.
Collapse
|
15
|
Julier Z, Park AJ, Briquez PS, Martino MM. Promoting tissue regeneration by modulating the immune system. Acta Biomater 2017; 53:13-28. [PMID: 28119112 DOI: 10.1016/j.actbio.2017.01.056] [Citation(s) in RCA: 502] [Impact Index Per Article: 62.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2016] [Revised: 01/03/2017] [Accepted: 01/20/2017] [Indexed: 02/07/2023]
Abstract
The immune system plays a central role in tissue repair and regeneration. Indeed, the immune response to tissue injury is crucial in determining the speed and the outcome of the healing process, including the extent of scarring and the restoration of organ function. Therefore, controlling immune components via biomaterials and drug delivery systems is becoming an attractive approach in regenerative medicine, since therapies based on stem cells and growth factors have not yet proven to be broadly effective in the clinic. To integrate the immune system into regenerative strategies, one of the first challenges is to understand the precise functions of the different immune components during the tissue healing process. While remarkable progress has been made, the immune mechanisms involved are still elusive, and there is indication for both negative and positive roles depending on the tissue type or organ and life stage. It is well recognized that the innate immune response comprising danger signals, neutrophils and macrophages modulates tissue healing. In addition, it is becoming evident that the adaptive immune response, in particular T cell subset activities, plays a critical role. In this review, we first present an overview of the basic immune mechanisms involved in tissue repair and regeneration. Then, we highlight various approaches based on biomaterials and drug delivery systems that aim at modulating these mechanisms to limit fibrosis and promote regeneration. We propose that the next generation of regenerative therapies may evolve from typical biomaterial-, stem cell-, or growth factor-centric approaches to an immune-centric approach. STATEMENT OF SIGNIFICANCE Most regenerative strategies have not yet proven to be safe or reasonably efficient in the clinic. In addition to stem cells and growth factors, the immune system plays a crucial role in the tissue healing process. Here, we propose that controlling the immune-mediated mechanisms of tissue repair and regeneration may support existing regenerative strategies or could be an alternative to using stem cells and growth factors. The first part of this review we highlight key immune mechanisms involved in the tissue healing process and marks them as potential target for designing regenerative strategies. In the second part, we discuss various approaches using biomaterials and drug delivery systems that aim at modulating the components of the immune system to promote tissue regeneration.
Collapse
Affiliation(s)
- Ziad Julier
- European Molecular Biology Laboratory Australia, Australian Regenerative Medicine Institute, Monash University, Victoria 3800, Australia
| | - Anthony J Park
- European Molecular Biology Laboratory Australia, Australian Regenerative Medicine Institute, Monash University, Victoria 3800, Australia
| | - Priscilla S Briquez
- Institute for Molecular Engineering, University of Chicago, Chicago, IL 60637, USA
| | - Mikaël M Martino
- European Molecular Biology Laboratory Australia, Australian Regenerative Medicine Institute, Monash University, Victoria 3800, Australia.
| |
Collapse
|
16
|
Wang LL, Burdick JA. Engineered Hydrogels for Local and Sustained Delivery of RNA-Interference Therapies. Adv Healthc Mater 2017; 6:10.1002/adhm.201601041. [PMID: 27976524 PMCID: PMC5226889 DOI: 10.1002/adhm.201601041] [Citation(s) in RCA: 67] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2016] [Revised: 10/21/2016] [Indexed: 12/20/2022]
Abstract
It has been nearly two decades since RNA-interference (RNAi) was first reported. While there are no approved clinical uses, several phase II and III clinical trials suggest the great promise of RNAi therapeutics. One challenge for RNAi therapies is the controlled localization and sustained presentation to target tissues, to both overcome systemic toxicity concerns and to enhance in vivo efficacy. One approach that is emerging to address these limitations is the entrapment of RNAi molecules within hydrogels for local and sustained release. In these systems, nucleic acids are either delivered as siRNA conjugates or within nanoparticles. A plethora of hydrogels has been implemented using these approaches, including both traditional hydrogels that have already been developed for other applications and new hydrogels developed specifically for RNAi delivery. These hydrogels have been applied to various applications in vivo, including cancer, bone regeneration, inflammation and cardiac repair. This review will examine the design and implementation of such hydrogel RNAi systems and will cover the most recent applications of these systems.
