51
|
Mehta P, Haj-Ahmad R, Rasekh M, Arshad MS, Smith A, van der Merwe SM, Li X, Chang MW, Ahmad Z. Pharmaceutical and biomaterial engineering via electrohydrodynamic atomization technologies. Drug Discov Today 2017; 22:157-165. [DOI: 10.1016/j.drudis.2016.09.021] [Citation(s) in RCA: 79] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2016] [Revised: 08/17/2016] [Accepted: 09/22/2016] [Indexed: 10/20/2022]
|
52
|
Tsekoura EK, K C RB, Uludag H. Biomaterials to Facilitate Delivery of RNA Agents in Bone Regeneration and Repair. ACS Biomater Sci Eng 2016; 3:1195-1206. [PMID: 33440509 DOI: 10.1021/acsbiomaterials.6b00387] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Bone healing after traumatic injuries or pathological diseases remains an important worldwide problem. In search of safer and more effective approaches to bone regeneration and repair, RNA-based therapeutic agents, specifically microRNAs (miRNAs) and short interfering RNA (siRNA), are beginning to be actively explored. In this review, we summarize current attempts to employ miRNAs and siRNAs in preclinical models of bone repair. We provide a summary of current limitations when attempting to utilize bioactive nucleic acids for therapeutic purposes and position the unique aspects of RNA reagents for clinical bone repair. Delivery strategies for RNA reagents are emphasized and nonviral carriers (biomaterial-based) employed to deliver such reagents are reviewed. Critical features of biomaterial carriers and various delivery technologies centered around nanoparticulate systems are highlighted. We conclude with the authors' perspectives on the future of the field, outlining main critical issues important to address as RNA reagents are explored for clinical applications.
Collapse
Affiliation(s)
- Eleni K Tsekoura
- Department of Chemical & Materials Engineering, Faculty of Engineering, ‡Department of Biomedical Engineering, Faculty of Medicine & Dentistry, and §Faculty of Pharmacy & Pharmaceutical Sciences, University of Alberta, Edmonton, Alberta T6G 2R3, Canada
| | - Remant Bahadur K C
- Department of Chemical & Materials Engineering, Faculty of Engineering, Department of Biomedical Engineering, Faculty of Medicine & Dentistry, and §Faculty of Pharmacy & Pharmaceutical Sciences, University of Alberta, Edmonton, Alberta T6G 2R3, Canada
| | - Hasan Uludag
- Department of Chemical & Materials Engineering, Faculty of Engineering, Department of Biomedical Engineering, Faculty of Medicine & Dentistry, and Faculty of Pharmacy & Pharmaceutical Sciences, University of Alberta, Edmonton, Alberta T6G 2R3, Canada
| |
Collapse
|
53
|
Zhou F, Jia X, Yang Y, Yang Q, Gao C, Hu S, Zhao Y, Fan Y, Yuan X. Nanofiber-mediated microRNA-126 delivery to vascular endothelial cells for blood vessel regeneration. Acta Biomater 2016; 43:303-313. [PMID: 27477849 DOI: 10.1016/j.actbio.2016.07.048] [Citation(s) in RCA: 69] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2016] [Revised: 07/19/2016] [Accepted: 07/27/2016] [Indexed: 11/28/2022]
Abstract
UNLABELLED As manipulation of gene expression by virtue of microRNAs (miRNAs) is one of the emerging strategies for cardiovascular disease remedy, local delivery of miRNAs to a specific vascular tissue is challenging. In this work, we developed an efficient delivery system composed of electrospun fibrous membranes and target carriers for the intracellular delivery of miRNA-126 (miR-126) to vascular endothelial cells (VECs) in the local specific vascular environment. A bilayer vascular scaffold was specially prepared via emulsion electrospinning of poly(ethylene glycol)-b-poly(l-lactide-co-ε-caprolactone) (PELCL) and dual-power electrospinning of poly(ε-caprolactone) (PCL) and gelatin. The inner layer of PELCL, which was loaded with complexes of miR-126 in REDV peptide-modified trimethyl chitosan-g-poly(ethylene glycol), regulated the response of VECs, while the outer layer of PCL/gelatin contributed to the mechanical stability. Biological activities of the miR-126-loaded electrospun membranes were evaluated by cell proliferation and SPRED-1 expression of a miR-126 target gene. By encapsulating targeting complexes of miR-126 in the electrospun membranes, a sustained release profile of miRNA was obtained for 56days. Significant down-regulation of SPRED-1 gene expression in VECs was detected on day 3, and it was found that miR-126 released from the electrospun membranes accelerated VEC proliferation in the first 9days. The bilayer vascular scaffold loaded with miR-126 complexes could also improve endothelialization in vivo. These results demonstrated the potential of this approach towards a new and more effective delivering system for local delivery of miRNAs to facilitate blood vessel regeneration. STATEMENT OF SIGNIFICANCE Tissue engineering of small-diameter blood vessels is still challenging because of thrombosis and low long-term patency. The manipulation of gene expression by miRNAs could be a novel strategy in vascular regeneration. Here, we report an efficient delivery system of electrospun fibrous scaffold combined with REDV peptide-modified trimethyl chitosan for targeted intracellular delivery of miR-126 to VECs in the local vascular environment. Results exhibited that miR-126 released from the electrospun membrane could modulate VEC proliferation via down-regulation of SPRED-1 gene expression. The electrospun scaffolds loaded with target-delivery carriers may serve as an ideal platform for local delivery of miRNAs in the vascular tissue engineering.
Collapse
Affiliation(s)
- Fang Zhou
- School of Materials Science and Engineering, and Tianjin Key Laboratory of Composite and Functional Materials, Tianjin University, Tianjin 300072, China
| | - Xiaoling Jia
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, School of Biological Science and Medical Engineering, Beihang University, Beijing 100083, China
| | - Yang Yang
- School of Materials Science and Engineering, and Tianjin Key Laboratory of Composite and Functional Materials, Tianjin University, Tianjin 300072, China
| | - Qingmao Yang
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, School of Biological Science and Medical Engineering, Beihang University, Beijing 100083, China
| | - Chao Gao
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, School of Biological Science and Medical Engineering, Beihang University, Beijing 100083, China
| | - Suli Hu
- School of Materials Science and Engineering, and Tianjin Key Laboratory of Composite and Functional Materials, Tianjin University, Tianjin 300072, China
| | - Yunhui Zhao
- School of Materials Science and Engineering, and Tianjin Key Laboratory of Composite and Functional Materials, Tianjin University, Tianjin 300072, China
| | - Yubo Fan
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, School of Biological Science and Medical Engineering, Beihang University, Beijing 100083, China; National Research Center for Rehabilitation Technical Aids, Beijing 100176, China.
| | - Xiaoyan Yuan
- School of Materials Science and Engineering, and Tianjin Key Laboratory of Composite and Functional Materials, Tianjin University, Tianjin 300072, China.
| |
Collapse
|
54
|
Mohammadian F, Eatemadi A. Drug loading and delivery using nanofibers scaffolds. ARTIFICIAL CELLS NANOMEDICINE AND BIOTECHNOLOGY 2016; 45:881-888. [PMID: 27188394 DOI: 10.1080/21691401.2016.1185726] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
In recent times, notable advancement has been made in the field of electrospinning for the fabrication of numerous types of nanofiber scaffolds. Due to the ultrathin fiber diameter, electrospun nanofiber scaffolds are considered to be an operational delivery system for biomolecules, genes, as well as drugs due to the high specific surface area and stereological porous structure. Here, we introduce some of methods for the integration of drugs and biomolecules within electrospun nanofiber scaffolds, such as blending, surface modification, coaxial process, and emulsion methods. Then, we describe some important biomedical applications of nanofibers in drug delivery systems along with their suitable examples in transdermal systems and wound dressings, cancer therapy, growth factor delivery, nucleic acid delivery, and stem cell delivery.
Collapse
Affiliation(s)
- Farideh Mohammadian
- a Department of Medical Biotechnology, Faculty of Advance Medical Sciences , Tabriz University of Medical Sciences , Tabriz , Iran
| | - Ali Eatemadi
- b Department of Medical Biotechnology, School of Advance Science in Medicine , Tehran University of Medical Sciences , Tehran , Iran
| |
Collapse
|
55
|
Amgoth C, Dharmapuri G, Kalle AM, Paik P. Nanoporous capsules of block co-polymers of [(MeO-PEG-NH)-b-(L-GluA)]-PCL for the controlled release of anticancer drugs for therapeutic applications. NANOTECHNOLOGY 2016; 27:125101. [PMID: 26891479 DOI: 10.1088/0957-4484/27/12/125101] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/05/2023]
Abstract
Herein, new nanoporous capsules of the block co-polymers of MeO-PEG-NH-(L-GluA)10 and polycaprolactone (PCL) have been synthesized through a surfactant-free cost-effective self-assembled soft-templating approach for the controlled release of drugs and for therapeutic applications. The nanoporous polymer capsules are designed to be biocompatible and are capable of encapsulating anticancer drugs (e.g., doxorubicin hydrochloride (DOX) and imatinib mesylate (ITM)) with a high extent (∼279 and ∼480 ng μg(-1), respectively). We have developed a nanoformulation of porous MeO-PEG-NH-(L-GluA)10-PCL capsules with DOX and ITM. The porous polymer nanoformulations have been programmed in terms of the release of anticancer drugs with a desired dose to treat the leukemia (K562) and human carcinoma cells (HepG2) in vitro and show promising IC50 values with a very high mortality of cancer cells (up to ∼96.6%). Our nanoformulation arrests the cell divisions due to 'cellular scenescence' and kills the cancer cells specifically. The present findings could enrich the effectiveness of idiosyncratic nanoporous polymer capsules for use in various other nanomedicinal and biomedical applications, such as for killing cancer cells, immune therapy, and gene delivery.
