1
|
Goyal MM, Shen SA, Lehar M, Martinez A, Hiel H, Wang C, Liu Y, Wang C, Sun DQ. A Benchtop Round Window Model for Studying Magnetic Nanoparticle Transport to the Inner Ear. Laryngoscope 2024; 134:3355-3362. [PMID: 38379206 DOI: 10.1002/lary.31345] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Revised: 01/09/2024] [Accepted: 01/30/2024] [Indexed: 02/22/2024]
Abstract
INTRODUCTION The round window membrane (RWM) presents a significant barrier to the local application of therapeutics to the inner ear. We demonstrate a benchtop preclinical RWM model and evaluate superparamagnetic iron oxide nanoparticles (SPIONs) as vehicles for magnetically assisted drug delivery. METHODS Guinea pig RWM explants were inset into a 3D-printed dual chamber benchtop device. Custom-synthesized 7-nm iron core nanoparticles were modified with different polyethylene glycol chains to yield two sizes of SPIONs (NP-PEG600 and NP-PEG3000) and applied to the benchtop model with and without a magnetic field. Histologic analysis of the RWM was performed using transmission electron microscopy (TEM) and confocal microscopy. RESULTS Over a 4-h period, 19.5 ± 1.9% of NP-PEG3000 and 14.6 ± 1.9% of NP-PEG600 were transported across the guinea pig RWM. The overall transport increased by 1.45× to 28.4 ± 5.8% and 21.0 ± 2.0%, respectively, when a magnetic field was applied. Paraformaldehyde fixation of the RWM decreased transport significantly (NP-PEG3000: 7.6 ± 1.5%; NP-PEG600: 7.0 ± 1.6%). Confocal and electron microscopy analysis demonstrated nanoparticle localization throughout all cellular layers and layer-specific transport characteristics within RWM. CONCLUSION The guinea pig RWM explant benchtop model allows for targeted and practical investigations of transmembrane transport in the development of nanoparticle drug delivery vehicles. The presence of a magnetic field increases SPION delivery by 45%-50% in a nanoparticle size- and cellular layer-dependent manner. LEVEL OF EVIDENCE NA Laryngoscope, 134:3355-3362, 2024.
Collapse
Affiliation(s)
- Mukund M Goyal
- Department of Chemical & Biomolecular Engineering, Johns Hopkins University, Baltimore, Maryland, USA
| | - Sarek A Shen
- Department of Otolaryngology-Head and Neck Surgery, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Mohamed Lehar
- Department of Otolaryngology-Head and Neck Surgery, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Angela Martinez
- Department of Otolaryngology-Head and Neck Surgery, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Hakim Hiel
- Department of Otolaryngology-Head and Neck Surgery, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Canhui Wang
- Department of Chemical & Biomolecular Engineering, Johns Hopkins University, Baltimore, Maryland, USA
| | - Yulin Liu
- Department of Chemical & Biomolecular Engineering, Johns Hopkins University, Baltimore, Maryland, USA
| | - Chao Wang
- Department of Otolaryngology-Head and Neck Surgery, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Daniel Q Sun
- Department of Chemical & Biomolecular Engineering, Johns Hopkins University, Baltimore, Maryland, USA
- Department of Otolaryngology-Head and Neck Surgery, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| |
Collapse
|
2
|
Wagle SR, Kovacevic B, Ionescu CM, Foster T, Jones M, Mikov M, Wise A, Mooranian A, Al-Salami H. Probucol-bile acid based nanoparticles protect auditory cells from oxidative stress: an in vitro study. Ther Deliv 2024; 15:237-252. [PMID: 38469721 DOI: 10.4155/tde-2023-0099] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/13/2024] Open
Abstract
Aim: Excessive free radicals contribute to oxidative stress and mitochondrial dysfunction in sensorineural hearing loss (SNHL). The antioxidant probucol holds promise, but its limited bioavailability and inner ear barriers hinder effective SNHL treatment. Methodology: We addressed this by developing probucol-loaded nanoparticles with polymers and lithocholic acid and tested them on House Ear Institute-Organ of Corti cells. Results: Probucol-based nanoparticles effectively reduced oxidative stress-induced apoptosis, enhanced cellular viability, improved probucol uptake and promoted mitochondrial function. Additionally, they demonstrated the capacity to reduce reactive oxygen species through the nuclear factor erythroid 2-related factor 2/heme oxygenase-1 pathway. Conclusion: This innovative nanoparticle system holds the potential to prevent oxidative stress-related hearing impairment, providing an effective solution for SNHL.
Collapse
Affiliation(s)
- Susbin Raj Wagle
- The Biotechnology & Drug Development Research Laboratory, Curtin Medical School & Curtin Health Innovation Research Institute, Curtin University, Bentley 6102, Perth, Western Australia, Australia
| | - Bozica Kovacevic
- The Biotechnology & Drug Development Research Laboratory, Curtin Medical School & Curtin Health Innovation Research Institute, Curtin University, Bentley 6102, Perth, Western Australia, Australia
| | - Corina Mihaela Ionescu
- The Biotechnology & Drug Development Research Laboratory, Curtin Medical School & Curtin Health Innovation Research Institute, Curtin University, Bentley 6102, Perth, Western Australia, Australia
| | - Thomas Foster
- The Biotechnology & Drug Development Research Laboratory, Curtin Medical School & Curtin Health Innovation Research Institute, Curtin University, Bentley 6102, Perth, Western Australia, Australia
| | - Melissa Jones
- The Biotechnology & Drug Development Research Laboratory, Curtin Medical School & Curtin Health Innovation Research Institute, Curtin University, Bentley 6102, Perth, Western Australia, Australia
| | - Momir Mikov
- Department of Pharmacology, Toxicology & Clinical Pharmacology, Faculty of Medicine, University of Novi Sad, Novi Sad (Hajduk Veljkova 3, 21101), Serbia
| | | | - Armin Mooranian
- The Biotechnology & Drug Development Research Laboratory, Curtin Medical School & Curtin Health Innovation Research Institute, Curtin University, Bentley 6102, Perth, Western Australia, Australia
- School of Pharmacy, University of Otago, Dunedin, Otago, New Zealand
| | - Hani Al-Salami
- The Biotechnology & Drug Development Research Laboratory, Curtin Medical School & Curtin Health Innovation Research Institute, Curtin University, Bentley 6102, Perth, Western Australia, Australia
- Medical School, University of Western Australia, Perth, Western Australia, Australia
| |
Collapse
|
3
|
Xu K, Xu B, Gu J, Wang X, Yu D, Chen Y. Intrinsic mechanism and pharmacologic treatments of noise-induced hearing loss. Theranostics 2023; 13:3524-3549. [PMID: 37441605 PMCID: PMC10334830 DOI: 10.7150/thno.83383] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Accepted: 06/12/2023] [Indexed: 07/15/2023] Open
Abstract
Noise accounts for one-third of hearing loss worldwide. Regretfully, noise-induced hearing loss (NIHL) is deemed to be irreversible due to the elusive pathogenic mechanisms that have not been fully elucidated. The complex interaction between genetic and environmental factors, which influences numerous downstream molecular and cellular events, contributes to the NIHL. In clinical settings, there are no effective therapeutic drugs other than steroids, which are the only treatment option for patients with NIHL. Therefore, the need for treatment of NIHL that is currently unmet, along with recent progress in our understanding of the underlying regulatory mechanisms, has led to a lot of new literatures focusing on this therapeutic field. The emergence of novel technologies that modify local drug delivery to the inner ear has led to the development of promising therapeutic approaches, which are currently under clinical investigation. In this comprehensive review, we focus on outlining and analyzing the basics and potential therapeutics of NIHL, as well as the application of biomaterials and nanomedicines in inner ear drug delivery. The objective of this review is to provide an incentive for NIHL's fundamental research and future clinical translation.
Collapse
Affiliation(s)
- Ke Xu
- Department of Otolaryngology-Head and Neck Surgery, Shanghai Ninth People's Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Ear Institute, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Key Laboratory of Translational Medicine on Ear and Nose Diseases, Shanghai, China
| | - Baoying Xu
- Materdicine Lab, School of Life Sciences, Shanghai University, Shanghai, China
| | - Jiayi Gu
- Department of Otolaryngology-Head and Neck Surgery, Shanghai Ninth People's Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Ear Institute, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Key Laboratory of Translational Medicine on Ear and Nose Diseases, Shanghai, China
| | - Xueling Wang
- Department of Otolaryngology-Head and Neck Surgery, Shanghai Ninth People's Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Ear Institute, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Key Laboratory of Translational Medicine on Ear and Nose Diseases, Shanghai, China
| | - Dehong Yu
- Materdicine Lab, School of Life Sciences, Shanghai University, Shanghai, China
| | - Yu Chen
- Materdicine Lab, School of Life Sciences, Shanghai University, Shanghai, China
| |
Collapse
|
4
|
Singh R, Birru B, Veit JGS, Arrigali EM, Serban MA. Development and Characterization of an In Vitro Round Window Membrane Model for Drug Permeability Evaluations. Pharmaceuticals (Basel) 2022; 15:ph15091105. [PMID: 36145326 PMCID: PMC9504332 DOI: 10.3390/ph15091105] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Revised: 08/29/2022] [Accepted: 08/30/2022] [Indexed: 12/11/2022] Open
Abstract
Hearing loss and balance disorders are highly common disorders, and the development of effective oto-therapeutics remains an area of intense research. Drug development and screening in the hearing research field heavily rely on the use of preclinical models with often ambiguous translational relevance. This often leads to failed advancement in the market of effective therapeutics. In this context, especially for inner ear-specific pathologies, the availability of an in vitro, physiologically relevant, round window membrane (RWM) model could enable rapid, high-throughput screening of potential topical drugs for inner ear and cochlear dysfunctions and could help accelerate the advancement to clinic and market of more viable drug candidates. In this study, we report the development and evaluation of an in vitro model that mimics the native RWM tissue morphology and microenvironment as shown via immunostaining and histological analyses. The developed three-dimensional (3D) in vitro model was additionally assessed for barrier integrity by transepithelial electrical resistance, and the permeability of lipophilic and hydrophilic drugs was determined. Our collective findings suggest that this in vitro model could serve as a tool for rapid development and screening of topically deliverable oto-therapeutics.