Collapse
Affiliation(s)
- Leo L. Wang
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA 19104
| | - Jason A. Burdick
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA 19104
| |
Collapse
|
17
|
Huynh CT, Nguyen MK, Tonga GY, Longé L, Rotello VM, Alsberg E. Photocleavable Hydrogels for Light-Triggered siRNA Release. Adv Healthc Mater 2016; 5:305-310. [PMID: 26639103 PMCID: PMC4755586 DOI: 10.1002/adhm.201500778] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2015] [Indexed: 01/22/2023]
Abstract
A photocleavable hydrogel system for on-demand delivery of genetic material is reported. The release of short interfering RNAs can be triggered by the application of UV light without any loss in bioactivity. This approach provides a promising external stimulus-based nucleic acid delivery platform for applications in disease therapeutics and tissue regeneration.
Collapse
Affiliation(s)
- Cong Truc Huynh
- Department of Biomedical Engineering, Case Western Reserve University, 10900 Euclid Avenue, Cleveland, OH 44106, USA
| | - Minh Khanh Nguyen
- Department of Biomedical Engineering, Case Western Reserve University, 10900 Euclid Avenue, Cleveland, OH 44106, USA
| | - Gulen Yesilbag Tonga
- Department of Chemistry, University of Massachusetts, 710 North Pleasant Street, Amherst, MA 01003, USA
| | - Lionel Longé
- Department of Chemistry, University of Massachusetts, 710 North Pleasant Street, Amherst, MA 01003, USA
- Département Chimie Physique, École Nationale Supérieure de Chimie, de Biologie et de Physique 16, avenue Pey Berland 33607 PESSAC Cedex, France
| | - Vincent M. Rotello
- Department of Chemistry, University of Massachusetts, 710 North Pleasant Street, Amherst, MA 01003, USA
| | - Eben Alsberg
- Department of Biomedical Engineering, Case Western Reserve University, 10900 Euclid Avenue, Cleveland, OH 44106, USA
- Department of Orthopaedic Surgery, Case Western Reserve University, 10900 Euclid Avenue, Cleveland, OH 44106, USA
| |
Collapse
|
18
|
Sarett SM, Nelson CE, Duvall CL. Technologies for controlled, local delivery of siRNA. J Control Release 2015; 218:94-113. [PMID: 26476177 PMCID: PMC4665980 DOI: 10.1016/j.jconrel.2015.09.066] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2015] [Revised: 09/25/2015] [Accepted: 09/29/2015] [Indexed: 12/24/2022]
Abstract
The discovery of RNAi in the late 1990s unlocked a new realm of therapeutic possibilities by enabling potent and specific silencing of theoretically any desired genetic target. Better elucidation of the mechanism of action, the impact of chemical modifications that stabilize and reduce nonspecific effects of siRNA molecules, and the key design considerations for effective delivery systems has spurred progress toward developing clinically-successful siRNA therapies. A logical aim for initial siRNA translation is local therapies, as delivering siRNA directly to its site of action helps to ensure that a sufficient dose reaches the target tissue, lessens the potential for off-target side effects, and circumvents the substantial systemic delivery barriers. While locally injected or topically applied siRNA has progressed into numerous clinical trials, an enormous opportunity exists to develop sustained-release, local delivery systems that enable both spatial and temporal control of gene silencing. This review focuses on material platforms that establish both localized and controlled gene silencing, with emphasis on the systems that show most promise for clinical translation.
Collapse
Affiliation(s)
- Samantha M Sarett
- Vanderbilt University Department of Biomedical Engineering, United States
| | | | - Craig L Duvall
- Vanderbilt University Department of Biomedical Engineering, United States.