Collapse
Affiliation(s)
- Chander Amgoth
- School of Engineering Sciences and Technology, University of Hyderabad, Hyderabad-500046, India
| | | | | | | |
Collapse
|
56
|
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: 45] [Impact Index Per Article: 5.0] [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
|
57
|
Repanas A, Andriopoulou S, Glasmacher B. The significance of electrospinning as a method to create fibrous scaffolds for biomedical engineering and drug delivery applications. J Drug Deliv Sci Technol 2016. [DOI: 10.1016/j.jddst.2015.12.007] [Citation(s) in RCA: 67] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
|
58
|
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
|
59
|
Incorporating small molecules or biologics into nanofibers for optimized drug release: A review. Int J Pharm 2015; 494:516-30. [DOI: 10.1016/j.ijpharm.2015.08.054] [Citation(s) in RCA: 95] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2015] [Revised: 08/18/2015] [Accepted: 08/19/2015] [Indexed: 12/23/2022]
|
60
|
Efficient RNA delivery by integrin-targeted glutathione responsive polyethyleneimine capped gold nanorods. Acta Biomater 2015; 23:136-146. [PMID: 26026304 DOI: 10.1016/j.actbio.2015.05.028] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2015] [Revised: 04/29/2015] [Accepted: 05/22/2015] [Indexed: 11/22/2022]
Abstract
RNA interference (RNAi) mediated gene silencing holds significant promises in gene therapy. A major obstacle to efficient RNAi is the systemic delivery of the therapeutic RNAs into the cytoplasmon without being trapped in intracellular endo-/lyso-somes. Herein we report the development of a PEGylated, RGD peptide modified, and disulfide cross-linked short polyethylenimines (DSPEIs) functionalized gold nanorod (RDG) for targeted small hairpin (sh)RNA delivery. The RDG effectively condensed shRNAs into stable nanoparticles, allowing for highly specific targeting of model human brain cancer cells (U-87 MG-GFP) via the αvβ3 integrins-mediated endocytosis. The combined effects of endosomal escape (via the proton-sponge effect of the PEIs) and efficient cleavage of the disulfide-cross-linked DSPEIs by the high intracellular glutathione content triggered rapid cytoplasma shRNAs release resulting in excellent RNAi efficiency and low cytotoxicity. Furthermore, the high stability and prolonged blood circulation afforded by PEGylation allowed for highly effective, targeted tumor accumulation and internalization of the carriers, resulting in outstanding intra-tumor gene silencing efficiency in U-87 MG-GFP tumor bearing BALB/c mice. Combining the capabilities of both passive and active targeting, intracellular glutathione-triggered "off-on" release and endosomal escape, the RDG nanocarrier developed herein appears to be a highly promising non-viral vector for efficient RNAi.
Collapse
|
61
|
MicroRNA delivery for regenerative medicine. Adv Drug Deliv Rev 2015; 88:108-22. [PMID: 26024978 DOI: 10.1016/j.addr.2015.05.014] [Citation(s) in RCA: 110] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2015] [Revised: 05/13/2015] [Accepted: 05/21/2015] [Indexed: 12/26/2022]
Abstract
MicroRNA (miRNA) directs post-transcriptional regulation of a network of genes by targeting mRNA. Although relatively recent in development, many miRNAs direct differentiation of various stem cells including induced pluripotent stem cells (iPSCs), a major player in regenerative medicine. An effective and safe delivery of miRNA holds the key to translating miRNA technologies. Both viral and nonviral delivery systems have seen success in miRNA delivery, and each approach possesses advantages and disadvantages. A number of studies have demonstrated success in augmenting osteogenesis, improving cardiogenesis, and reducing fibrosis among many other tissue engineering applications. A scaffold-based approach with the possibility of local and sustained delivery of miRNA is particularly attractive since the physical cues provided by the scaffold may synergize with the biochemical cues induced by miRNA therapy. Herein, we first briefly cover the application of miRNA to direct stem cell fate via replacement and inhibition therapies, followed by the discussion of the promising viral and nonviral delivery systems. Next we present the unique advantages of a scaffold-based delivery in achieving lineage-specific differentiation and tissue development.
Collapse
|
62
|
Hill MC, Nguyen MK, Jeon O, Alsberg E. Spatial control of cell gene expression by siRNA gradients in biodegradable hydrogels. Adv Healthc Mater 2015; 4:714-22. [PMID: 25530099 PMCID: PMC4406766 DOI: 10.1002/adhm.201400458] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2014] [Revised: 11/22/2014] [Indexed: 11/10/2022]
Abstract
The extracellular environment exposes cells to numerous biochemical and physical signals that regulate their behavior. Strategies for generating continuous gradients of signals in biomaterials may allow for spatial control and patterning of cell behavior, and ultimately aid in the engineering of complex tissues. Short interfering RNA (siRNA) can regulate gene expression by silencing specific mRNA molecules post-transcriptionally, which may be valuable when presented in a continuous gradient for regenerative or therapeutic applications. Here, a biodegradable hydrogel system containing a gradient of siRNA is presented, and its capacity to regulate protein expression of encapsulated cells in a spatially continuous manner is demonstrated. Photocross-linkable dextran hydrogels containing a gradient of siRNA have been successfully fabricated using a dual-programmable syringe pump system, and differential gene silencing in incorporated cells that is sustained over time has been shown using green fluorescent protein as a reporter. This platform technology may be applied in tissue engineering to spatially control biologically relevant cellular processes.
Collapse
Affiliation(s)
- Michael C. Hill
- Department of Biomedical Engineering, Case Western Reserve University, 10900 Euclid Ave., Cleveland, Ohio 44106, USA
| | - Minh K. Nguyen
- Department of Biomedical Engineering, Case Western Reserve University, 10900 Euclid Ave., Cleveland, Ohio 44106, USA
| | - Oju Jeon
- Department of Biomedical Engineering, Case Western Reserve University, 10900 Euclid Ave., Cleveland, Ohio 44106, USA
| | - Eben Alsberg
- Department of Biomedical Engineering, Case Western Reserve University, 10900 Euclid Ave., Cleveland, Ohio 44106, USA. Department of Orthopaedic Surgery, Case Western Reserve University, 10900 Euclid Ave., Cleveland, Ohio 44106, USA. National Center for Regenerative Medicine, Division of General Medical Sciences, Case Western Reserve University, 10900 Euclid Ave., Cleveland, Ohio 44106, USA
| |
Collapse
|
63
|
Pelipenko J, Kocbek P, Kristl J. Critical attributes of nanofibers: Preparation, drug loading, and tissue regeneration. Int J Pharm 2015; 484:57-74. [DOI: 10.1016/j.ijpharm.2015.02.043] [Citation(s) in RCA: 154] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2014] [Revised: 02/16/2015] [Accepted: 02/16/2015] [Indexed: 12/13/2022]
|
64
|
Karthikeyan K, Krishnaswamy VR, Lakra R, Kiran MS, Korrapati PS. Fabrication of electrospun zein nanofibers for the sustained delivery of siRNA. JOURNAL OF MATERIALS SCIENCE. MATERIALS IN MEDICINE 2015; 26:101. [PMID: 25655500 DOI: 10.1007/s10856-015-5439-x] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/21/2014] [Accepted: 11/29/2014] [Indexed: 06/04/2023]
Abstract
In this study, zein nanofibers based siRNA delivery system has been attempted for the first time. Here, the amphiphilic property of zein and the size advantage of nanofibers have been brought together in developing an ideal delivery system for siRNA. The morphological analysis of the GAPDH-siRNA loaded zein nanofibers revealed the proper encapsulation of the siRNA in the polymeric matrix. The loading efficiency of this delivery system was found to be 58.57±2.4% (w/w). The agarose gel analysis revealed that the zein nanofibers preserved the integrity of siRNA for a longer period even at the room temperature. The in vitro release studies not only depicted the sustaining potential of the zein nanofibers but also ensured the release of sufficient quantity of siRNA required to induce the gene silencing effect. The amphiphilic property of zein supported the cell attachment and thereby facilitated the transfection of siRNA into the cells. qRT-PCR analysis confirmed the potential of the developed system in inducing the desired gene silencing effect. Thus, electrospun zein nanofibers have been successfully employed for the delivery of siRNA which has a great therapeutic potential.