Collapse
Affiliation(s)
- Ruby Singh
- Department of Biomedical and Pharmaceutical Sciences, University of Montana, 32 Campus Dr., Skaggs 394, Missoula, MT 59812, USA
- Montana Biotechnology Center (BIOTECH), University of Montana, Missoula, MT 59812, USA
| | - Bhaskar Birru
- Department of Biomedical and Pharmaceutical Sciences, University of Montana, 32 Campus Dr., Skaggs 394, Missoula, MT 59812, USA
- Montana Biotechnology Center (BIOTECH), University of Montana, Missoula, MT 59812, USA
| | - Joachim G. S. Veit
- Department of Biomedical and Pharmaceutical Sciences, University of Montana, 32 Campus Dr., Skaggs 394, Missoula, MT 59812, USA
- Montana Biotechnology Center (BIOTECH), University of Montana, Missoula, MT 59812, USA
| | - Elizabeth M. Arrigali
- Department of Biomedical and Pharmaceutical Sciences, University of Montana, 32 Campus Dr., Skaggs 394, Missoula, MT 59812, USA
- Montana Biotechnology Center (BIOTECH), University of Montana, Missoula, MT 59812, USA
| | - Monica A. Serban
- Department of Biomedical and Pharmaceutical Sciences, University of Montana, 32 Campus Dr., Skaggs 394, Missoula, MT 59812, USA
- Montana Biotechnology Center (BIOTECH), University of Montana, Missoula, MT 59812, USA
- Correspondence:
| |
Collapse
|
5
|
Shivanna AT, Dash BS, Chen JP. Functionalized Magnetic Nanoparticles for Alternating Magnetic Field- or Near Infrared Light-Induced Cancer Therapies. MICROMACHINES 2022; 13:mi13081279. [PMID: 36014201 PMCID: PMC9413965 DOI: 10.3390/mi13081279] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Revised: 08/02/2022] [Accepted: 08/06/2022] [Indexed: 05/14/2023]
Abstract
The multi-faceted nature of functionalized magnetic nanoparticles (fMNPs) is well-suited for cancer therapy. These nanocomposites can also provide a multimodal platform for targeted cancer therapy due to their unique magnetic guidance characteristics. When induced by an alternating magnetic field (AMF), fMNPs can convert the magnetostatic energy to heat for magnetic hyperthermia (MHT), as well as for controlled drug release. Furthermore, with the ability to convert near-infrared (NIR) light energy to heat energy, fMNPs have attracted interest for photothermal therapy (PTT). Other than MHT and PTT, fMNPs also have a place in combination cancer therapies, such as chemo-MHT, chemo-PTT, and chemo-PTT-photodynamic therapy, among others, due to their versatile properties. Thus, this review presents multifunctional nanocomposites based on fMNPs for cancer therapies, induced by an AMF or NIR light. We will first discuss the different fMNPs induced with an AMF for cancer MHT and chemo-MHT. Secondly, we will discuss fMNPs irradiated with NIR lasers for cancer PTT and chemo-PTT. Finally, fMNPs used for dual-mode AMF + NIR-laser-induced magneto-photo-hyperthermia (MPHT) will be discussed.
Collapse
Affiliation(s)
| | - Banendu Sunder Dash
- Department of Chemical and Materials Engineering, Chang Gung University, Kwei-San, Taoyuan 33302, Taiwan
| | - Jyh-Ping Chen
- Department of Chemical and Materials Engineering, Chang Gung University, Kwei-San, Taoyuan 33302, Taiwan
- Department of Neurosurgery, Chang Gung Memorial Hospital at Linkou, Kwei-San, Taoyuan 33305, Taiwan
- Research Center for Food and Cosmetic Safety, College of Human Ecology, Chang Gung University of Science and Technology, Taoyuan 33305, Taiwan
- Department of Materials Engineering, Ming Chi University of Technology, Tai-Shan, New Taipei City 24301, Taiwan
- Correspondence: ; Tel.: +886-3-2118800
| |
Collapse
|
6
|
Zhang L, Bai X, Wang R, Xu L, Ma J, Xu Y, Lu Z. Advancements in the studies of novel nanomaterials for inner ear drug delivery. Nanomedicine (Lond) 2022; 17:1463-1475. [PMID: 36189895 DOI: 10.2217/nnm-2022-0121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Hearing loss is currently one of the most prevalent sensory disorders worldwide. Because both the blood-labyrinth barrier and the limited blood circulation in the inner ear restrain the effective delivery of most drugs to the inner ear tissues, current treatments for hearing loss are limited to mainly medication, hearing devices and cochlear surgery for therapeutic purposes, whereas treatments lack a noninvasive targeted drug-delivery system. With the continuously rapid development of new nanomaterials, the nanodelivery systems are expected to provide a potentially effective method of clinical treatment for hearing loss. This paper reviews the advantages and disadvantages of the commonly used drug-delivery methods and novel nanomaterials for inner ears as well as advancements in the targeted treatment of hearing loss.
Collapse
Affiliation(s)
- Li Zhang
- Department of Laboratory Medicine, Shandong Provincial Hospital Affiliated to Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, 250021, China
| | - Xiaohui Bai
- Department of Laboratory Medicine, Shandong Provincial Hospital Affiliated to Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, 250021, China
| | - Rongrong Wang
- Department of Clinical Laboratory, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, 250021, China
| | - Lulu Xu
- Department of Laboratory Medicine, Shandong Provincial Hospital Affiliated to Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, 250021, China
| | - Jingyu Ma
- Department of Laboratory Medicine, Shandong Provincial Hospital Affiliated to Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, 250021, China
| | - Yue Xu
- Department of Clinical Laboratory, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, 250021, China
| | - Zhiming Lu
- Department of Laboratory Medicine, Shandong Provincial Hospital Affiliated to Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, 250021, China
| |
Collapse
|
7
|
Martin MJ, Spitzmaul G, Lassalle V. Novel insights and perspectives for the diagnosis and treatment of hearing loss through the implementation of magnetic nanotheranostics. ChemMedChem 2022; 17:e202100685. [PMID: 34978134 DOI: 10.1002/cmdc.202100685] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2021] [Revised: 12/29/2021] [Indexed: 11/06/2022]
Abstract
Hearing loss (HL) is a sensory disability that affects 5% of the world's population. HL predominantly involves damage and death to the cochlear cells. Currently, there is no cure or specific medications for HL. Furthermore, the arrival of therapeutic molecules to the inner ear represents a challenge due to the limited blood supply to the sensory cells and the poor penetration of the blood-cochlear barrier. Superparamagnetic iron oxide nanoparticles (SPIONs) perfectly coordinate with the requirements for controlled drug delivery along with magnetic resonance imaging (MRI) diagnostic and monitoring capabilities. Besides, they are suitable tools to be applied to HL, expecting to be more effective and non-invasive. So far, the published literature only refers to some preclinical studies of SPIONs for HL management. This contribution aims to provide an integrated view of the best options and strategies that can be considered for future research punctually in the field of magnetic nanotechnology applied to HL.
Collapse
Affiliation(s)
- Maria Julia Martin
- INQUISUR: Instituto de Quimica del Sur, Departamento de Química, Universidad Nacional del Sur (CONICET-UNS), Alem 1253, 8000, Bahía Blanca, ARGENTINA
| | - Guillermo Spitzmaul
- Universidad Nacional del Sur Departamento de Biología Bioquímica y Farmacia: Universidad Nacional del Sur Departamento de Biologia Bioquimica y Farmacia, Departamento de Biología, Bioquímica Y farmacia, Camino La Carrindanga Km 7, 8000, Bahía Blanca, ARGENTINA
| | - Verónica Lassalle
- INQUISUR: Instituto de Quimica del Sur, Química, Av Alem 1253, 8000, Bahía Blanca, ARGENTINA
| |
Collapse
|
8
|
Dippong T, Levei EA, Deac IG, Petean I, Borodi G, Cadar O. Sol-Gel Synthesis, Structure, Morphology and Magnetic Properties of Ni 0.6Mn 0.4Fe 2O 4 Nanoparticles Embedded in SiO 2 Matrix. NANOMATERIALS 2021; 11:nano11123455. [PMID: 34947806 PMCID: PMC8708454 DOI: 10.3390/nano11123455] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Revised: 12/16/2021] [Accepted: 12/16/2021] [Indexed: 01/07/2023]
Abstract
The structure, morphology and magnetic properties of (Ni0.6Mn0.4Fe2O4)α(SiO2)100−α (α = 0–100%) nanocomposites (NCs) produced by sol-gel synthesis were investigated using X-ray diffraction (XRD), Fourier transform infrared spectroscopy (FT-IR), atomic force microscopy (AFM) and vibrating sample magnetometry (VSM). At low calcination temperatures (300 °C), poorly crystallized Ni0.6Mn0.4Fe2O4, while at high calcination temperatures, well-crystallized Ni0.6Mn0.4Fe2O4 was obtained along with α-Fe2O3, quartz, cristobalite or iron silicate secondary phase, depending on the Ni0.6Mn0.4Fe2O4 content in the NCs. The average crystallite size increases from 2.6 to 74.5 nm with the increase of calcination temperature and ferrite content embedded in the SiO2 matrix. The saturation magnetization (Ms) enhances from 2.5 to 80.5 emu/g, the remanent magnetization (MR) from 0.68 to 12.6 emu/g and the coercive field (HC) from 126 to 260 Oe with increasing of Ni0.6Mn0.4Fe2O4 content in the NCs. The SiO2 matrix has a diamagnetic behavior with a minor ferromagnetic fraction, Ni0.6Mn0.4Fe2O4 embedded in SiO2 matrix displays superparamagnetic behavior, while unembedded Ni0.6Mn0.4Fe2O4 has a high-quality ferromagnetic behavior.
Collapse
Affiliation(s)
- Thomas Dippong
- Faculty of Science, Technical University of Cluj-Napoca, 76 Victoriei Street, 430122 Baia Mare, Romania
- Correspondence:
| | - Erika Andrea Levei
- INCDO-INOE 2000, Research Institute for Analytical Instrumentation, 67 Donath Street, 400293 Cluj-Napoca, Romania; (E.A.L.); (O.C.)
| | - Iosif Grigore Deac
- Faculty of Physics, Babes-Bolyai University, 1 Kogalniceanu Street, 400084 Cluj-Napoca, Romania;
| | - Ioan Petean
- Faculty of Chemistry and Chemical Engineering, Babes-Bolyai University, 11 Arany Janos Street, 400028 Cluj-Napoca, Romania;
| | - Gheorghe Borodi
- National Institute for Research and Development of Isotopic and Molecular Technologies, 65-103 Donath Street, 400293 Cluj-Napoca, Romania;
| | - Oana Cadar
- INCDO-INOE 2000, Research Institute for Analytical Instrumentation, 67 Donath Street, 400293 Cluj-Napoca, Romania; (E.A.L.); (O.C.)
| |
Collapse
|
9
|
Abstract
With the increasing insight into molecular mechanisms of cardiovascular disease, a promising solution involves directly delivering genes, cells, and chemicals to the infarcted myocardium or impaired endothelium. However, the limited delivery efficiency after administration fails to reach the therapeutic dose and the adverse off-target effect even causes serious safety concerns. Controlled drug release via external stimuli seems to be a promising method to overcome the drawbacks of conventional drug delivery systems (DDSs). Microbubbles and magnetic nanoparticles responding to ultrasound and magnetic fields respectively have been developed as an important component of novel DDSs. In particular, several attempts have also been made for the design and fabrication of dual-responsive DDS. This review presents the recent advances in the ultrasound and magnetic fields responsive DDSs in cardiovascular application, followed by their current problems and future reformation.