| |
Collapse
|
19
|
siRNA Versus miRNA as Therapeutics for Gene Silencing. MOLECULAR THERAPY. NUCLEIC ACIDS 2015; 4:e252. [PMID: 26372022 PMCID: PMC4877448 DOI: 10.1038/mtna.2015.23] [Citation(s) in RCA: 703] [Impact Index Per Article: 70.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/23/2015] [Accepted: 07/18/2015] [Indexed: 02/06/2023]
Abstract
Discovered a little over two decades ago, small interfering RNAs (siRNAs) and microRNAs (miRNAs) are noncoding RNAs with important roles in gene regulation. They have recently been investigated as novel classes of therapeutic agents for the treatment of a wide range of disorders including cancers and infections. Clinical trials of siRNA- and miRNA-based drugs have already been initiated. siRNAs and miRNAs share many similarities, both are short duplex RNA molecules that exert gene silencing effects at the post-transcriptional level by targeting messenger RNA (mRNA), yet their mechanisms of action and clinical applications are distinct. The major difference between siRNAs and miRNAs is that the former are highly specific with only one mRNA target, whereas the latter have multiple targets. The therapeutic approaches of siRNAs and miRNAs are therefore very different. Hence, this review provides a comparison between therapeutic siRNAs and miRNAs in terms of their mechanisms of action, physicochemical properties, delivery, and clinical applications. Moreover, the challenges in developing both classes of RNA as therapeutics are also discussed.
Collapse
|
20
|
Zhu H, Yang F, Tang B, Li XM, Chu YN, Liu YL, Wang SG, Wu DC, Zhang Y. Mesenchymal stem cells attenuated PLGA-induced inflammatory responses by inhibiting host DC maturation and function. Biomaterials 2015; 53:688-98. [DOI: 10.1016/j.biomaterials.2015.03.005] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2014] [Revised: 02/27/2015] [Accepted: 03/04/2015] [Indexed: 12/29/2022]
|
21
|
Browne S, Pandit A. Biomaterial-mediated modification of the local inflammatory environment. Front Bioeng Biotechnol 2015; 3:67. [PMID: 26029692 PMCID: PMC4432793 DOI: 10.3389/fbioe.2015.00067] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2015] [Accepted: 04/30/2015] [Indexed: 12/14/2022] Open
Abstract
Inflammation plays a major role in the rejection of biomaterial implants. In addition, despite playing an important role in the early stages of wound healing, dysregulated inflammation has a negative impact on the wound healing processes. Thus, strategies to modulate excessive inflammation are needed. Through the use of biomaterials to control the release of anti-inflammatory therapeutics, increased control over inflammation is possible in a range of pathological conditions. However, the choice of biomaterial (natural or synthetic), and the form it takes (solid, hydrogel, or micro/nanoparticle) is dependent on both the cause and tissue location of inflammation. These considerations also influence the nature of the anti-inflammatory therapeutic that is incorporated into the biomaterial to be delivered. In this report, the range of biomaterials and anti-inflammatory therapeutics that have been combined will be discussed, as well as the functional benefit observed. Furthermore, we point toward future strategies in the field that will bring more efficacious anti-inflammatory therapeutics closer to realization.
Collapse
Affiliation(s)
- Shane Browne
- Network of Excellence for Functional Biomaterials (NFB), National University of Ireland, Galway, Ireland
| | - Abhay Pandit
- Network of Excellence for Functional Biomaterials (NFB), National University of Ireland, Galway, Ireland
| |
Collapse
|
22
|
He C, Yin L, Song Y, Tang C, Yin C. Optimization of multifunctional chitosan-siRNA nanoparticles for oral delivery applications, targeting TNF-α silencing in rats. Acta Biomater 2015; 17:98-106. [PMID: 25662912 DOI: 10.1016/j.actbio.2015.01.041] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2014] [Revised: 01/28/2015] [Accepted: 01/28/2015] [Indexed: 11/28/2022]
Abstract
Secretion of tumor necrosis factor-α (TNF-α) by macrophages plays a predominant role in the development and progression of various inflammatory diseases. In the current contribution, multifunctional nanoparticles (NPs) containing TNF-α siRNA targeting macrophages via oral administration were developed to knockdown TNF-α expression against acute hepatic injury in rats. Mannose-modified trimethyl chitosan-cysteine (MTC) NPs were prepared by self-assembly method (sa-MTC NPs), ionic gelation and siRNA entrapment method (en-MTC NPs), and ionic gelation and siRNA adsorption method (ad-MTC NPs). Among them, en-MTC NPs demonstrated the best stability against ionic challenges with desired siRNA integrity against nucleases. By targeting normal enterocytes and M cells that express mannose receptors, en-MTC NPs notably promoted intestinal absorption of siRNA in rats. They further facilitated siRNA internalization by rat peritoneal exudate cells (PECs) via lipid-raft involved endocytosis and macropinocytosis, thus inducing effective in vitro TNF-α knockdown. Orally delivered en-MTC NPs at a low siRNA dose of 50 μg/kg inhibited systemic TNF-α production and decreased TNF-α mRNA levels in macrophage-enriched liver, spleen, and lung tissues, which consequently protected rats from acute hepatic injury. Therefore, the en-MTC NPs would provide an effective approach to orally deliver TNF-α siRNA for the anti-inflammatory therapy.