Collapse
Affiliation(s)
- K Karthikeyan
- Biomaterials Department, CSIR-Central Leather Research Institute, Adyar, Chennai, 600020, India
| | | | | | | | | |
Collapse
|
65
|
Llorens E, Ibañez H, Del Valle LJ, Puiggalí J. Biocompatibility and drug release behavior of scaffolds prepared by coaxial electrospinning of poly(butylene succinate) and polyethylene glycol. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2015; 49:472-484. [PMID: 25686974 DOI: 10.1016/j.msec.2015.01.039] [Citation(s) in RCA: 73] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/18/2014] [Revised: 12/19/2014] [Accepted: 01/07/2015] [Indexed: 11/16/2022]
Abstract
Scaffolds constituted by electrospun microfibers of poly(ethylene glycol) (PEG) and poly(butylene succinate) (PBS) were studied. Specifically, coaxial microfibers having different core-shell distributions and compositions were considered as well as uniaxial micro/nanofibers prepared from mixtures of both polymers. Processing conditions were optimized for all geometries and compositions and resulting morphologies (i.e. diameter and surface texture) characterized by scanning electron microscopy. Chemical composition, molecular interactions and thermal properties were evaluated by FTIR, NMR, XPS and differential scanning calorimetry. The PEG component of electrospun fibers could be solubilized by immersion of scaffolds in aqueous medium, giving rise to high porosity and hydrophobic samples. Nevertheless, a small amount of PEG was retained in the PBS matrix, suggesting some degree of mixing. Solubilization was slightly dependent on fiber structure; specifically, the distribution of PEG in the core or shell of coaxial fibers led to higher or lower retention levels, respectively. Scaffolds could be effectively loaded with hydrophobic drugs having antibacterial and anticarcinogenic activities like triclosan and curcumin, respectively. Their release was highly dependent on their chemical structure and medium composition. Thus, low and high release rates were observed in phosphate buffer saline (SS) and SS/ethanol (30:70 v/v), respectively. Slight differences in the release of triclosan were found depending on fiber distribution and composition. Antibacterial activity and biocompatibility were evaluated for both loaded and unloaded scaffolds.
Collapse
Affiliation(s)
- E Llorens
- Departament d'Enginyeria Química, Universitat Politècnica de Catalunya, Av. Diagonal 647, Barcelona E-08028, Spain
| | - H Ibañez
- Departament d'Enginyeria Química, Universitat Politècnica de Catalunya, Av. Diagonal 647, Barcelona E-08028, Spain
| | - L J Del Valle
- Departament d'Enginyeria Química, Universitat Politècnica de Catalunya, Av. Diagonal 647, Barcelona E-08028, Spain.
| | - J Puiggalí
- Departament d'Enginyeria Química, Universitat Politècnica de Catalunya, Av. Diagonal 647, Barcelona E-08028, Spain; Center for Research in Nano-Engineering (CrNE), Universitat Politècnica de Catalunya, Edifici C, C/Pasqual i Vila s/n, Barcelona E-08028, Spain
| |
Collapse
|
66
|
Balaji A, Vellayappan MV, John AA, Subramanian AP, Jaganathan SK, Supriyanto E, Razak SIA. An insight on electrospun-nanofibers-inspired modern drug delivery system in the treatment of deadly cancers. RSC Adv 2015. [DOI: 10.1039/c5ra07595e] [Citation(s) in RCA: 69] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
This review gives an insight into the process of electrospinning, its essential parameters, the types of drug incorporation and the works reported on common dreadful cancers.
Collapse
Affiliation(s)
- A. Balaji
- IJN-UTM Cardiovascular Engineering Centre
- Faculty of Biosciences and Medical Engineering
- Universiti Teknologi Malaysia
- Johor Bahru 81310
- Malaysia
| | - M. V. Vellayappan
- IJN-UTM Cardiovascular Engineering Centre
- Faculty of Biosciences and Medical Engineering
- Universiti Teknologi Malaysia
- Johor Bahru 81310
- Malaysia
| | - A. A. John
- IJN-UTM Cardiovascular Engineering Centre
- Faculty of Biosciences and Medical Engineering
- Universiti Teknologi Malaysia
- Johor Bahru 81310
- Malaysia
| | - A. P. Subramanian
- IJN-UTM Cardiovascular Engineering Centre
- Faculty of Biosciences and Medical Engineering
- Universiti Teknologi Malaysia
- Johor Bahru 81310
- Malaysia
| | - S. K. Jaganathan
- IJN-UTM Cardiovascular Engineering Centre
- Faculty of Biosciences and Medical Engineering
- Universiti Teknologi Malaysia
- Johor Bahru 81310
- Malaysia
| | - Eko Supriyanto
- IJN-UTM Cardiovascular Engineering Centre
- Faculty of Biosciences and Medical Engineering
- Universiti Teknologi Malaysia
- Johor Bahru 81310
- Malaysia
| | - S. I. A. Razak
- IJN-UTM Cardiovascular Engineering Centre
- Faculty of Biosciences and Medical Engineering
- Universiti Teknologi Malaysia
- Johor Bahru 81310
- Malaysia
| |
Collapse
|
67
|
Li YF, Rubert M, Yu Y, Besenbacher F, Chen M. Delivery of dexamethasone from electrospun PCL–PEO binary fibers and their effects on inflammation regulation. RSC Adv 2015. [DOI: 10.1039/c5ra03099d] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Differences in surface topography, chemical composition, wettability and release kinetics of the anti-inflammatory drug dexamethasone among different PCL–PEO fibers collectively affected the regulation of inflammatory related gene expression.
Collapse
Affiliation(s)
- Yan-Fang Li
- Interdisciplinary Nanoscience Center (iNANO)
- Aarhus University
- DK-8000 Aarhus C
- Denmark
- Institute of Nanoscience and Nanotechnology
| | - Marina Rubert
- Interdisciplinary Nanoscience Center (iNANO)
- Aarhus University
- DK-8000 Aarhus C
- Denmark
| | - Ying Yu
- Institute of Nanoscience and Nanotechnology
- Central China Normal University
- Wuhan 430079
- China
| | - Flemming Besenbacher
- Interdisciplinary Nanoscience Center (iNANO)
- Aarhus University
- DK-8000 Aarhus C
- Denmark
| | - Menglin Chen
- Interdisciplinary Nanoscience Center (iNANO)
- Aarhus University
- DK-8000 Aarhus C
- Denmark
| |
Collapse
|
68
|
Kumar K, Kumar Doddi S, Kalle Arunasree M, Paik P. CPMV-induced synthesis of hollow mesoporous SiO2 nanocapsules with excellent performance in drug delivery. Dalton Trans 2015; 44:4308-17. [DOI: 10.1039/c4dt02549k] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Synthesis of CPMV- hollow silica nanocapsules and their use in nanomedicine.
Collapse
Affiliation(s)
- Koushi Kumar
- School of Engineering Sciences and Technology
- University of Hyderabad
- India
| | | | | | - Pradip Paik
- School of Engineering Sciences and Technology
- University of Hyderabad
- India
- Advanced Research Centre for High Energy Materials
- University of Hyderabad
| |
Collapse
|
69
|
Lee S, Jin G, Jang JH. Electrospun nanofibers as versatile interfaces for efficient gene delivery. J Biol Eng 2014; 8:30. [PMID: 25926887 PMCID: PMC4414388 DOI: 10.1186/1754-1611-8-30] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2014] [Accepted: 11/27/2014] [Indexed: 12/31/2022] Open
Abstract
The integration of gene delivery technologies with electrospun nanofibers is a versatile strategy to increase the potential of gene therapy as a key platform technology that can be readily utilized for numerous biomedical applications, including cancer therapy, stem cell therapy, and tissue engineering. As a spatial template for gene delivery, electrospun nanofibers possess highly advantageous characteristics, such as their ease of production, their ECM-analogue nature, the broad range of choices for materials, the feasibility of producing structures with varied physical and chemical properties, and their large surface-to-volume ratios. Thus, electrospun fiber-mediated gene delivery exhibits a great capacity to modulate the spatial and temporal release kinetics of gene vectors and enhance gene delivery efficiency. This review discusses the powerful characteristics of electrospun nanofibers, which can function as spatial interfaces capable of promoting controlled and efficient gene delivery.
Collapse
Affiliation(s)
- Slgirim Lee
- Department of Chemical and Biomolecular Engineering, Yonsei University, Seoul, 120-749 Korea
| | - Gyuhyung Jin
- Department of Chemical and Biomolecular Engineering, Yonsei University, Seoul, 120-749 Korea
| | - Jae-Hyung Jang
- Department of Chemical and Biomolecular Engineering, Yonsei University, Seoul, 120-749 Korea
| |
Collapse
|
70
|
James EN, Delany AM, Nair LS. Post-transcriptional regulation in osteoblasts using localized delivery of miR-29a inhibitor from nanofibers to enhance extracellular matrix deposition. Acta Biomater 2014; 10:3571-80. [PMID: 24816265 DOI: 10.1016/j.actbio.2014.04.026] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2014] [Revised: 03/31/2014] [Accepted: 04/24/2014] [Indexed: 02/03/2023]
Abstract
MicroRNAs are important post-transcriptional regulators of skeletal biology, and miRNA-based therapeutics have the potential to aid bone repair. However, efficient tools for delivering miRNA mimics or inhibitors to specific target tissues are limited. Polymeric nanofibers closely mimic natural extracellular matrix (ECM) morphology, and are attractive candidates for supporting delivery of cells and bone-anabolic reagents. It is hypothesized that gelatin nanofibers could be used for the localized transient delivery of miRNA-based therapeutics, using miR-29a inhibitor as a prototype to increase ECM deposition. miR-29 family members are negative regulators of ECM synthesis, targeting the mRNAs of selected collagens and osteonectin/SPARC. Inhibiting miR-29 activity may therefore increase ECM production by cells. miR-29a inhibitor-loaded gelatin nanofibers, prepared by electrospinning, demonstrated continuous release of miRNA inhibitor over 72h. Pre-osteoblastic murine MC3T3-E1 cell line seeded on miR-29a inhibitor-loaded nanofibers synthesized more osteonectin, indicating efficient inhibitor delivery. These cells also displayed increased Igf1 and Tgfb1 mRNA. Moreover, primary bone marrow stromal cells from transgenic pOBCol3.6cyan reporter mice, grown on miR-29a inhibitor scaffolds, displayed increased col3.6 cyan expression as well as collagen production. This study demonstrates that ECM mimicking nanostructured scaffolds, in conjunction with bioactive miRNA-based therapeutics, may serve as a novel platform for developing biologically active localized cell delivery systems.