Collapse
|
10
|
Lin YC, Shih CP, Chen HC, Chou YL, Sytwu HK, Fang MC, Lin YY, Kuo CY, Su HH, Hung CL, Chen HK, Wang CH. Ultrasound Microbubble-Facilitated Inner Ear Delivery of Gold Nanoparticles Involves Transient Disruption of the Tight Junction Barrier in the Round Window Membrane. Front Pharmacol 2021; 12:689032. [PMID: 34262458 PMCID: PMC8273281 DOI: 10.3389/fphar.2021.689032] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Accepted: 06/16/2021] [Indexed: 11/15/2022] Open
Abstract
The application of ultrasound microbubbles (USMBs) enhances the permeability of the round window membrane (RWM) and improves drug delivery to the inner ear. In this study, we investigated the efficiency of USMB-aided delivery of chitosan-coated gold nanoparticles (CS-AuNPs) and the mechanism of USMB-mediated enhancement of RMW permeability. We exposed mouse inner ears to USMBs at an intensity of 2 W/cm2 and then filled the tympanic bulla with CS-AuNPs or fluorescein isothiocyanate-decorated CS-AuNPs (FITC-CS-AuNPs). The membrane uptake of FITC-CS-AuNPs and their depth of permeation into the three-layer structure of the RWM, with or without prior USMB treatment, were visualized by z-stack confocal laser scanning microscopy. Ultrastructural changes in the RWM due to USMB-mediated cavitation appeared as sunburn-like peeling and various degrees of depression in the RWM surface, with pore-like openings forming in the outer epithelium. This disruption of the outer epithelium was paralleled by a transient reduction in tight junction (TJ)-associated protein levels in the RWM and an enhanced delivery of FITC-CS-AuNPs into the RWM. Without prior USMB exposure, the treatment with CS-AuNPs also caused a noticeable reduction in TJ proteins of the RWM. Our findings indicated that the combined treatment with USMBs and CS-AuNPs represents a promising and efficient drug and gene delivery vehicle for a trans-RWM approach for inner ear therapy. The outer epithelial layer of the RWM plays a decisive role in controlling the transmembrane transport of substances such as CS-AuNPs following the administration of USMBs. Most importantly, the enhanced permeation of AuNPs involved the transient disruption of the TJ-created paracellular barrier in the outer epithelium of the RWM.
Collapse
Affiliation(s)
- Yi-Chun Lin
- National Defense Medical Center, Graduate Institute of Medical Sciences, Taipei, Taiwan
| | - Cheng-Ping Shih
- Department of Otolaryngology-Head and Neck Surgery, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Hsin-Chien Chen
- Department of Otolaryngology-Head and Neck Surgery, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Ying-Liang Chou
- Department of Otolaryngology-Head and Neck Surgery, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan.,Department of Otorhinolaryngology, Taichung Armed Forces General Hospital, Taichung, Taiwan
| | - Huey-Kang Sytwu
- National Defense Medical Center, Graduate Institute of Medical Sciences, Taipei, Taiwan.,National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Zhunan Town, Miaoli County, Taiwan
| | - Mei-Cho Fang
- Laboratory Animal Center, National Defense Medical Center, Taipei, Taiwan
| | - Yuan-Yung Lin
- National Defense Medical Center, Graduate Institute of Medical Sciences, Taipei, Taiwan.,Department of Otolaryngology-Head and Neck Surgery, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Chao-Yin Kuo
- Department of Otolaryngology-Head and Neck Surgery, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Hsiao-Han Su
- Department of Otolaryngology-Head and Neck Surgery, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Chia-Lien Hung
- Department of Otorhinolaryngology, Taichung Armed Forces General Hospital, Taichung, Taiwan
| | - Hang-Kang Chen
- Department of Otolaryngology-Head and Neck Surgery, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan.,Department of Otorhinolaryngology, Taichung Armed Forces General Hospital, Taichung, Taiwan
| | - Chih-Hung Wang
- National Defense Medical Center, Graduate Institute of Medical Sciences, Taipei, Taiwan.,Department of Otolaryngology-Head and Neck Surgery, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan.,Department of Otorhinolaryngology, Taichung Armed Forces General Hospital, Taichung, Taiwan.,National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Zhunan Town, Miaoli County, Taiwan
| |
Collapse
|
11
|
Guigou C, Lalande A, Millot N, Belharet K, Bozorg Grayeli A. Use of Super Paramagnetic Iron Oxide Nanoparticles as Drug Carriers in Brain and Ear: State of the Art and Challenges. Brain Sci 2021; 11:358. [PMID: 33799690 PMCID: PMC7998448 DOI: 10.3390/brainsci11030358] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Revised: 03/02/2021] [Accepted: 03/05/2021] [Indexed: 12/16/2022] Open
Abstract
Drug delivery and distribution in the central nervous system (CNS) and the inner ear represent a challenge for the medical and scientific world, especially because of the blood-brain and the blood-perilymph barriers. Solutions are being studied to circumvent or to facilitate drug diffusion across these structures. Using superparamagnetic iron oxide nanoparticles (SPIONs), which can be coated to change their properties and ensure biocompatibility, represents a promising tool as a drug carrier. They can act as nanocarriers and can be driven with precision by magnetic forces. The aim of this study was to systematically review the use of SPIONs in the CNS and the inner ear. A systematic PubMed search between 1999 and 2019 yielded 97 studies. In this review, we describe the applications of the SPIONS, their design, their administration, their pharmacokinetic, their toxicity and the methods used for targeted delivery of drugs into the ear and the CNS.
Collapse
Affiliation(s)
- Caroline Guigou
- Department of Otolaryngology-Head and Neck Surgery, Dijon University Hospital, 21000 Dijon, France;
- ImVia Laboratory, EA 7535, Université Bourgogne Franche-Comté, 21079 Dijon, France;
| | - Alain Lalande
- ImVia Laboratory, EA 7535, Université Bourgogne Franche-Comté, 21079 Dijon, France;
| | - Nadine Millot
- Laboratoire Interdisciplinaire Carnot de Bourgogne, UMR 6303, CNRS, Université Bourgogne Franche-Comté, BP 47870, 21078 Dijon, France;
| | - Karim Belharet
- Laboratoire PRISME, JUNIA Campus Centre, 36000 Châteauroux, France;
| | - Alexis Bozorg Grayeli
- Department of Otolaryngology-Head and Neck Surgery, Dijon University Hospital, 21000 Dijon, France;
- ImVia Laboratory, EA 7535, Université Bourgogne Franche-Comté, 21079 Dijon, France;
| |
Collapse
|
12
|
An X, Zha D. Development of nanoparticle drug-delivery systems for the inner ear. Nanomedicine (Lond) 2020; 15:1981-1993. [PMID: 32605499 DOI: 10.2217/nnm-2020-0198] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Hearing loss has become the most common sensory nerve disorder worldwide, with no effective treatment strategy. Low-permeability and limited blood supply to the blood-labyrinth barrier limit the effective delivery and efficacy of therapeutic drugs in the inner ear. Nanoparticle (NP)-based drugs have shown benefits of stable controlled release and functional surface modification, and NP-based delivery systems have become a research hotspot. In this review, we discuss the development of new targeted drug-delivery systems based on the biocompatibility and safety of different NPs in the cochlea, as well as the advantages and disadvantages of their prescription methods and approaches. We believe that targeted NP-based drug-delivery systems will be effective treatments for hearing loss.
Collapse
Affiliation(s)
- Xiaogang An
- Department of Otolaryngology - Head & Neck Surgery, Xijing Hospital, Fourth Military Medical University, 127 Changle Western Road, Xi'an, Shaanxi Province, 710032, PR China
| | - Dingjun Zha
- Department of Otolaryngology - Head & Neck Surgery, Xijing Hospital, Fourth Military Medical University, 127 Changle Western Road, Xi'an, Shaanxi Province, 710032, PR China
| |
Collapse
|
13
|
Optimized phospholipid-based nanoparticles for inner ear drug delivery and therapy. Biomaterials 2018; 171:133-143. [PMID: 29689410 DOI: 10.1016/j.biomaterials.2018.04.038] [Citation(s) in RCA: 54] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2018] [Revised: 03/26/2018] [Accepted: 04/14/2018] [Indexed: 12/26/2022]
Abstract
To develop efficient carriers for inner ear drug delivery, we prepared four kinds of phospholipid-based nanoparticles: neutral, anionic, cationic, and cationic-PEG (polyethyleneglycol) particles. PEG was used to maintain long-term particle circulation in the perilymph, avoiding non-specific binding of particles to proteins. All four nanoparticles were about 200 nm in diameter, and their zeta potentials were -4.32, -26.0, +25.8, and -0.28, respectively, for neutral, anionic, cationic, and cationic-PEG nanoparticles. To test particle efficacy in vitro, we used an artificial mucosa 100 μm in thickness to model the round window membrane (RWM) and HEI-OC1 cells, which were treated with particles containing Nile Red dye. Based on the levels of particle penetration and cellular uptake in this model system, we selected an optimal particle for further study. We also observed the movement of particles in ex vivo organotypic cultures of the organ of Corti. In mice, we analyzed the biodistribution of dexamethasone (Dex) in the inner ear after intratympanic injection of Dex-loaded nanoparticles. Then, we tested the therapeutic utility of the Dex-loaded nanoparticles in a mouse model of ototoxicity. In the auditory brainstem response (ABR) test, particle provided improved hearing loss recovery at all tested frequencies, more so than did the Dex sodium phosphate (Dex-SP) solution in current clinical use. Furthermore, quantitative PCR showed that nanoparticles reduced the levels of pro-inflammatory cytokines, exhibiting anti-inflammatory effects superior to those of Dex-SP. Thus, the surface properties of nanoparticles play pivotal roles in particle penetration and distribution after intratympanic injection. Our in vitro screening system using an artificial mucosa will also be valuable in the development of carriers for inner ear drug delivery.
Collapse
|
14
|
Martinac AD, Bavi N, Bavi O, Martinac B. Pulling MscL open via N-terminal and TM1 helices: A computational study towards engineering an MscL nanovalve. PLoS One 2017; 12:e0183822. [PMID: 28859093 PMCID: PMC5578686 DOI: 10.1371/journal.pone.0183822] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2017] [Accepted: 08/11/2017] [Indexed: 11/18/2022] Open
Abstract
There are great opportunities in the manipulation of bacterial mechanosensitive (MS) ion channels for specific and targeted drug delivery purposes. Recent research has shown that these ion channels have the potential to be converted into nanovalves through clever use of magnetic nanoparticles and magnetic fields. Using a combination of molecular dynamics (MD) simulations and the finite element (FE) modelling, this study investigates the theoretical feasibility of opening the MscL channel (MS channel of large conductance of E. coli) by applying mechanical force directly to its N-terminus. This region has already been reported to function as a major mechanosensor in this channel. The stress-strain behaviour of each MscL helix was obtained using all atom MD simulations. Using the same method, we simulated two models, the wild-type (WT) MscL and the G22N mutant MscL, both embedded in a POPE lipid bilayer. In addition to indicating the main interacting residues at the hydrophobic pore, their pairwise interaction energies were monitored during the channel gating. We implemented these inputs into our FE model of MscL using curve-fitting codes and continuum mechanics equations. In the FE model, the channel could be fully opened via pulling directly on the N-terminus and bottom of TM1 by mutating dominant van der Waals interactions in the channel pore; otherwise the stress generated on the channel protein can irreversibly unravel the N-secondary structure. This is a significant finding suggesting that applying force in this manner is sufficient to open an MscL nanovalve delivering various drugs used, for example, in cancer chemotherapy. More importantly, the FE model indicates that to fully operate an MscL nanovalve by pulling directly on the N-terminus and bottom of TM1, gain-of-function (GOF) mutants (e.g., G22N MscL) would have to be employed rather than the WT MscL channel.