Collapse
Affiliation(s)
- Chunbai He
- State Key Laboratory of Genetic Engineering, Department of Pharmaceutical Sciences, School of Life Sciences, Fudan University, Shanghai 200433, China
| | - Lichen Yin
- State Key Laboratory of Genetic Engineering, Department of Pharmaceutical Sciences, School of Life Sciences, Fudan University, Shanghai 200433, China
| | - Yudong Song
- State Key Laboratory of Genetic Engineering, Department of Pharmaceutical Sciences, School of Life Sciences, Fudan University, Shanghai 200433, China
| | - Cui Tang
- State Key Laboratory of Genetic Engineering, Department of Pharmaceutical Sciences, School of Life Sciences, Fudan University, Shanghai 200433, China
| | - Chunhua Yin
- State Key Laboratory of Genetic Engineering, Department of Pharmaceutical Sciences, School of Life Sciences, Fudan University, Shanghai 200433, China.
| |
Collapse
|
23
|
Merkel OM, Kissel T. Quo vadis polyplex? J Control Release 2014; 190:415-23. [DOI: 10.1016/j.jconrel.2014.06.009] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2014] [Revised: 06/06/2014] [Accepted: 06/06/2014] [Indexed: 12/24/2022]
|
24
|
Drugs and Polymers for Delivery Systems in OA Joints: Clinical Needs and Opportunities. Polymers (Basel) 2014. [DOI: 10.3390/polym6030799] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
|
25
|
Nelson CE, Kim AJ, Adolph EJ, Gupta MK, Yu F, Hocking KM, Davidson JM, Guelcher SA, Duvall CL. Tunable delivery of siRNA from a biodegradable scaffold to promote angiogenesis in vivo. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2014; 26:607-14, 506. [PMID: 24338842 PMCID: PMC3951880 DOI: 10.1002/adma.201303520] [Citation(s) in RCA: 89] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/29/2013] [Revised: 10/14/2013] [Indexed: 05/17/2023]
Abstract
A system has been engineered for temporally controlled delivery of siRNA from biodegradable tissue regenerative scaffolds. Therapeutic application of this approach to silence prolyl hydroxylase domain 2 promoted expression of pro-angiogenic genes controlled by HIF1α and enhanced scaffold vascularization in vivo. This technology provides a new standard for efficient and controllable gene silencing to modulate host response within regenerative biomaterials.