Collapse
|
71
|
Eatemadi A, Daraee H, Zarghami N, Melat Yar H, Akbarzadeh A. Nanofiber: Synthesis and biomedical applications. ARTIFICIAL CELLS NANOMEDICINE AND BIOTECHNOLOGY 2014; 44:111-21. [DOI: 10.3109/21691401.2014.922568] [Citation(s) in RCA: 114] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
|
72
|
Sustained localized presentation of RNA interfering molecules from in situ forming hydrogels to guide stem cell osteogenic differentiation. Biomaterials 2014; 35:6278-6286. [PMID: 24831973 DOI: 10.1016/j.biomaterials.2014.04.048] [Citation(s) in RCA: 112] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2014] [Accepted: 04/13/2014] [Indexed: 02/07/2023]
Abstract
To date, RNA interfering molecules have been used to differentiate stem cells on two-dimensional (2D) substrates that do not mimic three-dimensional (3D) microenvironments in the body. Here, in situ forming poly(ethylene glycol) (PEG) hydrogels were engineered for controlled, localized and sustained delivery of RNA interfering molecules to differentiate stem cells encapsulated within the 3D polymer network. RNA interfering molecules were released from the hydrogels in a sustained and controlled manner over the course of 3-6 weeks, and exhibited high bioactivity. Importantly, it was demonstrated that the delivery of siRNA and/or miRNA from the hydrogel constructs enhanced the osteogenic differentiation of encapsulated stem cells. Prolonged delivery of siRNA and/or miRNA from this polymeric scaffold permitted extended regulation of cell behavior, unlike traditional siRNA experiments performed in vitro. This approach presents a powerful new methodology for controlling cell fate, and is promising for multiple applications in tissue engineering and regenerative medicine.
Collapse
|
73
|
Hu X, Liu S, Zhou G, Huang Y, Xie Z, Jing X. Electrospinning of polymeric nanofibers for drug delivery applications. J Control Release 2014; 185:12-21. [PMID: 24768792 DOI: 10.1016/j.jconrel.2014.04.018] [Citation(s) in RCA: 709] [Impact Index Per Article: 64.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2014] [Revised: 03/21/2014] [Accepted: 04/10/2014] [Indexed: 01/19/2023]
Abstract
Electrospinning has been recognized as a simple and versatile method for fabrication of polymer nanofibers. Various polymers that include synthetic, natural, and hybrid materials have been successfully electrospun into ultrafine fibers. The inherently high surface to volume ratio of electrospun fibers can enhance cell attachment, drug loading, and mass transfer properties. Drugs ranging from antibiotics and anticancer agents to proteins, DNA, RNA, living cells, and various growth factors have been incorporated into electrospun fibers. This article presents an overview of electrospinning techniques and their application in drug delivery.
Collapse
Affiliation(s)
- Xiuli Hu
- State Key Laboratory of Polymer Physics and Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, PR China
| | - Shi Liu
- State Key Laboratory of Polymer Physics and Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, PR China; The University of Chinese Academy of Sciences, Beijing 100049, PR China
| | - Guangyuan Zhou
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, PR China
| | - Yubin Huang
- State Key Laboratory of Polymer Physics and Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, PR China.
| | - Zhigang Xie
- State Key Laboratory of Polymer Physics and Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, PR China.
| | - Xiabin Jing
- State Key Laboratory of Polymer Physics and Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, PR China
| |
Collapse
|
74
|
Monteiro N, Martins A, Pires R, Faria S, Fonseca NA, Moreira JN, Reis RL, Neves NM. Immobilization of bioactive factor-loaded liposomes on the surface of electrospun nanofibers targeting tissue engineering. Biomater Sci 2014; 2:1195-1209. [DOI: 10.1039/c4bm00069b] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Dexamethasone-loaded liposomes immobilized at the surface of electrospun polycaprolactone nanofiber meshes successfully promoted the osteogenic differentiation of human bone marrow-derived mesenchymal stem cells.
Collapse
Affiliation(s)
- Nelson Monteiro
- 3B's Research Group – Biomaterials
- Biodegradables and Biomimetics
- Department of Polymer Engineering
- University of Minho
- Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine
| | - Albino Martins
- 3B's Research Group – Biomaterials
- Biodegradables and Biomimetics
- Department of Polymer Engineering
- University of Minho
- Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine
| | - Ricardo Pires
- 3B's Research Group – Biomaterials
- Biodegradables and Biomimetics
- Department of Polymer Engineering
- University of Minho
- Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine
| | - Susana Faria
- Department of Mathematics for Science and Technology
- Research CMAT
- University of Minho
- 4800-058 Guimarães, Portugal
| | - Nuno A. Fonseca
- CNC – Center for Neurosciences and Cell Biology
- FFUC – Faculty of Pharmacy of the University of Coimbra
- 3000 Coimbra, Portugal
| | - João N. Moreira
- CNC – Center for Neurosciences and Cell Biology
- FFUC – Faculty of Pharmacy of the University of Coimbra
- 3000 Coimbra, Portugal
| | - Rui L. Reis
- 3B's Research Group – Biomaterials
- Biodegradables and Biomimetics
- Department of Polymer Engineering
- University of Minho
- Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine
| | - Nuno M. Neves
- 3B's Research Group – Biomaterials
- Biodegradables and Biomimetics
- Department of Polymer Engineering
- University of Minho
- Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine
| |
Collapse
|
75
|
Yin L, Zhao X, Ji S, He C, Wang G, Tang C, Gu S, Yin C. The use of gene activated matrix to mediate effective SMAD2 gene silencing against hypertrophic scar. Biomaterials 2013; 35:2488-98. [PMID: 24388384 DOI: 10.1016/j.biomaterials.2013.12.015] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2013] [Accepted: 12/08/2013] [Indexed: 10/25/2022]
Abstract
Hypertrophic scar (HS) originates from the over-expression of transforming growth factor β (TGF-β) and downstream SMAD2. With attempts to rectify HS by RNA interference (RNAi) against SMAD2, we report the design of plasmid DNA encoding SMAD2 siRNA (pSUPER-SMAD2), and identify the optimal siRNA sequence toward maximal RNAi efficiency. To realize effective and sustained RNAi, we developed gene activated matrix (GAM) based on porous atelocollagen scaffold and embedded trimethyl chitosan-cysteine (TMCC)/pSUPER-SMAD2 polyplexes for promoting cell growth and gene transfection. The GAM exhibited porosity higher than 80%, pore size of 200-250 μm, desired mechanical strength, and sustained pSUPER-SMAD2 release profiles. Normal skin fibroblasts (NSFs) and hypertrophic scar fibroblasts (HSFs) were allowed to infiltrate and proliferate in GAM; at the meantime they were transfected with TMCC/pSUPER-SMAD2 polyplexes to display remarkably reduced SMAD2 levels that lasted for up to 10 days, consequently inhibiting the over-production of type I and type III collagen. We further unraveled the notably higher transfection levels of GAM in three-dimensional (3D) than in 2D environment, which was attributed to the improved cell-matrix interactions that promote cell proliferation and polyplex internalization. This highly safe and effective GAM may serve as a promising candidate towards HS treatment.
Collapse
Affiliation(s)
- Lichen Yin
- State Key Laboratory of Genetic Engineering, Department of Pharmaceutical Sciences, School of Life Sciences, Fudan University, Shanghai 200433, China
| | - Xin Zhao
- State Key Laboratory of Genetic Engineering, Department of Pharmaceutical Sciences, School of Life Sciences, Fudan University, Shanghai 200433, China
| | - Shizhao Ji
- Center of Burns and Traumatic Surgery, Changhai Hospital, Second Military Medical University, Shanghai 200433, China
| | - Chunbai He
- State Key Laboratory of Genetic Engineering, Department of Pharmaceutical Sciences, School of Life Sciences, Fudan University, Shanghai 200433, China
| | - Guangyi Wang
- Center of Burns and Traumatic Surgery, Changhai Hospital, Second Military Medical 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.
| | - Shaohua Gu
- 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
|
76
|
Blakney AK, Ball C, Krogstad EA, Woodrow KA. Electrospun fibers for vaginal anti-HIV drug delivery. Antiviral Res 2013; 100 Suppl:S9-16. [PMID: 24188701 DOI: 10.1016/j.antiviral.2013.09.022] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2013] [Revised: 09/19/2013] [Accepted: 09/26/2013] [Indexed: 01/11/2023]
Abstract
Diversity of microbicide delivery systems is essential for future success in the prevention and treatment of HIV in order to account for the varied populations of women all over the world that may benefit from use of these products. Recently, a novel dosage form for intravaginal drug delivery has been developed using drug-eluting fibers fabricated by electrospinning. There is a strong rationale to support the idea that drug-eluting fibers can be designed to realize multiple design constraints in a single product for topical HIV prevention: fibers are able to deliver a wide range of agents, incorporate multiple agents via composites, and facilitate controlled release over relevant time frames for pericoital and sustained (coitally-independent) use. It is also technologically feasible to scale-up production of fiber-based microbicides. Electrospun fibers may allow for prioritization of physical attributes that affect user perceptions without compromising biological efficacy. Challenges with using fibers as a microbicide include issues related to vehicle deployment, spreading and retention in the vaginal vault. In addition, studies will need to address the interaction of the fibers with the mucosal environment, including unknown safety and toxicity. Sustained release fiber microbicides capable of delivering multiple antiretroviral drugs while simultaneously exhibiting tunable degradation or dissolution of the fibers is also a challenge. However, electrospun fibers are a promising new platform for vaginal delivery of anti-HIV agents and future research will inform their place in the field. This article is based on a presentation at the "Product Development Workshop 2013: HIV and Multipurpose Prevention Technologies", held in Arlington, Virginia on February 20-21, 2013. It forms part of a special supplement to Antiviral Research.