Collapse
Affiliation(s)
- Adam D. Martinac
- Molecular Cardiology and Biophysics Division, Victor Chang Cardiac Research Institute, Darlinghurst, NSW, Australia
| | - Navid Bavi
- Molecular Cardiology and Biophysics Division, Victor Chang Cardiac Research Institute, Darlinghurst, NSW, Australia
- St Vincent’s Clinical School, Faculty of Medicine, University of New South Wales, Darlinghurst, NSW, Australia
| | - Omid Bavi
- Department of Physics, University of Tehran, Tehran, Iran
| | - Boris Martinac
- Molecular Cardiology and Biophysics Division, Victor Chang Cardiac Research Institute, Darlinghurst, NSW, Australia
- St Vincent’s Clinical School, Faculty of Medicine, University of New South Wales, Darlinghurst, NSW, Australia
- * E-mail:
| |
Collapse
|
15
|
Amin FU, Hoshiar AK, Do TD, Noh Y, Shah SA, Khan MS, Yoon J, Kim MO. Osmotin-loaded magnetic nanoparticles with electromagnetic guidance for the treatment of Alzheimer's disease. NANOSCALE 2017; 9:10619-10632. [PMID: 28534925 DOI: 10.1039/c7nr00772h] [Citation(s) in RCA: 62] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/20/2023]
Abstract
Alzheimer's disease (AD) is the most prevalent age-related neurodegenerative disease, pathologically characterized by the accumulation of aggregated amyloid beta (Aβ) in the brain. Here, we describe for the first time the development of a new, pioneering nanotechnology-based drug delivery approach for potential therapies for neurodegenerative diseases, particularly AD. We demonstrated the delivery of fluorescent carboxyl magnetic Nile Red particles (FMNPs) to the brains of normal mice using a functionalized magnetic field (FMF) composed of positive- and negative-pulsed magnetic fields generated by electromagnetic coils. The FMNPs successfully reached the brain in a few minutes and showed evidence of blood-brain barrier (BBB) crossing. Moreover, the best FMF conditions were found for inducing the FMNPs to reach the cortex and hippocampus regions. Under the same FMF conditions, dextran-coated Fe3O4 magnetic nanoparticles (MNPs) loaded with osmotin (OMNP) were transported to the brains of Aβ1-42-treated mice. Compared with native osmotin, the OMNP potently attenuates Aβ1-42-induced synaptic deficits, Aβ accumulation, BACE-1 expression and tau hyperphosphorylation. This magnetic drug delivery approach can be extended to preclinical and clinical use and may advance the chances of success in the treatment of neurological disorders like AD in the future.
Collapse
Affiliation(s)
- Faiz Ul Amin
- Division of Life Science (BK 21), College of Natural Sciences, Gyeongsang National University (GNU), Jinju, 660-701, Republic of Korea.
| | | | | | | | | | | | | | | |
Collapse
|
16
|
Valente F, Astolfi L, Simoni E, Danti S, Franceschini V, Chicca M, Martini A. Nanoparticle drug delivery systems for inner ear therapy: An overview. J Drug Deliv Sci Technol 2017. [DOI: 10.1016/j.jddst.2017.03.003] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
17
|
Li L, Chao T, Brant J, O'Malley B, Tsourkas A, Li D. Advances in nano-based inner ear delivery systems for the treatment of sensorineural hearing loss. Adv Drug Deliv Rev 2017; 108:2-12. [PMID: 26796230 DOI: 10.1016/j.addr.2016.01.004] [Citation(s) in RCA: 72] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2015] [Revised: 01/03/2016] [Accepted: 01/04/2016] [Indexed: 01/31/2023]
Abstract
Sensorineural hearing loss (SNHL) is one of the most common diseases, accounting for about 90% of all hearing loss. Leading causes of SNHL include advanced age, ototoxic medications, noise exposure, inherited and autoimmune disorders. Most of SNHL is irreversible and managed with hearing aids or cochlear implants. Although there is increased understanding of the molecular pathophysiology of SNHL, biologic treatment options are limited due to lack of noninvasive targeted delivery systems. Obstacles of targeted inner ear delivery include anatomic inaccessibility, biotherapeutic instability, and nonspecific delivery. Advances in nanotechnology may provide a solution to these barriers. Nanoparticles can stabilize and carry biomaterials across the round window membrane into the inner ear, and ligand bioconjugation onto nanoparticle surfaces allows for specific targeting. A newer technology, nanohydrogel, may offer noninvasive and sustained biotherapeutic delivery into specific inner ear cells. Nanohydrogel may be used for inner ear dialysis, a potential treatment for ototoxicity-induced SNHL.
Collapse
Affiliation(s)
- Lilun Li
- Department of Otorhinolaryngology-Head & Neck Surgery, University of Pennsylvania School of Medicine, Philadelphia, PA 19104, USA; New York University School of Medicine, New York, NY 10016, USA
| | - Tiffany Chao
- Department of Otorhinolaryngology-Head & Neck Surgery, University of Pennsylvania School of Medicine, Philadelphia, PA 19104, USA
| | - Jason Brant
- Department of Otorhinolaryngology-Head & Neck Surgery, University of Pennsylvania School of Medicine, Philadelphia, PA 19104, USA
| | - Bert O'Malley
- Department of Otorhinolaryngology-Head & Neck Surgery, University of Pennsylvania School of Medicine, Philadelphia, PA 19104, USA
| | - Andrew Tsourkas
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Daqing Li
- Department of Otorhinolaryngology-Head & Neck Surgery, University of Pennsylvania School of Medicine, Philadelphia, PA 19104, USA.
| |
Collapse
|
18
|
Zou J, Ostrovsky S, Israel LL, Feng H, Kettunen MI, Lellouche JPM, Pyykkö I. Efficient penetration of ceric ammonium nitrate oxidant-stabilized gamma-maghemite nanoparticles through the oval and round windows into the rat inner ear as demonstrated by MRI. J Biomed Mater Res B Appl Biomater 2016; 105:1883-1891. [DOI: 10.1002/jbm.b.33719] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2015] [Revised: 04/04/2016] [Accepted: 05/09/2016] [Indexed: 12/13/2022]
Affiliation(s)
- Jing Zou
- Department of Otolaryngology-Head and Neck Surgery; Center for Otolaryngology-Head & Neck Surgery of Chinese PLA, Changhai Hospital, Second Military Medical University; Shanghai China
- Hearing and Balance Research Unit, Field of Oto-laryngology, School of Medicine, University of Tampere; Tampere Finland
| | - Stella Ostrovsky
- Laboratory of Nanoscale Materials and Systems, Department of Chemistry, Bar-Ilan University; Ramat-Gan Israel
| | - Liron L. Israel
- Laboratory of Nanoscale Materials and Systems, Department of Chemistry, Bar-Ilan University; Ramat-Gan Israel
| | - Hao Feng
- Hearing and Balance Research Unit, Field of Oto-laryngology, School of Medicine, University of Tampere; Tampere Finland
| | - Mikko I. Kettunen
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland; Kuopio Finland
| | - Jean-Paul Moshe Lellouche
- Laboratory of Nanoscale Materials and Systems, Department of Chemistry, Bar-Ilan University; Ramat-Gan Israel
| | - Ilmari Pyykkö
- Hearing and Balance Research Unit, Field of Oto-laryngology, School of Medicine, University of Tampere; Tampere Finland
| |
Collapse
|
19
|
Nguyen Y, Celerier C, Pszczolinski R, Claver J, Blank U, Ferrary E, Sterkers O. Superparamagnetic nanoparticles as vectors for inner ear treatments: driving and toxicity evaluation. Acta Otolaryngol 2016; 136:402-8. [PMID: 26982172 DOI: 10.3109/00016489.2015.1129069] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Conclusion Super paramagnetic nanoparticles (MNP) are a promising vector to achieve controlled drug delivery into the cochlea. Objective The goal of the study was to evaluate the toxicological risk of MNP upon the inner ear. Methods Fe3O4-MNP displacement was studied in various catheter materials, shape, and solvent with a local magnetic field. EC5V cells (derived from the inner ear) were cultured with MNP (100 and 500 nm) at various concentrations or without MNP. Cell survival was assessed with a flow cytometry analysis. Localization of MNP within the cells was studied with confocal microscopy. In vivo, a single intra-cochlear administration of 200 nm MNP (3 × 10(10)MNP/mL, n = 8; 1.5 × 10(12) MNP/mL, n = 6) or saline (n = 14) was performed in guinea pigs. Hearing thresholds were assessed with auditory brainstem responses at Day 7. Results MNP could be concentrated at different locations of the catheter with sequential activation of solenoids. MNP were internalized in the cytoplasm, but not in the nuclei nor in endosomes at 48 h. After 48 h of incubation, no difference for cell survival between the groups was observed, whatever the MNP concentration. A size effect was observed with less survival in the 100 nm group. In guinea pigs at day 7, hearing threshold shift was not different in the three groups.
Collapse
Affiliation(s)
- Yann Nguyen
- a Inserm, 'Minimally Invasive Robot-based Hearing Rehabilitation', UMR-S 1159 , Paris , France
- b Sorbonne University, University Pierre et Marie Curie, UPMC Univ Paris 06 , Paris , France
- c Otolaryngology Department , Unit of Otology, Auditory Implants and Skull Base Surgery, Hospital Pitié Salpêtrière, 47-83 boulevard de l'Hôpital, Cedex 13, , Paris , France
| | - Charlotte Celerier
- a Inserm, 'Minimally Invasive Robot-based Hearing Rehabilitation', UMR-S 1159 , Paris , France
- b Sorbonne University, University Pierre et Marie Curie, UPMC Univ Paris 06 , Paris , France
- c Otolaryngology Department , Unit of Otology, Auditory Implants and Skull Base Surgery, Hospital Pitié Salpêtrière, 47-83 boulevard de l'Hôpital, Cedex 13, , Paris , France
| | - Romain Pszczolinski
- a Inserm, 'Minimally Invasive Robot-based Hearing Rehabilitation', UMR-S 1159 , Paris , France
- b Sorbonne University, University Pierre et Marie Curie, UPMC Univ Paris 06 , Paris , France
- c Otolaryngology Department , Unit of Otology, Auditory Implants and Skull Base Surgery, Hospital Pitié Salpêtrière, 47-83 boulevard de l'Hôpital, Cedex 13, , Paris , France
| | - Julien Claver
- d Inserm 'Kidney Immunopathology, Receptors and Inflammation", UMR-S 1149 , Paris , France
| | - Ulrick Blank
- d Inserm 'Kidney Immunopathology, Receptors and Inflammation", UMR-S 1149 , Paris , France
| | - Evelyne Ferrary
- a Inserm, 'Minimally Invasive Robot-based Hearing Rehabilitation', UMR-S 1159 , Paris , France
- b Sorbonne University, University Pierre et Marie Curie, UPMC Univ Paris 06 , Paris , France
- c Otolaryngology Department , Unit of Otology, Auditory Implants and Skull Base Surgery, Hospital Pitié Salpêtrière, 47-83 boulevard de l'Hôpital, Cedex 13, , Paris , France
| | - Olivier Sterkers
- a Inserm, 'Minimally Invasive Robot-based Hearing Rehabilitation', UMR-S 1159 , Paris , France
- b Sorbonne University, University Pierre et Marie Curie, UPMC Univ Paris 06 , Paris , France
- c Otolaryngology Department , Unit of Otology, Auditory Implants and Skull Base Surgery, Hospital Pitié Salpêtrière, 47-83 boulevard de l'Hôpital, Cedex 13, , Paris , France
| |
Collapse
|
20
|
Leach JC, Wang A, Ye K, Jin S. A RNA-DNA Hybrid Aptamer for Nanoparticle-Based Prostate Tumor Targeted Drug Delivery. Int J Mol Sci 2016; 17:380. [PMID: 26985893 PMCID: PMC4813238 DOI: 10.3390/ijms17030380] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2016] [Revised: 03/08/2016] [Accepted: 03/10/2016] [Indexed: 12/03/2022] Open
Abstract
The side effects of radio- and chemo-therapy pose long-term challenges on a cancer patient’s health. It is, therefore, highly desirable to develop more effective therapies that can specifically target carcinoma cells without damaging normal and healthy cells. Tremendous efforts have been made in the past to develop targeted drug delivery systems for solid cancer treatment. In this study, a new aptamer, A10-3-J1, which recognizes the extracellular domain of the prostate specific membrane antigen (PSMA), was designed. A super paramagnetic iron oxide nanoparticle-aptamer-doxorubicin (SPIO-Apt-Dox) was fabricated and employed as a targeted drug delivery platform for cancer therapy. This DNA RNA hybridized aptamer antitumor agent was able to enhance the cytotoxicity of targeted cells while minimizing collateral damage to non-targeted cells. This SPIO-Apt-Dox nanoparticle has specificity to PSMA+ prostate cancer cells. Aptamer inhibited nonspecific uptake of membrane-permeable doxorubic to the non-target cells, leading to reduced untargeted cytotoxicity and endocytic uptake while enhancing targeted cytotoxicity and endocytic uptake. The experimental results indicate that the drug delivery platform can yield statistically significant effectiveness being more cytotoxic to the targeted cells as opposed to the non-targeted cells.