Collapse
Affiliation(s)
- Christopher E. Nelson
- Biomedical Engineering – Vanderbilt University, VU Station B, Box 351631, Nashville TN 37235
| | - Arnold J. Kim
- Biomedical Engineering – Vanderbilt University, VU Station B, Box 351631, Nashville TN 37235
| | - Elizabeth J. Adolph
- Chemical and Biomolecular Engineering – Vanderbilt University, Nashville TN 37235
| | - Mukesh K. Gupta
- Biomedical Engineering – Vanderbilt University, VU Station B, Box 351631, Nashville TN 37235
| | - Fang Yu
- Department of Pathology – Vanderbilt University, Nashville TN 37235
| | - Kyle M. Hocking
- Biomedical Engineering – Vanderbilt University, VU Station B, Box 351631, Nashville TN 37235
| | | | - Scott A. Guelcher
- Chemical and Biomolecular Engineering – Vanderbilt University, Nashville TN 37235
| | - Craig L. Duvall
- Biomedical Engineering – Vanderbilt University, VU Station B, Box 351631, Nashville TN 37235
- Prof. C.L. Duvall, Vanderbilt University, VU Station B, Box 351631, Nashville, TN, 37235, USA,
| |
Collapse
|
26
|
Needham CJ, Shah SR, Mountziaris PM, Kasper FK, Mikos AG. Modulation of Polyplex Release from Biodegradable Microparticles through Poly(ethylenimine) Modification and Varying Loading Concentration. Pharm Res 2013; 31:77-85. [DOI: 10.1007/s11095-013-1133-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2013] [Accepted: 06/24/2013] [Indexed: 10/26/2022]
|
27
|
Höbel S, Aigner A. Polyethylenimines for siRNA and miRNA delivery in vivo. WILEY INTERDISCIPLINARY REVIEWS-NANOMEDICINE AND NANOBIOTECHNOLOGY 2013; 5:484-501. [PMID: 23720168 DOI: 10.1002/wnan.1228] [Citation(s) in RCA: 145] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Received: 12/06/2012] [Revised: 02/28/2013] [Accepted: 03/19/2013] [Indexed: 12/23/2022]
Abstract
The discovery of RNA interference (RNAi) as a naturally occurring mechanism for gene knockdown has attracted considerable attention toward the use of small interfering RNAs (siRNAs) for therapeutic purposes. Likewise, microRNAs (miRNAs) have emerged as important cellular regulators of gene expression, and their pathological underexpression allows for novel therapeutic strategies ('miRNA replacement therapy'). To address issues related to the instability, charge, and molecular weight of small RNA molecules, nanoparticle formulations have been explored for their in vivo application. Polyethylenimines (PEIs) are positively charged, linear, or branched polymers that are able to form nanoscale complexes with small RNAs, leading to RNA protection, cellular delivery, and intracellular release. This review highlights the important properties of various PEIs with regard to their use for in vivo RNA delivery. PEI modifications for increased efficacy, altered pharmacokinetic properties, improved biocompatibility and, upon covalent coupling of ligands, targeted delivery are described. An overview of various modified PEIs and a comprehensive list of representative studies using PEI-based siRNA or miRNA delivery in vivo are given.
Collapse
Affiliation(s)
- Sabrina Höbel
- Clinical Pharmacology, Faculty of Medicine, Rudolf-Boehm-Institute of Pharmacology and Toxicology, University Leipzig, Leipzig, Germany
| | | |
Collapse
|
28
|
Liu X, Ma L, Liang J, Zhang B, Teng J, Gao C. RNAi functionalized collagen-chitosan/silicone membrane bilayer dermal equivalent for full-thickness skin regeneration with inhibited scarring. Biomaterials 2012; 34:2038-48. [PMID: 23261213 DOI: 10.1016/j.biomaterials.2012.11.062] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2012] [Accepted: 11/29/2012] [Indexed: 12/24/2022]
Abstract
Scar inhibition of dermal equivalent is one of the key issues for treatment of full thickness skin defects. To yield a bioactive RNAi functionalized matrix for skin regeneration with inhibited scarring, collagen-chitosan/silicone membrane bilayer dermal equivalent (BDE) was combined with trimetylchitosan (TMC)/siRNA complexes which could induce suppression of transforming growth factor-β1 (TGF-β1) pathway. The RNAi-BDE functioned as a reservoir for the incorporated TMC/siRNA complexes, enabling a prolonged siRNA release. The seeded fibroblasts in the RNAi-BDE showed good viability, internalized the TMC/siRNA complexes effectively and suppressed TGF-β1 expression constantly until 14 d. Application of the RNAi-BDE on the full-thickness skin defects of pig backs confirmed the in vivo inhibition of TGF-β1 expression by immunohistochemistry, real-time quantitative PCR and western blotting during 30 d post surgery. The levels of other scar-related factors such as collagen type I, collagen type III and α-smooth muscle actin (α-SMA) were also down-regulated. In combination with the ultra-thin skin graft transplantation for 73 d, the regenerated skin by RNAi-BDE had an extremely similar structure to that of the normal one. Our study reflects the latest paradigm of tissue engineering by incorporating the emerging biomolecule siRNA. The 3-D scaffolding materials for siRNA delivery may have general implications in generation of bioactive matrix as well.
Collapse
Affiliation(s)
- Xing Liu
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou 310027, China
| | | | | | | | | | | |
Collapse
|