Collapse
Affiliation(s)
- Anna K Blakney
- Department of Bioengineering, University of Washington, Seattle, Washington, United States
| | | | | | | |
Collapse
|
77
|
Zamani M, Prabhakaran MP, Ramakrishna S. Advances in drug delivery via electrospun and electrosprayed nanomaterials. Int J Nanomedicine 2013; 8:2997-3017. [PMID: 23976851 PMCID: PMC3746732 DOI: 10.2147/ijn.s43575] [Citation(s) in RCA: 126] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023] Open
Abstract
Electrohydrodynamic (EHD) techniques refer to procedures that utilize electrostatic forces to fabricate fibers or particles of different shapes with sizes in the nano-range to a few microns through electrically charged fluid jet. Employing different techniques, such as blending, surface modification, and coaxial process, there is a great possibility of incorporating bioactive such molecules as drugs, DNA, and growth factors into the nanostructures fabricated via EHD techniques. By careful selection of materials and processing conditions, desired encapsulation efficiency as well as preserved bioactivity of the therapeutic agents can be achieved. The drug-loaded nanostructures produced can be applied via different routes, such as implantation, injection, and topical or oral administration for a wide range of disease treatment. Taking advantage of the recent developments in EHD techniques like the coaxial process or multilayered structures, individually controlled delivery of multiple drugs is achievable, which is of great demand in cancer therapy and growth-factor delivery. This review summarizes the most recent techniques and postmodification methods to fabricate electrospun nanofibers and electrosprayed particles for drug-delivery applications.
Collapse
Affiliation(s)
- Maedeh Zamani
- Department of Mechanical Engineering, National University of Singapore, Singapore
| | | | | |
Collapse
|
78
|
Buzgo M, Jakubova R, Mickova A, Rampichova M, Prosecka E, Kochova P, Lukas D, Amler E. Time-regulated drug delivery system based on coaxially incorporated platelet α-granules for biomedical use. Nanomedicine (Lond) 2013. [DOI: 10.2217/nnm.12.140] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Aim: Platelet derivatives serve as an efficient source of natural growth factors. In the current study, α-granules were incorporated into coaxial nanofibers. Materials & methods: A nanofiber scaffold containing α-granules was prepared by coaxial electrospinning. The biological potential of the nanofiber scaffold was evaluated in chondrocyte and mesenchymal stem cell cultivation studies. Additionally, the concentration of TGF-β1 was determined. Results: Microscopy studies showed that intact α-granules were incorporated into the coaxial nanofibers. The cultivation tests showed that the novel scaffold stimulated viability and extracellular matrix production of chondrocytes and mesenchymal stem cells. In addition, the concentration of growth factors necessary for the induction of cell proliferation significantly decreased. Conclusion: The system preserved α-granule bioactivity and stimulated cell viability and chondrogenic differentiation of mesenchymal stem cells. Core/shell nanofibers incorporating α-granules are a promising system for tissue engineering, particularly cartilage engineering. Original submitted 21 March 2012; Revised submitted 8 August 2012; Published online 2 December 2012
Collapse
Affiliation(s)
- Matej Buzgo
- Institute of Experimental Medicine, Academy of Sciences of the Czech Republic, v.v.i, Vídeňská 1083, 142 20 Prague 4, Czech Republic
- Department of Biophysics, 2nd Faculty of Medicine, Charles University in Prague, V Úvalu 84, 150 06 Prague 5, Czech Republic.
| | - Radka Jakubova
- Department of Biophysics, 2nd Faculty of Medicine, Charles University in Prague, V Úvalu 84, 150 06 Prague 5, Czech Republic
- Institute of Experimental Medicine, Academy of Sciences of the Czech Republic, v.v.i, Vídeňská 1083, 142 20 Prague 4, Czech Republic
| | - Andrea Mickova
- Department of Biophysics, 2nd Faculty of Medicine, Charles University in Prague, V Úvalu 84, 150 06 Prague 5, Czech Republic
- Institute of Experimental Medicine, Academy of Sciences of the Czech Republic, v.v.i, Vídeňská 1083, 142 20 Prague 4, Czech Republic
| | - Michala Rampichova
- Department of Biophysics, 2nd Faculty of Medicine, Charles University in Prague, V Úvalu 84, 150 06 Prague 5, Czech Republic
- Institute of Experimental Medicine, Academy of Sciences of the Czech Republic, v.v.i, Vídeňská 1083, 142 20 Prague 4, Czech Republic
| | - Eva Prosecka
- Department of Biophysics, 2nd Faculty of Medicine, Charles University in Prague, V Úvalu 84, 150 06 Prague 5, Czech Republic
- Institute of Experimental Medicine, Academy of Sciences of the Czech Republic, v.v.i, Vídeňská 1083, 142 20 Prague 4, Czech Republic
| | - Petra Kochova
- Department of Mechanics, Faculty of Applied Sciences, University of West Bohemia, Univerzitni 8, 30614 Pilsen, Czech Republic
| | - David Lukas
- Department of Nonwovens, Technical University of Liberec, Studentska 2, 461 17 Liberec, Czech Republic
| | - Evžen Amler
- Department of Biophysics, 2nd Faculty of Medicine, Charles University in Prague, V Úvalu 84, 150 06 Prague 5, Czech Republic
- Institute of Experimental Medicine, Academy of Sciences of the Czech Republic, v.v.i, Vídeňská 1083, 142 20 Prague 4, Czech Republic
| |
Collapse
|
79
|
Gabriel D, Dvir T, Kohane DS. Delivering bioactive molecules as instructive cues to engineered tissues. Expert Opin Drug Deliv 2013; 9:473-92. [PMID: 22432691 DOI: 10.1517/17425247.2012.668521] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
INTRODUCTION Growth factors and other bioactive molecules play a crucial role in the creation of functional engineered tissues from dissociated cells. AREAS COVERED This review discusses the delivery of bioactive molecules - particularly growth factors - to affect cellular function in the context of tissue engineering. We discuss the primary biological themes that are addressed by delivering bioactives, the types of molecules that are to be delivered, the major materials used in producing scaffolds and/or drug delivery systems, and the principal drug delivery strategies. EXPERT OPINION Drug delivery systems have allowed the sustained release of bioactive molecules to engineered tissues, with marked effects on tissue function. Sophisticated drug delivery techniques will allow precise recapitulation of developmental milestones by providing temporally distinct patterns of release of multiple bioactives. High-resolution patterning techniques will allow tissue constructs to be designed with precisely defined areas where bioactives can act. New biological discoveries, just as the development of small molecules with potent effects on cell differentiation, will likely have a marked impact on the field.
Collapse
Affiliation(s)
- Doris Gabriel
- Children's Hospital Boston, Harvard Medical School, Division of Critical Care Medicine, Department of Anesthesiology, Laboratory for Biomaterials and Drug Delivery, 300 Longwood Avenue, Boston, MA 02115, USA
| | | | | |
Collapse
|
80
|
Low WC, Rujitanaroj PO, Lee DK, Messersmith PB, Stanton LW, Goh E, Chew SY. Nanofibrous scaffold-mediated REST knockdown to enhance neuronal differentiation of stem cells. Biomaterials 2013; 34:3581-90. [PMID: 23415645 DOI: 10.1016/j.biomaterials.2013.01.093] [Citation(s) in RCA: 72] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2013] [Accepted: 01/26/2013] [Indexed: 02/06/2023]
Abstract
At present, the recovery prospect for patients with chronic neurodegenerative diseases or acute trauma in the central nervous system is sub-optimal. The controlled differentiation of neural stem/progenitor cells (NPCs) to functional neurons is a possible treatment strategy. In contrast to the classical approach of biochemicals supplementation for guided stem cell commitment, this study explores the feasibility of directing neuronal differentiation through synergistic integration of three-dimensional nanofibrous topographical cues and scaffold-mediated knockdown of RE-1 silencing transcription factor (REST) in mouse NPCs. Taking advantage of the strong adhesive property and latent reactivity of mussel-inspired polydopamine (PD) coating, electrospun polycaprolactone (PCL) nanofibers were successfully functionalized with REST siRNAs (denoted as siREST PD-fiber). Sustained REST knockdown in NPCs was achieved for up to five days in vitro and the silencing efficiency was significantly higher than that mediated through siRNA adsorption onto non-PD coated sample controls. The silencing of REST, together with nanofiber topographical effect, significantly enhanced NPC neuronal commitment (57.5% Map2(+) cells in siREST PD-fiber vs. 43.5% in siREST PD-film vs. 50% in PD-fiber controls, p < 0.05) while reducing astrocytic and oligodendrocytic differentiation (10.7% O4(+) cells vs. ∼30% in siREST PD-film, p < 0.01). Taken together, the synergistic effects of scaffold-mediated REST knockdown and topographical cues from PD-modified nanofibers may be a useful strategy for generating functional neurons for therapeutic purposes.