Collapse
Affiliation(s)
- John C Leach
- Department of Biomedical Engineering, College of Engineering, University of Arkansas, Fayetteville, AR 72701, USA.
| | - Andrew Wang
- Ocean Nanotech, 2143 Worth Lane, Springdale, AR 72764, USA.
| | - Kaiming Ye
- Department of Biomedical Engineering, College of Engineering, University of Arkansas, Fayetteville, AR 72701, USA.
- Department of Biomedical Engineering, Thomas J. Watson School of Engineering and Applied Sciences, State University of New York in Binghamton, Binghamton, NY 13902, USA.
| | - Sha Jin
- Department of Biomedical Engineering, College of Engineering, University of Arkansas, Fayetteville, AR 72701, USA.
- Department of Biomedical Engineering, Thomas J. Watson School of Engineering and Applied Sciences, State University of New York in Binghamton, Binghamton, NY 13902, USA.
| |
Collapse
|
21
|
Pyykkö I, Zou J, Schrott-Fischer A, Glueckert R, Kinnunen P. An Overview of Nanoparticle Based Delivery for Treatment of Inner Ear Disorders. Methods Mol Biol 2016; 1427:363-415. [PMID: 27259938 DOI: 10.1007/978-1-4939-3615-1_21] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Nanoparticles offer new possibilities for inner ear treatment as they can carry a variety of drugs, protein, and nucleic acids to inner ear. Nanoparticles are equipped with several functions such as targetability, immuno-transparency, biochemical stability, and ability to be visualized in vivo and in vitro. A group of novel peptides can be attached to the surface of nanoparticles that will enhance the cell entry, endosomal escape, and nuclear targeting. Eight different types of nanoparticles with different payload carrying strategies are available now. The transtympanic delivery of nanoparticles indicates that, depending on the type of nanoparticle, different migration pathways into the inner ear can be employed, and that optimal carriers can be designed according to the intended cargo. The use of nanoparticles as drug/gene carriers is especially attractive in conjunction with cochlear implantation or even as an inclusion in the implant as a drug/gene reservoir.
Collapse
Affiliation(s)
- Ilmari Pyykkö
- Department of Otolaryngology, University of Tampere and University Hospital of Tampere, Tampere, 33014, Finland. .,Hearing and Balance Research Unit, Field of Otolaryngology, School of Medicine, University of Tampere, Medisiinarinkatu 3, Tampere, 33520, Finland.
| | - Jing Zou
- BECS, Department of Biomedical Engineering and Computational Science, Aalto University, Aalto, 02150, Espoo, Finland
| | - Annelies Schrott-Fischer
- Department of Otolaryngology, Medical University of Innsbruck, Anichstrasse 35, Innsbruck, 6020, Austria
| | - Rudolf Glueckert
- Department of Otolaryngology, Medical University of Innsbruck, Anichstrasse 35, Innsbruck, 6020, Austria
| | - Paavo Kinnunen
- BECS, Department of Biomedical Engineering and Computational Science, Aalto University, Aalto, Finland
| |
Collapse
|
22
|
Durymanov MO, Rosenkranz AA, Sobolev AS. Current Approaches for Improving Intratumoral Accumulation and Distribution of Nanomedicines. Theranostics 2015; 5:1007-20. [PMID: 26155316 PMCID: PMC4493538 DOI: 10.7150/thno.11742] [Citation(s) in RCA: 125] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2015] [Accepted: 05/09/2015] [Indexed: 12/22/2022] Open
Abstract
The ability of nanoparticles and macromolecules to passively accumulate in solid tumors and enhance therapeutic effects in comparison with conventional anticancer agents has resulted in the development of various multifunctional nanomedicines including liposomes, polymeric micelles, and magnetic nanoparticles. Further modifications of these nanoparticles have improved their characteristics in terms of tumor selectivity, circulation time in blood, enhanced uptake by cancer cells, and sensitivity to tumor microenvironment. These "smart" systems have enabled highly effective delivery of drugs, genes, shRNA, radioisotopes, and other therapeutic molecules. However, the resulting therapeutically relevant local concentrations of anticancer agents are often insufficient to cause tumor regression and complete elimination. Poor perfusion of inner regions of solid tumors as well as vascular barrier, high interstitial fluid pressure, and dense intercellular matrix are the main intratumoral barriers that impair drug delivery and impede uniform distribution of nanomedicines throughout a tumor. Here we review existing methods and approaches for improving tumoral uptake and distribution of nano-scaled therapeutic particles and macromolecules (i.e. nanomedicines). Briefly, these strategies include tuning physicochemical characteristics of nanomedicines, modulating physiological state of tumors with physical impacts or physiologically active agents, and active delivery of nanomedicines using cellular hitchhiking.
Collapse
|
23
|
Lewis EEL, Child HW, Hursthouse A, Stirling D, McCully M, Paterson D, Mullin M, Berry CC. The influence of particle size and static magnetic fields on the uptake of magnetic nanoparticles into three dimensional cell-seeded collagen gel cultures. J Biomed Mater Res B Appl Biomater 2014; 103:1294-301. [PMID: 25358626 DOI: 10.1002/jbm.b.33302] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2014] [Revised: 08/18/2014] [Accepted: 10/01/2014] [Indexed: 12/31/2022]
Abstract
Over recent decades there has been and continues to be major advances in the imaging, diagnosis and potential treatment of medical conditions, by the use of magnetic nanoparticles. However, to date the majority of cell delivery studies employ a traditional 2D monolayer culture. This article aims to determine the ability of various sized magnetic nanoparticles to penetrate and travel through a cell seeded collagen gel model, in the presence or absence of a magnetic field. Three different sized (100, 200, and 500 nm) nanoparticles were employed in the study. The results showed cell viability was unaffected by the presence of nanoparticles over a 24-h test period. The initial uptake of the 100 nm nanoparticle into the collagen gel structure was superior compared to the larger sized nanoparticles under the influence of a magnetic field and incubated for 24 h. Interestingly, it was the 200 nm nanoparticles, which proved to penetrate the gel furthest, under the influence of a magnetic field, during the initial culture stage after 1-h incubation.
Collapse
Affiliation(s)
- Emily E L Lewis
- Centre for Cell Engineering, University of Glasgow, Glasgow, UK, G12 8QQ
| | - Hannah W Child
- Centre for Cell Engineering, University of Glasgow, Glasgow, UK, G12 8QQ
| | - Andrew Hursthouse
- School of Science, University of the West of Scotland, Paisley, UK, PA1 2BE
| | - David Stirling
- School of Science, University of the West of Scotland, Paisley, UK, PA1 2BE
| | - Mark McCully
- Centre for Cell Engineering, University of Glasgow, Glasgow, UK, G12 8QQ
| | - David Paterson
- Centre for Cell Engineering, University of Glasgow, Glasgow, UK, G12 8QQ
| | - Margaret Mullin
- Electron Microscopy Unit, University of Glasgow, Glasgow, UK, G12 8QQ
| | - Catherine C Berry
- Centre for Cell Engineering, University of Glasgow, Glasgow, UK, G12 8QQ
| |
Collapse
|
24
|
Barrefelt Å, Saghafian M, Kuiper R, Ye F, Egri G, Klickermann M, Brismar TB, Aspelin P, Muhammed M, Dähne L, Hassan M. Biodistribution, kinetics, and biological fate of SPION microbubbles in the rat. Int J Nanomedicine 2013; 8:3241-54. [PMID: 24023513 PMCID: PMC3767493 DOI: 10.2147/ijn.s49948] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
Background In the present investigation, we studied the kinetics and biodistribution of a contrast agent consisting of poly(vinyl alcohol) (PVA) microbubbles containing superparamagnetic iron oxide (SPION) trapped between the PVA layers (SPION microbubbles). Methods The biological fate of SPION microbubbles was determined in Sprague-Dawley rats after intravenous administration. Biodistribution and elimination of the microbubbles were studied in rats using magnetic resonance imaging for a period of 6 weeks. The rats were sacrificed and perfusion-fixated at different time points. The magnetic resonance imaging results obtained were compared with histopathologic findings in different organs. Results SPION microbubbles could be detected in the liver using magnetic resonance imaging as early as 10 minutes post injection. The maximum signal was detected between 24 hours and one week post injection. Histopathology showed the presence of clustered SPION microbubbles predominantly in the lungs from the first time point investigated (10 minutes). The frequency of microbubbles declined in the pulmonary vasculature and increased in pulmonary, hepatic, and splenic macrophages over time, resulting in a relative shift from the lungs to the spleen and liver. Meanwhile, macrophages showed increasing signs of cytoplasmic iron accumulation, initially in the lungs, then followed by other organs. Conclusion The present investigation highlights the biological behavior of SPION microbubbles, including organ distribution over time and indications for biodegradation. The present results are essential for developing SPION microbubbles as a potential contrast agent and/or a drug delivery vehicle for specific organs. Such a vehicle will facilitate the use of multimodality imaging techniques, including ultrasound, magnetic resonance imaging, and single positron emission computed tomography, and hence improve diagnostics, therapy, and the ability to monitor the efficacy of treatment.
Collapse
Affiliation(s)
- Åsa Barrefelt
- Department of Clinical Science, Intervention and Technology, Division of Medical Imaging and Technology, Karolinska Institutet, and Department of Radiology, Karolinska University Hospital-Huddinge, Stockholm, Sweden ; Experimental Cancer Medicine, Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Du X, Li W, Gao X, West MB, Saltzman WM, Cheng CJ, Stewart C, Zheng J, Cheng W, Kopke RD. Regeneration of mammalian cochlear and vestibular hair cells through Hes1/Hes5 modulation with siRNA. Hear Res 2013; 304:91-110. [PMID: 23850665 DOI: 10.1016/j.heares.2013.06.011] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/27/2012] [Revised: 05/16/2013] [Accepted: 06/27/2013] [Indexed: 12/31/2022]
Abstract
The Notch pathway is a cell signaling pathway determining initial specification and subsequent cell fate in the inner ear. Previous studies have suggested that new hair cells (HCs) can be regenerated in the inner ear by manipulating the Notch pathway. In the present study, delivery of siRNA to Hes1 and Hes5 using a transfection reagent or siRNA to Hes1 encapsulated within poly(lactide-co-glycolide acid) (PLGA) nanoparticles increased HC numbers in non-toxin treated organotypic cultures of cochleae and maculae of postnatal day 3 mouse pups. An increase in HCs was also observed in cultured cochleae and maculae of mouse pups pre-conditioned with a HC toxin (4-hydroxy-2-nonenal or neomycin) and then treated with the various siRNA formulations. Treating cochleae with siRNA to Hes1 associated with a transfection reagent or siRNA to Hes1 delivered by PLGA nanoparticles decreased Hes1 mRNA and up-regulated Atoh1 mRNA expression allowing supporting cells (SCs) to acquire a HC fate. Experiments using cochleae and maculae of p27(kip1)/-GFP transgenic mouse pups demonstrated that newly generated HCs trans-differentiated from SCs. Furthermore, PLGA nanoparticles are non-toxic to inner ear tissue, readily taken up by cells within the tissue of interest, and present a synthetic delivery system that is a safe alternative to viral vectors. These results indicate that when delivered using a suitable vehicle, Hes siRNAs are potential therapeutic molecules that may have the capacity to regenerate new HCs in the inner ear and possibly restore human hearing and balance function.
Collapse
Affiliation(s)
- Xiaoping Du
- Hough Ear Institute, P.O. Box 23206, Oklahoma City, OK 73112, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Whatley BR, Li X, Zhang N, Wen X. Magnetic-directed patterning of cell spheroids. J Biomed Mater Res A 2013; 102:1537-47. [DOI: 10.1002/jbm.a.34797] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2013] [Accepted: 04/30/2013] [Indexed: 12/18/2022]
Affiliation(s)
- Benjamin R. Whatley
- Department of Bioengineering; Clemson-MUSC Bioengineering Program; Clemson University; Clemson South Carolina 29634
| | - Xiaowei Li
- Department of Bioengineering; Clemson-MUSC Bioengineering Program; Clemson University; Clemson South Carolina 29634
| | - Ning Zhang
- Department of Bioengineering; Clemson-MUSC Bioengineering Program; Clemson University; Clemson South Carolina 29634
- Department of Regenerative Medicine & Cell Biology; Orthopaedic Surgery; Neuroscience; Dental Medicine; and Hollings Cancer Center; Medical University of South Carolina; Charleston South Carolina 29425
- Department of Biomedical Engineering; Virginia Commonwealth University; Richmond Virginia 23284
| | - Xuejun Wen
- Department of Bioengineering; Clemson-MUSC Bioengineering Program; Clemson University; Clemson South Carolina 29634
- Department of Regenerative Medicine & Cell Biology; Orthopaedic Surgery; Neuroscience; Dental Medicine; and Hollings Cancer Center; Medical University of South Carolina; Charleston South Carolina 29425
- Department of Chemical and Life Science Engineering; Virginia Commonwealth University; Richmond Virginia 23284
| |
Collapse
|
27
|
Liu H, Feng L, Tolia G, Liddell MR, Hao J, Li SK. Evaluation of intratympanic formulations for inner ear delivery: methodology and sustained release formulation testing. Drug Dev Ind Pharm 2013; 40:896-903. [PMID: 23631539 DOI: 10.3109/03639045.2013.789054] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
A convenient and efficient in vitro diffusion cell method to evaluate formulations for inner ear delivery via the intratympanic route is currently not available. The existing in vitro diffusion cell systems commonly used to evaluate drug formulations do not resemble the physical dimensions of the middle ear and round window membrane. The objectives of this study were to examine a modified in vitro diffusion cell system of a small diffusion area for studying sustained release formulations in inner ear drug delivery and to identify a formulation for sustained drug delivery to the inner ear. Four formulations and a control were examined in this study using cidofovir as the model drug. Drug release from the formulations in the modified diffusion cell system was slower than that in the conventional diffusion cell system due to the decrease in the diffusion surface area of the modified diffusion cell system. The modified diffusion cell system was able to show different drug release behaviors among the formulations and allowed formulation evaluation better than the conventional diffusion cell system. Among the formulations investigated, poly(lactic-co-glycolic acid)-poly(ethylene glycol)-poly(lactic-co-glycolic acid) triblock copolymer systems provided the longest sustained drug delivery, probably due to their rigid gel structures and/or polymer-to-cidofovir interactions.
Collapse
Affiliation(s)
- Hongzhuo Liu
- Division of Pharmaceutical Sciences, James L. Winkle College of Pharmacy, University of Cincinnati , Cincinnati, OH , USA
| | | | | | | | | | | |
Collapse
|
28
|
|
29
|
Min KA, Shin MC, Yu F, Yang M, David AE, Yang VC, Rosania GR. Pulsed magnetic field improves the transport of iron oxide nanoparticles through cell barriers. ACS NANO 2013; 7:2161-2171. [PMID: 23373613 PMCID: PMC3609927 DOI: 10.1021/nn3057565] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/27/2023]
Abstract
Understanding how a magnetic field affects the interaction of magnetic nanoparticles (MNPs) with cells is fundamental to any potential downstream applications of MNPs as gene and drug delivery vehicles. Here, we present a quantitative analysis of how a pulsed magnetic field influences the manner in which MNPs interact with and penetrate across a cell monolayer. Relative to a constant magnetic field, the rate of MNP uptake and transport across cell monolayers was enhanced by a pulsed magnetic field. MNP transport across cells was significantly inhibited at low temperature under both constant and pulsed magnetic field conditions, consistent with an active mechanism (i.e., endocytosis) mediating MNP transport. Microscopic observations and biochemical analysis indicated that, in a constant magnetic field, transport of MNPs across the cells was inhibited due to the formation of large (>2 μm) magnetically induced MNP aggregates, which exceeded the size of endocytic vesicles. Thus, a pulsed magnetic field enhances the cellular uptake and transport of MNPs across cell barriers relative to a constant magnetic field by promoting accumulation while minimizing magnetically induced MNP aggregation at the cell surface.
Collapse
Affiliation(s)
- Kyoung Ah Min
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Michigan, 428 Church St., Ann Arbor, MI 48109, USA
| | - Meong Cheol Shin
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Michigan, 428 Church St., Ann Arbor, MI 48109, USA
| | - Faquan Yu
- Key Laboratory for Green Chemical Process of Ministry of Education, Wuhan Institute of Technology, Wuhan 430073, China
| | - Meizhu Yang
- Key Laboratory for Green Chemical Process of Ministry of Education, Wuhan Institute of Technology, Wuhan 430073, China
| | - Allan E. David
- Department of Chemical Engineering, Auburn University, Auburn, AL 36849, USA
| | - Victor C. Yang
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Michigan, 428 Church St., Ann Arbor, MI 48109, USA
- Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnosis, School of Pharmacy, Tianjin Medical University, Tianjin 300070, China
| | - Gus R. Rosania
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Michigan, 428 Church St., Ann Arbor, MI 48109, USA
| |
Collapse
|
30
|
Sensenig R, Sapir Y, MacDonald C, Cohen S, Polyak B. Magnetic nanoparticle-based approaches to locally target therapy and enhance tissue regeneration in vivo. Nanomedicine (Lond) 2013; 7:1425-42. [PMID: 22994959 DOI: 10.2217/nnm.12.109] [Citation(s) in RCA: 134] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Magnetic-based systems utilizing superparamagnetic nanoparticles and a magnetic field gradient to exert a force on these particles have been used in a wide range of biomedical applications. This review is focused on drug targeting applications that require penetration of a cellular barrier as well as strategies to improve the efficacy of targeting in these biomedical applications. Another focus of this review is regenerative applications utilizing tissue engineered scaffolds prepared with the aid of magnetic particles, the use of remote actuation for release of bioactive molecules and magneto-mechanical cell stimulation, cell seeding and cell patterning.
Collapse
Affiliation(s)
- Richard Sensenig
- Department of Surgery, Drexel University College of Medicine, PA 19102, USA
| | | | | | | | | |
Collapse
|
31
|
Wahajuddin, Arora S. Superparamagnetic iron oxide nanoparticles: magnetic nanoplatforms as drug carriers. Int J Nanomedicine 2012; 7:3445-71. [PMID: 22848170 PMCID: PMC3405876 DOI: 10.2147/ijn.s30320] [Citation(s) in RCA: 553] [Impact Index Per Article: 46.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
A targeted drug delivery system is the need of the hour. Guiding magnetic iron oxide nanoparticles with the help of an external magnetic field to its target is the principle behind the development of superparamagnetic iron oxide nanoparticles (SPIONs) as novel drug delivery vehicles. SPIONs are small synthetic γ-Fe2O3 (maghemite) or Fe3O4 (magnetite) particles with a core ranging between 10 nm and 100 nm in diameter. These magnetic particles are coated with certain biocompatible polymers, such as dextran or polyethylene glycol, which provide chemical handles for the conjugation of therapeutic agents and also improve their blood distribution profile. The current research on SPIONs is opening up wide horizons for their use as diagnostic agents in magnetic resonance imaging as well as for drug delivery vehicles. Delivery of anticancer drugs by coupling with functionalized SPIONs to their targeted site is one of the most pursued areas of research in the development of cancer treatment strategies. SPIONs have also demonstrated their efficiency as nonviral gene vectors that facilitate the introduction of plasmids into the nucleus at rates multifold those of routinely available standard technologies. SPION-induced hyperthermia has also been utilized for localized killing of cancerous cells. Despite their potential biomedical application, alteration in gene expression profiles, disturbance in iron homeostasis, oxidative stress, and altered cellular responses are some SPION-related toxicological aspects which require due consideration. This review provides a comprehensive understanding of SPIONs with regard to their method of preparation, their utility as drug delivery vehicles, and some concerns which need to be resolved before they can be moved from bench top to bedside.
Collapse
Affiliation(s)
- Wahajuddin
- Pharmacokinetics and Metabolism Division, CSIR-Central Drug Research Institute, Lucknow, Uttar Pradesh, India.
| | | |
Collapse
|
32
|
Eustaquio T, Leary JF. Single-cell nanotoxicity assays of superparamagnetic iron oxide nanoparticles. Methods Mol Biol 2012; 926:69-85. [PMID: 22975957 DOI: 10.1007/978-1-62703-002-1_5] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Properly evaluating the nanotoxicity of nanoparticles involves much more than bulk-cell assays of cell death by necrosis. Cells exposed to nanoparticles may undergo repairable oxidative stress and DNA damage or be induced into apoptosis. Exposure to nanoparticles may cause the cells to alter their proliferation or differentiation or their cell-cell signaling with neighboring cells in a tissue. Nanoparticles are usually more toxic to some cell subpopulations than others, and toxicity often varies with cell cycle. All of these facts dictate that any nanotoxicity assay must be at the single-cell level and must try whenever feasible and reasonable to include many of these other factors. Focusing on one type of quantitative measure of nanotoxicity, we describe flow and scanning image cytometry approaches to measuring nanotoxicity at the single-cell level by using a commonly used assay for distinguishing between necrotic and apoptotic causes of cell death by one type of nanoparticle. Flow cytometry is fast and quantitative, provided that the cells can be prepared into a single-cell suspension for analysis. But when cells cannot be put into suspension without altering nanotoxicity results, or if morphology, attachment, and stain location are important, a scanning image cytometry approach must be used. Both methods are described with application to a particular type of nanoparticle, a superparamagnetic iron oxide nanoparticle (SPION), as an example of how these assays may be applied to the more general problem of determining the effects of nanomaterial exposure to living cells.