Collapse
Affiliation(s)
- Wei Ching Low
- Division of Chemical and Biomolecular Engineering, School of Chemical and Biomedical Engineering, Nanyang Technological University, Singapore, Singapore
| | | | | | | | | | | | | |
Collapse
|
81
|
Abstract
Microbicides are topically applied, user controlled dosage forms that are being developed to prevent the transmission of HIV during coitus. Early candidates focused on coitally dependent dosage forms such as gels and creams. More recent development has focused on broadening the coitally dependent options through the introduction of films and fast dissolving tablets. Additionally, it has become important to have longer acting products to minimize the burden of user compliance and thus vaginal rings have been developed providing sustained delivery of antiretroviral drugs. This chapter discusses the history of microbicides along with a detailed description of coitally dependent products, gels, films, tablets diaphragms, as well as coitally independent dosage forms such as vaginal rings and the introduction of a new technology, electrospun fibers.
Collapse
Affiliation(s)
- L C Rohan
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh Magee Women's Research Institute, Pittsburgh, PA, USA,
| | | | | |
Collapse
|
82
|
Andersen MØ, Dillschneider P, Kjems J. The Role of MicroRNAs in Natural Tissue Development and Application in Regenerative Medicine. ADVANCES IN DELIVERY SCIENCE AND TECHNOLOGY 2013. [DOI: 10.1007/978-1-4614-4744-3_3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
83
|
Rujitanaroj PO, Jao B, Yang J, Wang F, Anderson JM, Wang J, Chew SY. Controlling fibrous capsule formation through long-term down-regulation of collagen type I (COL1A1) expression by nanofiber-mediated siRNA gene silencing. Acta Biomater 2013; 9:4513-24. [PMID: 23036951 PMCID: PMC3523808 DOI: 10.1016/j.actbio.2012.09.029] [Citation(s) in RCA: 68] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2012] [Revised: 09/22/2012] [Accepted: 09/24/2012] [Indexed: 10/27/2022]
Abstract
The foreign body reaction often interferes with the long-term functionality and performance of implanted biomedical devices through fibrous capsule formation. While many implant modification techniques have been adopted in attempts to control fibrous encapsulation, the outcomes remained sub-optimal. Nanofiber scaffold-mediated RNA interference may serve as an alternative approach through the localized and sustained delivery of siRNA at implant sites. In this study, we investigated the efficacy of siRNA-poly(caprolactone-co-ethylethylene phosphate) nanofibers in controlling fibrous capsule formation through the down-regulation of collagen type I (COL1A1) in vitro and in vivo. By encapsulating complexes of COL1A1 siRNA with a transfection reagent (Transit TKO) or the cell penetrating peptides CADY or MPG within the nanofibers (550-650 nm in diameter), a sustained release of siRNA was obtained for at least 28 days (loading efficiency ~60-67%). Scaffold-mediated transfection significantly enhanced cellular uptake of oligonucleotides and prolonged in vitro gene silencing duration by at least 2-3 times as compared to conventional bolus delivery of siRNA (14 days vs. 5-7 days by bolus delivery). In vivo subcutaneous implantation of siRNA scaffolds revealed a significant decrease in fibrous capsule thickness at weeks 2 and 4 as compared to plain nanofibers (p<0.05). Taken together, the results demonstrated the efficacy of scaffold-mediated siRNA gene-silencing in providing effective long-term control of fibrous capsule formation.
Collapse
Affiliation(s)
- Pim-on Rujitanaroj
- School of Chemical and Biomedical Engineering, Nanyang Technological University, 637459, Singapore
| | - Brian Jao
- Department of Pathology, Case Western Reserve University, Cleveland, Ohio 44106, USA
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, Ohio 44106, USA
| | - Junghoon Yang
- Department of Pathology, Case Western Reserve University, Cleveland, Ohio 44106, USA
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, Ohio 44106, USA
| | - Feng Wang
- Hefei National Laboratory for Physical Sciences at the Microscale and School of Life Sciences, University of Science and Technology of China, Hefei, Anhui 230027, P. R. China
| | - James M. Anderson
- Department of Pathology, Case Western Reserve University, Cleveland, Ohio 44106, USA
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, Ohio 44106, USA
| | - Jun Wang
- Hefei National Laboratory for Physical Sciences at the Microscale and School of Life Sciences, University of Science and Technology of China, Hefei, Anhui 230027, P. R. China
| | - Sing Yian Chew
- School of Chemical and Biomedical Engineering, Nanyang Technological University, 637459, Singapore
| |
Collapse
|
84
|
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
|
85
|
Achille C, Sundaresh S, Chu B, Hadjiargyrou M. Cdk2 silencing via a DNA/PCL electrospun scaffold suppresses proliferation and increases death of breast cancer cells. PLoS One 2012; 7:e52356. [PMID: 23285007 PMCID: PMC3527537 DOI: 10.1371/journal.pone.0052356] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2012] [Accepted: 11/13/2012] [Indexed: 11/18/2022] Open
Abstract
RNA interference (RNAi) is a promising approach for cancer treatment. Site specific and controlled delivery of RNAi could be beneficial to the patient, while at the same time reducing undesirable off-target side effects. We utilized electrospinning to generate a biodegradable scaffold capable of incorporating and delivering a bioactive plasmid encoding for short hairpin (sh) RNA against the cell cycle specific protein, Cdk2. Three electrospun scaffolds were constructed, one using polycaprolactone (PCL) alone (Control) and PCL with plasmid DNA encoding for either Cdk2 (Cdk2i) and EGFP (EGFPi, also served as a control) shRNA. Scaffold fiber diameters ranged from 1 to 20 µm (DNA containing) and 0.2-3 µm (Control). While the electrospun fibers remained intact for more than two weeks in physiological buffer, degradation was visible during the third week of incubation. Approximately 20-60 ng/ml (~2.5% cumulative release) of intact and bioactive plasmid DNA was released over 21 days. Further, Cdk2 mRNA expression in cells plated on the Cdk2i scaffold was decreased by ~51% and 30%, in comparison with that of cells plated on Control or EGFPi scaffold, respectively. This decrease in Cdk2 mRNA by the Cdk2i scaffold translated to a ~40% decrease in the proliferation of the breast cancer cell line, MCF-7, as well as the presence of increased number of dead cells. Taken together, these results represent the first successful demonstration of the delivery of bioactive RNAi-based plasmid DNA from an electrospun polymer scaffold, specifically, in disrupting cell cycle regulation and suppressing proliferation of cancer cells.
Collapse
Affiliation(s)
- Clément Achille
- Institut Supérieur des Biosciences de Paris, Université de Paris Est Créteil, Créteil, France
| | - Sowmya Sundaresh
- Department of Biomedical Engineering, Stony Brook University, Stony Brook, New York, United States of America
| | - Benjamin Chu
- Department of Chemistry, Stony Brook University, Stony Brook, New York, United States of America
| | - Michael Hadjiargyrou
- Department of Life Sciences, New York Institute of Technology, Old Westbury, New York, United States of America
- * E-mail:
| |
Collapse
|
86
|
Benoit DSW, Boutin ME. Controlling mesenchymal stem cell gene expression using polymer-mediated delivery of siRNA. Biomacromolecules 2012; 13:3841-9. [PMID: 23020123 DOI: 10.1021/bm301294n] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
siRNA treatment has great promise to specifically control gene expression and select cell behaviors but has delivery challenges limiting its use. Particularly for applications in regenerative medicine, uniform and consistent delivery of siRNA to control gene expression and subsequent stem cell functions, such as differentiation, is paramount. Therefore, a diblock copolymer was examined for its ability to effectively deliver siRNA to mesenchymal stem cells (MSCs). The diblock copolymers, which are composed of cationic blocks for siRNA complexation, protection, and uptake and pH-responsive blocks for endosomal escape, were shown to facilitate nearly 100% MSC uptake of siRNA. This is vastly superior to a commercially available control, DharmaFECT, which resulted in only ~60% siRNA positive MSCs. Moreover, the diblock copolymer, at conditions that result in excellent knockdown (down to ~10% of control gene expression), was cytocompatible, causing no negative effects on MSC survivability. In contrast, DharmaFECT/siRNA treatment resulted in only ~60% survivability of MSCs. Longitudinal knockdown after siRNA treatment was examined and protein knockdown persists for ~6 days regardless of delivery system (diblock copolymer or DharmaFECT). Finally, MSC phenotype and differentiation capacity was examined after treatment with control siRNA. There was no statistically significant differences on cell surface markers of diblock copolymer/siRNA or DharmaFECT/siRNA-treated or cells measured 2 weeks after siRNA delivery compared to untreated cells. Upon differentiation with typical media/culture conditions to adipogenic, chondrogenic, and osteogenic lineages and examination of histological staining markers, there was no discernible differences between treated and untreated cells, regardless of delivery mechanism. Thus, diblock copolymers examined herein facilitated uniform siRNA treatment of MSCs, inducing siRNA-specific gene and protein knockdown without adversely affecting MSC survival or differentiation capacity and therefore show great promise for use within regenerative medicine applications.