Collapse
Affiliation(s)
- Trisha Eustaquio
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN, USA
| | | |
Collapse
|
33
|
Plank C, Zelphati O, Mykhaylyk O. Magnetically enhanced nucleic acid delivery. Ten years of magnetofection-progress and prospects. Adv Drug Deliv Rev 2011; 63:1300-31. [PMID: 21893135 PMCID: PMC7103316 DOI: 10.1016/j.addr.2011.08.002] [Citation(s) in RCA: 251] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2011] [Revised: 08/18/2011] [Accepted: 08/19/2011] [Indexed: 12/28/2022]
Abstract
Nucleic acids carry the building plans of living systems. As such, they can be exploited to make cells produce a desired protein, or to shut down the expression of endogenous genes or even to repair defective genes. Hence, nucleic acids are unique substances for research and therapy. To exploit their potential, they need to be delivered into cells which can be a challenging task in many respects. During the last decade, nanomagnetic methods for delivering and targeting nucleic acids have been developed, methods which are often referred to as magnetofection. In this review we summarize the progress and achievements in this field of research. We discuss magnetic formulations of vectors for nucleic acid delivery and their characterization, mechanisms of magnetofection, and the application of magnetofection in viral and nonviral nucleic acid delivery in cell culture and in animal models. We summarize results that have been obtained with using magnetofection in basic research and in preclinical animal models. Finally, we describe some of our recent work and end with some conclusions and perspectives.
Collapse
|
34
|
Mondalek FG, Ashley RA, Roth CC, Kibar Y, Shakir N, Ihnat MA, Fung KM, Grady BP, Kropp BP, Lin HK. Enhanced angiogenesis of modified porcine small intestinal submucosa with hyaluronic acid-poly(lactide-co-glycolide) nanoparticles: from fabrication to preclinical validation. J Biomed Mater Res A 2010; 94:712-9. [PMID: 20213816 DOI: 10.1002/jbm.a.32748] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
Hyaluronic acid-poly(de-co-glycolide) nanoparticles (HA-PLGA NPs) were synthesized to stabilize the porous structure of porcine small intestinal submucosa (SIS), to improve surface biocompatibility and to enhance performance in tissue regeneration. HA-PLGA NPs were characterized for size, zeta potential, surface morphology, and HA loading. Human microvascular endothelial cells responded to HA-PLGA NPs and HA-PLGA modified SIS (HA-PLGA-SIS) with elevated cell proliferation. HA-PLGA-SIS significantly enhanced neo-vascularization in an in ovo chorioallantoic membrane angiogenesis model. The angiogenic capability of the newly fabricated HA-PLGA-SIS was tested in a canine bladder augmentation model. Urinary bladder augmentation was performed in beagle dogs following hemi-cystectomy using HA-PLGA-SIS. The regenerated bladder was harvested at 10 weeks post augmentation and vascularization was evaluated using CD31 immunohistochemical staining. Bladder regenerated with HA-PLGA-SIS had significantly higher vascular ingrowth compared to unmodified SIS. This study shows that HA-PLGA NPs may represent a new approach for modifying naturally derived SIS biomaterials in regenerative medicine.
Collapse
Affiliation(s)
- Fadee G Mondalek
- Department of Chemical, Biological and Materials Engineering, University of Oklahoma, Norman, Oklahoma 73019, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Gao X, Wang Y, Chen K, Grady BP, Dormer KJ, Kopke RD. Magnetic Assisted Transport of PLGA Nanoparticles Through a Human Round Window Membrane Model. J Nanotechnol Eng Med 2010. [DOI: 10.1115/1.4002043] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
The lack of an effective method for inner ear drug delivery is a clinical problem for the prevention and treatment of hearing loss. With technology advances in nanomedicine and the use of hydrogels, more drug delivery options are becoming available. This study tested the feasibility of using a tripartite layer round window membrane (RWM) model to evaluate the effectiveness of a magnetic assisted transport of poly(lactic-co-glycolic acid) (PLGA)/superparamagnetic iron oxide nanoparticles (SPIONs). A RWM model was constructed as a three-cell-layer model with epithelial cells cultured on both sides of a small intestinal submucosal (SIS) matrix with fibroblasts seeded within the matrix. PLGA encapsulated coumarin-6/SPION nanoparticles 100 nm in diameter were formulated by an oil-in-water emulsion/solvent evaporation method and pulled through the RWM model using permanent magnets with a flux density 0.410 T at the pole face. Independent variables such as external magnetic force and exposure time, composition of hyaluronic acid (HA) hydrogel suspending media, and particle characteristics including magnetic susceptibility were studied. Magnetic assisted transport of coumarin-6 labeled magnetic nanoparticles through the RWM inserts increased 2.1-fold in 1 h compared with the controls. HA hydrogel did prevent particle accumulation on the surface of RWM in a magnetic field but also impaired the mobility of these particles. Greater particle susceptibility or stronger external magnetic fields did not significantly improve the transmembrane transport. A RWM model was designed consisting of a SIS membrane and three co-cultured layers of cells, which was structurally and physically similar to the human. PLGA particles (100 nm) with encapsulated ∼15 nm SPIONs were transported through this model with the assistance of an external magnet, allowing quantitative evaluation of prospective targeted drug delivery through the RWM via the assistance of a magnetic field.
Collapse
Affiliation(s)
- Xinsheng Gao
- Hough Ear Institute, INTEGRIS Health, 3400 Northwest 56th Street, Oklahoma City, OK 73112
| | - Youdan Wang
- Hough Ear Institute, INTEGRIS Health, 3400 Northwest 56th Street, Oklahoma City, OK 73112
| | - Kejian Chen
- Hough Ear Institute, INTEGRIS Health, 3400 Northwest 56th Street, Oklahoma City, OK 73112
| | - Brian P. Grady
- School of Chemical, Biological and Materials Engineering, University of Oklahoma, 100 East Boyd, Norman, OK 73019
| | - Kenneth J. Dormer
- Department of Physiology, University of Oklahoma Health Sciences Center, 940 Stanton L. Young Boulevard, Oklahoma City, OK 73104
| | - Richard D. Kopke
- Hough Ear Institute, INTEGRIS Health, 3400 Northwest 56th Street, Oklahoma City, OK 73112
| |
Collapse
|
36
|
Min KA, Yu F, Yang VC, Zhang X, Rosania GR. Transcellular Transport of Heparin-coated Magnetic Iron Oxide Nanoparticles (Hep-MION) Under the Influence of an Applied Magnetic Field. Pharmaceutics 2010; 2:119-135. [PMID: 21152371 PMCID: PMC2997712 DOI: 10.3390/pharmaceutics2020119] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
In this study, magnetic iron oxide nanoparticles coated with heparin (Hep-MION) were synthesized and the transcellular transport of the nanoparticles across epithelial cell monolayers on porous polyester membranes was investigated. An externally applied magnetic field facilitated the transport of the Hep-MION across cell monolayers. However, high Hep-MION concentrations led to an increased aggregation of nanoparticles on the cell monolayer after application of the magnetic field. Our results indicate that magnetic guidance of Hep-MION most effectively promotes transcellular transport under conditions that minimize formation of magnetically-induced nanoparticle aggregates. Across cell monolayers, the magnet’s attraction led to the greatest increase in mass transport rate in dilute dispersions and in high serum concentrations, suggesting that magnetic guidance may be useful for in vivo targeting of Hep-MION.
Collapse
Affiliation(s)
- Kyoung Ah Min
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Michigan, 428 Church St., Ann Arbor, MI 48109, USA; E-Mails: (K.A.M.); (F.Y.); (V.C.Y.); (X.Z.)
| | - Faquan Yu
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Michigan, 428 Church St., Ann Arbor, MI 48109, USA; E-Mails: (K.A.M.); (F.Y.); (V.C.Y.); (X.Z.)
- Key Laboratory for Green Chemical Process of Ministry of Education, Wuhan Institute of Technology, Wuhan 430073, China
| | - Victor C. Yang
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Michigan, 428 Church St., Ann Arbor, MI 48109, USA; E-Mails: (K.A.M.); (F.Y.); (V.C.Y.); (X.Z.)
- Tianjin Key Laboratory for Modern Drug Delivery and High Efficiency, Tianjin University, Tianjin 300072, China
| | - Xinyuan Zhang
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Michigan, 428 Church St., Ann Arbor, MI 48109, USA; E-Mails: (K.A.M.); (F.Y.); (V.C.Y.); (X.Z.)
| | - Gus R. Rosania
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Michigan, 428 Church St., Ann Arbor, MI 48109, USA; E-Mails: (K.A.M.); (F.Y.); (V.C.Y.); (X.Z.)
- Author to whom correspondence should be addressed; E-Mail: ; Tel.: +1-734-763-1032; Fax: +1-734-615-6162
| |
Collapse
|
37
|
MacDonald C, Friedman G, Alamia J, Barbee K, Polyak B. Time-varied magnetic field enhances transport of magnetic nanoparticles in viscous gel. Nanomedicine (Lond) 2010; 5:65-76. [PMID: 20025465 DOI: 10.2217/nnm.09.97] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
AIM The potential of magnetic nanoparticles (MNPs) to deliver various forms of therapy has not been fully realized, in part due to difficulties in transporting the carriers through soft tissue to different target sites. The aim of this study was to demonstrate that transport of MNPs through a viscous gel can be controlled by a combined AC (time-varying) magnetic field and static field gradient. MATERIALS & METHODS MNP velocity and transport efficiency were measured in a viscous gel at various settings of magnetic field and magnetite loadings. RESULTS Combined application of an AC magnetic field with the static field gradient resulted in a nearly 30-fold increase in MNP transport efficiency in viscous gel for 30% (w/w) magnetite-loaded particles as compared with static field conditions. CONCLUSION The 'oscillating' effect of an AC magnetic field greatly improves the ability to transport MNPs within soft media by decreasing the effective viscosity of the gel.
Collapse
Affiliation(s)
- Cristin MacDonald
- School of Biomedical Engineering, Drexel University Philadelphia, PA 19104, USA
| | | | | | | | | |
Collapse
|
38
|
Abstract
Nonviral gene delivery has been gaining considerable attention recently. Although the efficacy of DNA transfection, which is a major concern, is low in nonviral vector-mediated gene transfer compared with viral ones, nonviral vectors are relatively easy to prepare, less immunogenic and oncogenic, and have no potential of virus recombination and no limitation on the size of a transferred gene. The ability to incorporate genetic materials such as plasmid DNA, RNA, and siRNA into functionalized nanoparticles with little toxicity demonstrates a new era in pharmacotherapy for delivering genes selectively to tissues and cells. In this chapter, we highlight the basic concepts and applications of nonviral gene delivery using super paramagnetic iron oxide nanoparticles and functionalized silica nanoparticles. The experimental protocols related to these topics are described in the chapter.
Collapse
Affiliation(s)
- Sha Jin
- College of Engineering, University of Arkansas, Fayetteville, AR, USA
| | | | | |
Collapse
|
39
|
Williams PS, Carpino F, Zborowski M. Magnetic nanoparticle drug carriers and their study by quadrupole magnetic field-flow fractionation. Mol Pharm 2009; 6:1290-306. [PMID: 19591456 PMCID: PMC2757515 DOI: 10.1021/mp900018v] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Magnetic nanoparticle drug carriers continue to attract considerable interest for drug targeting in the treatment of cancers and other pathological conditions. The efficient delivery of therapeutic levels of drug to a target site while limiting nonspecific, systemic toxicity requires optimization of the drug delivery materials, the applied magnetic field, and the treatment protocol. The history and current state of magnetic drug targeting is reviewed. While initial studies involved micrometer-sized and larger carriers, and work with these microcarriers continues, it is the sub-micrometer carriers or nanocarriers that are of increasing interest. An aspect of magnetic drug targeting using nanoparticle carriers that has not been considered is then addressed. This aspect involves the variation in the magnetic properties of the nanocarriers. Quadrupole magnetic field-flow fractionation (QMgFFF) is a relatively new technique for characterizing magnetic nanoparticles. It is unique in its capability of determining the distribution in magnetic properties of a nanoparticle sample in suspension. The development and current state of this technique is also reviewed. Magnetic nanoparticle drug carriers have been found by QMgFFF analysis to be highly polydisperse in their magnetic properties, and the strength of response of the particles to magnetic field gradients is predicted to vary by orders of magnitude. It is expected that the least magnetic fraction of a formulation will contribute the most to systemic toxicity, and the depletion of this fraction will result in a more effective drug carrying material. A material that has a reduced systemic toxicity will allow higher doses of cytotoxic drugs to be delivered to the tumor with reduced side effects. Preliminary experiments involving a novel method of refining a magnetic nanoparticle drug carrier to achieve this result are described. QMgFFF is used to characterize the refined and unrefined material.
Collapse
Affiliation(s)
- P Stephen Williams
- Department of Biomedical Engineering, Lerner Research Institute, Cleveland Clinic, 9500 Euclid Avenue, Cleveland, Ohio 44195, USA.
| | | | | |
Collapse
|
40
|
Mondalek FG, Lawrence BJ, Kropp BP, Grady BP, Fung KM, Madihally SV, Lin HK. The incorporation of poly(lactic-co-glycolic) acid nanoparticles into porcine small intestinal submucosa biomaterials. Biomaterials 2008; 29:1159-66. [PMID: 18076986 DOI: 10.1016/j.biomaterials.2007.11.020] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2007] [Accepted: 11/15/2007] [Indexed: 11/18/2022]
Abstract
Small intestinal submucosa (SIS) derived from porcine small intestine has been intensively studied for its capacity in repairing and regenerating wounded and dysfunctional tissues. However, SIS suffers from a large spectrum of heterogeneity in microarchitecture leading to inconsistent results. In this study, we introduced nanoparticles (NPs) to SIS with an intention of decreasing the heterogeneity and improving the consistency of this biomaterial. As determined by scanning electron microscopy and urea permeability, the optimum NP size was estimated to be between 200 nm and 500 nm using commercial monodisperse latex spheres. The concentration of NPs that is required to alter pore sizes of SIS as determined by urea permeability was estimated to be 1 mg/ml 260 nm poly(lactic-co-glycolic) acid (PLGA) NPs. The 1mg/ml PLGA NPs loaded in the SIS did not change the tensile properties of the unmodified SIS or even alter pH values in a cell culture environment. More importantly, PLGA NP modified SIS did not affect human mammary endothelial cells (HMEC-1) morphology or adhesion, but actually enhanced HEMC-1 cell growth.
Collapse
Affiliation(s)
- Fadee G Mondalek
- Department of Chemical, Biological and Materials Engineering, University of Oklahoma, Norman, OK 73019, USA
| | | | | | | | | | | | | |
Collapse
|
41
|
Soloviev M. Nanobiotechnology today: focus on nanoparticles. J Nanobiotechnology 2007; 5:11. [PMID: 18163916 PMCID: PMC2211307 DOI: 10.1186/1477-3155-5-11] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2007] [Accepted: 12/30/2007] [Indexed: 12/16/2022] Open
Abstract
In the recent years the nanobiotechnology field and the Journal of Nanobiotechnology readership have witnessed an increase in interest towards the nanoparticles and their biological effects and applications. These include bottom-up and molecular self-assembly, biological effects of naked nanoparticles and nano-safety, drug encapsulation and nanotherapeutics, and novel nanoparticles for use in microscopy, imaging and diagnostics. This review highlights recent Journal of Nanobiotechnology publications in some of these areas .
Collapse
Affiliation(s)
- Mikhail Soloviev
- Royal Holloway University of London, Egham, Surrey, TW20 0EX, UK.
| |
Collapse
|
42
|
Ji X, Shao R, Elliott AM, Stafford RJ, Esparza-Coss E, Bankson JA, Liang G, Luo ZP, Park K, Markert JT, Li C. Bifunctional Gold Nanoshells with a Superparamagnetic Iron Oxide-Silica Core Suitable for Both MR Imaging and Photothermal Therapy. THE JOURNAL OF PHYSICAL CHEMISTRY. C, NANOMATERIALS AND INTERFACES 2007; 111:6245-6251. [PMID: 20165552 PMCID: PMC2822349 DOI: 10.1021/jp0702245] [Citation(s) in RCA: 146] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/20/2023]
Abstract
We describe the synthesis, characterization, and use of hybrid nanoparticles with a superparamagnetic iron oxide (SPIO) core and a gold nanoshell. These multifunctional nanoparticles, designated SPIO-Au nanoshells, displayed superparamagnetic characteristics and a significant absorbance in the near-infrared (NIR) region of the electromagnetic spectrum. In addition, they exhibited high transverse relaxivity, r2 , and a large r2/r1 ratio and therefore could be imaged by MRI to obtain T2-weighted images. Moreover, SPIO-Au nanoshells showed efficient photo-thermal effect when exposed to NIR light. The use of SPIO-Au nanoshells, with their combination of unique magnetic and optical properties, should enhance the efficacy of nanoshell-mediated photo-thermal therapy by making it possible to direct more nanoparticles to tumors through the application of external magnetic field and by permitting real-time in vivo MRI imaging of the distribution of the nanoparticles before, during, and after photo-thermal therapy.
Collapse
Affiliation(s)
- Xiaojun Ji
- Department of Experimental Diagnostic Imaging, The University of Texas M. D. Anderson Cancer Center, Houston, Texas 77030
| | - Ruping Shao
- Department of Experimental Diagnostic Imaging, The University of Texas M. D. Anderson Cancer Center, Houston, Texas 77030
| | - Andrew M. Elliott
- Department of Imaging Physics, The University of Texas M. D. Anderson Cancer Center, Houston, Texas 77030
| | - R. Jason Stafford
- Department of Imaging Physics, The University of Texas M. D. Anderson Cancer Center, Houston, Texas 77030
| | - Emilio Esparza-Coss
- Department of Imaging Physics, The University of Texas M. D. Anderson Cancer Center, Houston, Texas 77030
| | - James A. Bankson
- Department of Imaging Physics, The University of Texas M. D. Anderson Cancer Center, Houston, Texas 77030
| | - Gan Liang
- Department of Physics, Sam Houston State University, Huntsville, Texas 77341
| | - Zhi-Ping Luo
- Microscopy and Imaging Center, Texas A&M University, College Station, Texas 77843
| | - Keeseong Park
- Department of Physics, The University of Texas at Austin, Austin, Texas 78712
| | - John T. Markert
- Department of Physics, The University of Texas at Austin, Austin, Texas 78712
| | - Chun Li
- Department of Experimental Diagnostic Imaging, The University of Texas M. D. Anderson Cancer Center, Houston, Texas 77030
- Corresponding author: Chun Li, PhD, Department of Experimental Diagnostic Imaging-Box 57, The University of Texas M. D. Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, Texas 77030. Phone: (713) 792-5182. Fax: (713) 794-5456.
| |
Collapse
|
43
|
Barnes AL, Wassel RA, Mondalek F, Chen K, Dormer KJ, Kopke RD. Magnetic characterization of superparamagnetic nanoparticles pulled through model membranes. BIOMAGNETIC RESEARCH AND TECHNOLOGY 2007; 5:1. [PMID: 17204157 PMCID: PMC1785374 DOI: 10.1186/1477-044x-5-1] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/18/2006] [Accepted: 01/04/2007] [Indexed: 11/10/2022]
Abstract
BACKGROUND To quantitatively compare in-vitro and in vivo membrane transport studies of targeted delivery, one needs characterization of the magnetically-induced mobility of superparamagnetic iron oxide nanoparticles (SPION). Flux densities, gradients, and nanoparticle properties were measured in order to quantify the magnetic force on the SPION in both an artificial cochlear round window membrane (RWM) model and the guinea pig RWM. METHODS Three-dimensional maps were created for flux density and magnetic gradient produced by a 24-well casing of 4.1 kilo-Gauss neodymium-iron-boron (NdFeB) disc magnets. The casing was used to pull SPION through a three-layer cell culture RWM model. Similar maps were created for a 4 inch (10.16 cm) cube 48 MGOe NdFeB magnet used to pull polymeric-nanoparticles through the RWM of anesthetized guinea pigs. Other parameters needed to compute magnetic force were nanoparticle and polymer properties, including average radius, density, magnetic susceptibility, and volume fraction of magnetite. RESULTS A minimum force of 5.04 x 10(-16) N was determined to adequately pull nanoparticles through the in-vitro model. For the guinea pig RWM, the magnetic force on the polymeric nanoparticles was 9.69 x 10-20 N. Electron microscopy confirmed the movement of the particles through both RWM models. CONCLUSION As prospective carriers of therapeutic substances, polymers containing superparamagnetic iron oxide nanoparticles were succesfully pulled through the live RWM. The force required to achieve in vivo transport was significantly lower than that required to pull nanoparticles through the in-vitro RWM model. Indeed very little force was required to accomplish measurable delivery of polymeric-SPION composite nanoparticles across the RWM, suggesting that therapeutic delivery to the inner ear by SPION is feasible.
Collapse
Affiliation(s)
- Allison L Barnes
- Department of Physiology, College of Medicine, University of Oklahoma Health Sciences Center, 940 S.L. Young Blvd., Oklahoma City, OK 73104-0505, USA
| | - Ronald A Wassel
- Hough Ear Institute, 3400 N.W. 56Street, Oklahoma City, OK 73112, USA
| | - Fadee Mondalek
- School of Chemical, Biological & Materials Engineering, University of Oklahoma 100 East Boyd EC, Norman, OK 73019, USA
| | - Kejian Chen
- Hough Ear Institute, 3400 N.W. 56Street, Oklahoma City, OK 73112, USA
| | - Kenneth J Dormer
- Department of Physiology, College of Medicine, University of Oklahoma Health Sciences Center, 940 S.L. Young Blvd., Oklahoma City, OK 73104-0505, USA
- Hough Ear Institute, 3400 N.W. 56Street, Oklahoma City, OK 73112, USA
| | - Richard D Kopke
- Hough Ear Institute, 3400 N.W. 56Street, Oklahoma City, OK 73112, USA
| |
Collapse
|