Collapse
Affiliation(s)
- Danielle S W Benoit
- Department of Biomedical Engineering, University of Rochester Medical Center, University of Rochester, Rochester, NY 14627, USA.
| | | |
Collapse
|
87
|
Low WC, Yau WWY, Stanton LW, Marcy G, Goh E, Chew SY. Directing neuronal differentiation of primary neural progenitor cells by gene knockdown approach. DNA Cell Biol 2012; 31:1148-60. [PMID: 22339269 PMCID: PMC3391493 DOI: 10.1089/dna.2011.1557] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2011] [Revised: 01/10/2012] [Accepted: 01/12/2012] [Indexed: 01/07/2023] Open
Abstract
Directing differentiation of neural stem/progenitor cells (NPCs) to produce functional neurons is a promising remedy for neural pathological conditions. The major challenge, however, lies in the effective and efficient generation of a sizable population of neurons. A potential strategy is to incorporate RNA interference (RNAi) during directed stem cell differentiation to recapitulate the complex cell-signaling cascades that often occurs during the process. In this study, in vitro silencing of RE1-silencing transcription factor (REST) was carried out using small-interfering RNAs (siRNAs) to evaluate the efficacy of combining REST knockdown with conventional differentiation approaches to enhance neurogenesis. While earlier studies have demonstrated enhanced neuronal lineage commitment from embryonic stem cells and mesenchymal stem cells upon REST knockdown, the effects of REST silencing during other stages of neural development have not been extensively evaluated. We hypothesize that REST knockdown would enhance NPC development to mature neurons and that induced REST silencing can serve as a potential biochemical approach to direct cell fate. Under nonspecific induction conditions, REST knockdown induced eightfold higher Tuj1 mRNA expression at day 14 compared with untransfected cells and cells subjected to scrambled-siRNA treatment (controls). Immunostaining also revealed greater percentage of Tuj1 positive cells with REST knockdown. Combined with neuronal induction, REST silencing enhanced the kinetics of neuronal differentiation and the rate of maturation of committed neuronal cells. Specifically, upregulation of MAP2 occurred as early as 3 days after induction with REST silencing and the expression was comparable to the controls at day 14. Likewise, downregulation of REST generated more than twice the percentage of Tuj1 and MAP2 positive cells compared with controls at day 5 (p<0.05). Morphologically, REST-silencing enhanced the number and length of neurite extensions from Tuj1 positive cells (p<0.05), which was not evaluated in previous differentiation studies with REST knockdown. Taken together, these results demonstrate the efficacy of combining REST silencing during directed NPC differentiation to enhance the rate of differentiation and subsequent maturation of NPCs. This study also highlights the potential of RNAi as a biomedical strategy for guided stem cell differentiation.
Collapse
Affiliation(s)
- Wei Ching Low
- Division of Chemical and Biomolecular Engineering, School of Chemical and Biomedical Engineering, Nanyang Technological University, Singapore, Singapore
| | - Winifred Wing Yiu Yau
- Division of Chemical and Biomolecular Engineering, School of Chemical and Biomedical Engineering, Nanyang Technological University, Singapore, Singapore
| | - Lawrence W. Stanton
- Stem Cell and Developmental Biology Group, Genome Institute of Singapore, Singapore, Singapore
| | - Guillaume Marcy
- Duke-NUS Graduate Medical School, National University of Singapore, Singapore, Singapore
| | - Eyleen Goh
- Duke-NUS Graduate Medical School, National University of Singapore, Singapore, Singapore
| | - Sing Yian Chew
- Division of Chemical and Biomolecular Engineering, School of Chemical and Biomedical Engineering, Nanyang Technological University, Singapore, Singapore
| |
Collapse
|
88
|
Chen M, Gao S, Dong M, Song J, Yang C, Howard KA, Kjems J, Besenbacher F. Chitosan/siRNA nanoparticles encapsulated in PLGA nanofibers for siRNA delivery. ACS NANO 2012; 6:4835-4844. [PMID: 22621383 DOI: 10.1021/nn300106t] [Citation(s) in RCA: 142] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/01/2023]
Abstract
Composite nanofibers of biodegradable poly(D,L-lactic-co-glycolic acid) (PLGA) encapsulating chitosan/siRNA nanoparticles (NPs) were prepared by electrospinning. Acidic/alkaline hydrolysis and a bulk/surface degradation mechanism were investigated in order to achieve an optimized release profile for prolonged and efficient gene silencing. Thermo-controlled AFM in situ imaging not only revealed the integrity of the encapsulated chitosan/siRNA polyplex but also shed light on the decreasing T(g) of PLGA on the fiber surfaces during release. A triphasic release profile based on bulk erosion was obtained at pH 7.4, while a triphasic release profile involving both surface erosion and bulk erosion was obtained at pH 5.5. A short alkaline pretreatment provided a homogeneous hydrolysis and consequently a nearly zero-order release profile. The interesting release profile was further investigated for siRNA transfection, where the encapsulated chitosan/siRNA NPs exhibited up to 50% EGFP gene silencing activity after 48 h post-transfection on H1299 cells.
Collapse
Affiliation(s)
- Menglin Chen
- Interdisciplinary Nanoscience Center (iNANO), Aarhus University, DK-8000 Aarhus C, Denmark.
| | | | | | | | | | | | | | | |
Collapse
|
89
|
Electrospun matrices for localized drug delivery: Current technologies and selected biomedical applications. Eur J Pharm Biopharm 2012; 81:1-13. [DOI: 10.1016/j.ejpb.2012.01.016] [Citation(s) in RCA: 212] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2011] [Revised: 01/28/2012] [Accepted: 01/31/2012] [Indexed: 01/13/2023]
|
90
|
Cao H, Marcy G, Goh ELK, Wang F, Wang J, Chew SY. The Effects of Nanofiber Topography on Astrocyte Behavior and Gene Silencing Efficiency. Macromol Biosci 2012; 12:666-74. [DOI: 10.1002/mabi.201100436] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2011] [Revised: 01/08/2012] [Indexed: 01/10/2023]
|
91
|
Jiang X, Cao HQ, Shi LY, Ng SY, Stanton LW, Chew SY. Nanofiber topography and sustained biochemical signaling enhance human mesenchymal stem cell neural commitment. Acta Biomater 2012; 8:1290-302. [PMID: 22154861 DOI: 10.1016/j.actbio.2011.11.019] [Citation(s) in RCA: 86] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2011] [Revised: 10/20/2011] [Accepted: 11/14/2011] [Indexed: 11/26/2022]
Abstract
Stem cells hold great promise in enhancing nerve regeneration. In particular, human mesenchymal stem cells (MSC) represent a clinically viable cell source due in part to their abundance and accessibility. Unfortunately, current methods to direct the fate of stem cells remains largely limited to biochemical-based approaches on two-dimensional substrates with restricted efficacies. Here we have evaluated a scaffold-based approach to directing stem cell differentiation. We demonstrate the combined effects of nanofiber topography and controlled drug release on enhancing MSC neural commitment. By encapsulating up to 0.3 wt.% retinoic acid (RA) within aligned poly(ε-caprolactone) (PCL) nanofibers (average diameter ∼270 nm, AF750), sustained released of RA was obtained for at least 14 days (∼60% released). Compared with tissue culture polystyrene (TCPS), the nanofiber topography arising from plain PCL nanofibers significantly up-regulated the expressions of neural markers, Tuj-1, MAP2, GalC and RIP at the mRNA and protein levels. Combined with sustained drug availability, more significant changes in cell morphology and enhancement of neural marker expression were observed. In particular, scaffold-based controlled delivery of RA enhanced MAP2 and RIP expression compared with bolus delivery despite lower amounts of drug (>8 times lower). The generally higher expression of the mature neuronal marker MAP2 compared with glial markers at the mRNA and protein levels suggested an enhanced potential of MSC neuronal differentiation. In addition, positive staining for synaptophysin was detected only in cells cultured on aligned scaffolds in the presence of RA. Taken together, the results highlight the advantage of the scaffold-based approach in enhancing the potential of MSC neuronal differentiation and demonstrated the importance of the drug delivery approach in directing cell fate. Such biomimicking drug-encapsulating scaffolds may permit subsequent direct cell transplantation and provide guidance cues to control the fate of endogenously recruited stem cells.
Collapse
|
92
|
Liu T, Houle JD, Xu J, Chan BP, Chew SY. Nanofibrous collagen nerve conduits for spinal cord repair. Tissue Eng Part A 2012; 18:1057-66. [PMID: 22220714 DOI: 10.1089/ten.tea.2011.0430] [Citation(s) in RCA: 108] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Nerve regeneration in an injured spinal cord is often restricted, contributing to the devastating outcome of neurologic impairment below the site of injury. Although implantation of tissue-engineered scaffolds has evolved as a potential treatment method, the outcomes remain sub-optimal. One possible reason may be the lack of topographical signals from these constructs to provide contact guidance to invading cells or regrowing axons. Nanofibers mimic the natural extracellular matrix architecturally and may therefore promote physiologically relevant cellular phenotypes. In this study, the potential application of electrospun collagen nanofibers (diameter=208.2±90.4 nm) for spinal cord injury (SCI) treatment was evaluated in vitro and in vivo. Primary rat astrocytes and dorsal root ganglias (DRGs) were seeded on collagen-coated glass cover slips (two-dimensional [2D] substrate controls), and randomly oriented or aligned collagen fibers to evaluate scaffold topographical effects on astrocyte behavior and neurite outgrowth, respectively. When cultured on collagen nanofibers, astrocyte proliferation and expression of glial fibrillary acidic protein (GFAP) were suppressed as compared to cells on 2D controls at days 3 (p<0.05) and 7 (p<0.01). Aligned fibers resulted in elongated astrocytes (elongation factor >4, p<0.01) and directed the orientation of neurite outgrowth from DRGs along fiber axes. In the contrast, neurites emanated radially on randomly oriented collagen fibers. By forming collagen scaffolds into spiral tubular structures, we demonstrated the feasibility of using electrospun nanofibers for the treatment of acute SCI using a rat hemi-section model. At days 10 and 30 postimplantation, extensive cellular penetration into the constructs was observed regardless of fiber orientation. However, scaffolds with aligned fibers appeared more structurally intact at day 30. ED1 immunofluorescent staining revealed macrophage invasion by day 10, which decreased significantly by day 30. Neural fiber sprouting as evaluated by neurofilament staining was observed as early as day 10. In addition, GFAP immunostained astrocytes were found only at the boundary of the lesion site, and no astrocyte accumulation was observed in the implantation area at any time point. These findings indicate the feasibility of fabricating 3D spiral constructs using electrospun collagen fibers and demonstrated the potential of these scaffolds for SCI repair.
Collapse
Affiliation(s)
- Ting Liu
- School of Chemical and Biomedical Engineering, Nanyang Technological University, Singapore, Singapore
| | | | | | | | | |
Collapse
|
93
|
Thin-layer hydroxyapatite deposition on a nanofiber surface stimulates mesenchymal stem cell proliferation and their differentiation into osteoblasts. J Biomed Biotechnol 2012; 2012:428503. [PMID: 22319242 PMCID: PMC3272836 DOI: 10.1155/2012/428503] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2011] [Accepted: 10/19/2011] [Indexed: 01/13/2023] Open
Abstract
Pulsed laser deposition was proved as a suitable method for hydroxyapatite (HA) coating of coaxial poly-ɛ-caprolactone/polyvinylalcohol (PCL/PVA) nanofibers. The fibrous morphology of PCL/PVA nanofibers was preserved, if the nanofiber scaffold was coated with thin layers of HA (200 nm and 400 nm). Increasing thickness of HA, however, resulted in a gradual loss of fibrous character. In addition, biomechanical properties were improved after HA deposition on PCL/PVA nanofibers as the value of Young's moduli of elasticity significantly increased. Clearly, thin-layer hydroxyapatite deposition on a nanofiber surface stimulated mesenchymal stem cell viability and their differentiation into osteoblasts. The optimal depth of HA was 800 nm.
Collapse
|
94
|
Biomimicking Polysaccharide Nanofibers Promote Vascular Phenotypes: A Potential Application for Vascular Tissue Engineering. Macromol Biosci 2011; 12:395-401. [DOI: 10.1002/mabi.201100336] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2011] [Revised: 10/26/2011] [Indexed: 11/07/2022]
|
95
|
Yau WWY, Rujitanaroj PO, Lam L, Chew SY. Directing stem cell fate by controlled RNA interference. Biomaterials 2011; 33:2608-28. [PMID: 22209557 DOI: 10.1016/j.biomaterials.2011.12.021] [Citation(s) in RCA: 70] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2011] [Accepted: 12/09/2011] [Indexed: 01/26/2023]
Abstract
Directing stem cell fate remains a major area of interest and also a hurdle to many, particularly in the field of regenerative medicine. Unfortunately, conventional methods of over-expressing inductive factors through the use of biochemical induction cocktails have led to sub-optimal outcomes. A potential alternative may be to adopt the opposite by selectively silencing genes or pathways that are pivotal to stem cell differentiation. Indeed, over recent years, there have been an increasing number of studies on directing stem cell fate through gene knockdown via RNA interference (RNAi). While the effectiveness of RNAi in controlling stem cell differentiation is evident from the myriad of studies, a chaotically vast collection of gene silencing targets have also been identified. Meanwhile, variations in methods of transfecting stem cells have also affected silencing efficiencies and the subsequent extent of stem cell differentiation. This review serves to unite the pioneers who have ventured into the emerging field of RNAi-enhanced stem cell differentiation by summarizing and evaluating the current approaches adopted in utilizing gene silencing to direct stem cell fate and their corresponding outcomes.
Collapse
Affiliation(s)
- Winifred Wing Yiu Yau
- School of Chemical and Biomedical Engineering, Nanyang Technological University, 637459, Singapore
| | | | | | | |
Collapse
|
96
|
Dahlin RL, Kasper FK, Mikos AG. Polymeric nanofibers in tissue engineering. TISSUE ENGINEERING. PART B, REVIEWS 2011; 17:349-64. [PMID: 21699434 PMCID: PMC3179616 DOI: 10.1089/ten.teb.2011.0238] [Citation(s) in RCA: 202] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/29/2011] [Accepted: 06/22/2011] [Indexed: 01/07/2023]
Abstract
Polymeric nanofibers can be produced using methods such as electrospinning, phase separation, and self-assembly, and the fiber composition, diameter, alignment, degradation, and mechanical properties can be tailored to the intended application. Nanofibers possess unique advantages for tissue engineering. The small diameter closely matches that of extracellular matrix fibers, and the relatively large surface area is beneficial for cell attachment and bioactive factor loading. This review will update the reader on the aspects of nanofiber fabrication and characterization important to tissue engineering, including control of porous structure, cell infiltration, and fiber degradation. Bioactive factor loading will be discussed with specific relevance to tissue engineering. Finally, applications of polymeric nanofibers in the fields of bone, cartilage, ligament and tendon, cardiovascular, and neural tissue engineering will be reviewed.
Collapse
Affiliation(s)
- Rebecca L Dahlin
- Department of Bioengineering, Rice University, Houston, Texas 77251-1892, USA
| | | | | |
Collapse
|
97
|
Rujitanaroj PO, Wang YC, Wang J, Chew SY. Nanofiber-mediated controlled release of siRNA complexes for long term gene-silencing applications. Biomaterials 2011; 32:5915-23. [PMID: 21596430 DOI: 10.1016/j.biomaterials.2011.04.065] [Citation(s) in RCA: 113] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2011] [Accepted: 04/23/2011] [Indexed: 12/20/2022]
Abstract
Nanofiber scaffold-mediated delivery of small-interfering RNA (siRNA) holds great potential in regenerative medicine by providing biomimicking topographical signals and enhanced gene silencing effects to seeded cells. While the delivery of naked siRNA was demonstrated previously using poly (ε-caprolactone) (PCL) nanofibers, the resulting siRNA release kinetics and gene knockdown efficiencies were sub-optimal. In this study, we investigated the feasibility of encapsulating siRNA and transfection reagent (TKO) complexes within nanofibers comprising of a copolymer of caprolactone and ethyl ethylene phosphate (PCLEEP, diameter ∼ 400 nm). Sustained release of bioactive naked siRNA and siRNA/TKO complexes were obtained for at least 28 days. By copolymerizing EEP with caprolactone, siRNA release was significantly enhanced (total siRNA that was released by day 49 was ∼ 89.3-97.2% as compared to previously reported 3% by plain PCL nanofiber delivery). Using GAPDH as the model protein, bioactivity analyses by supernatant transfection revealed the partial retention of bioactivity of naked siRNA and siRNA/TKO complexes for at least 30 days. In particular, GAPDH siRNA/TKO supernatant alone induced significant gene silencing (∼40%), indicating the feasibility of co-encapsulating siRNA and transfection reagent within a single scaffold construct for sustained delivery. Direct culture of cells on siRNA incorporated scaffolds for scaffold-mediated gene transfection revealed significant gene knockdown even in the absence of transfection reagent (21.3% knockdown efficiency by scaffolds incorporating naked siRNA only). By encapsulating siRNA/TKO complexes, more significant gene knockdown was obtained (30.9% knockdown efficiency as compared to previously reported 18% by plain PCL scaffold-mediated transfection). Taken together, the results demonstrated the feasibility of co-encapsulating siRNA-transfection reagent complexes within a single nanofiber construct for sustained siRNA delivery and enhanced gene knockdown efficiency. The study also highlights the potential of PCLEEP as a platform for tailoring siRNA release kinetics for long-term gene silencing applications.
Collapse
Affiliation(s)
- Pim-on Rujitanaroj
- Division of Chemical and Biomolecular Engineering, School of Chemical and Biomedical Engineering, Nanyang Technological University, Singapore, Singapore
| | | | | | | |
Collapse
|
98
|
Monaghan M, Pandit A. RNA interference therapy via functionalized scaffolds. Adv Drug Deliv Rev 2011; 63:197-208. [PMID: 21241760 DOI: 10.1016/j.addr.2011.01.006] [Citation(s) in RCA: 69] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2010] [Revised: 12/16/2010] [Accepted: 01/05/2011] [Indexed: 12/27/2022]
Abstract
Tissue engineering aims to provide structural and biomolecular cues to compromised tissues through scaffolds. An emerging biomolecular cue is that of RNA interference by which the expression of genes can be silenced through a potent endogenous pathway. Recombinant viral-based approaches in RNAi delivery exist; however non-viral strategies offer many opportunities to exploit this mechanism of regulation in a safer way. Current RNAi therapies in clinical trials are without a vector (naked) or have slightly modified structures. Modification of these molecules with efficient backbone moieties for improved stability and potency, protecting and buffering them with delivery vehicles, and using scaffolds as reservoirs of delivery is at the frontier of current research. However, to enable an efficient sustained therapeutic effect scaffolds have a potentially significant role to play. This review presents non-viral delivery of RNAi that have been attempted via tissue engineered scaffolds. For RNAi to have a clinical impact, it is imperative to evaluate optimal delivery systems to ensure that the efficacy of this promising technology can be maximized.
Collapse
Affiliation(s)
- Michael Monaghan
- Network of Excellence for Functional Biomaterials, National University of Ireland-Galway, Ireland
| | | |
Collapse
|
99
|
Chew SY, Low WC. Scaffold-based approach to direct stem cell neural and cardiovascular differentiation: An analysis of physical and biochemical effects. J Biomed Mater Res A 2011; 97:355-74. [DOI: 10.1002/jbm.a.33064] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2010] [Revised: 01/11/2011] [Accepted: 01/24/2011] [Indexed: 01/12/2023]
|
100
|
|