151
|
Barth C, Spreen H, Mulac D, Keuter L, Behrens M, Humpf HU, Langer K. Spacer length and serum protein adsorption affect active targeting of trastuzumab-modified nanoparticles. BIOMATERIALS AND BIOSYSTEMS 2021; 5:100032. [PMID: 36825111 PMCID: PMC9934468 DOI: 10.1016/j.bbiosy.2021.100032] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Revised: 11/15/2021] [Accepted: 12/09/2021] [Indexed: 12/15/2022] Open
Abstract
Receptor-mediated active targeting of nanocarriers is a widely investigated approach to specifically address cancerous cells and tissues in the human body. The idea is to use these formulations as drug carriers with enhanced specificity and therefore reduced systemic side effects. Until today a big obstacle to reach this goal remains the adsorption of serum proteins to the nanocarrier's surface after contact with biological fluids. In this context different nanoparticle characteristics could be beneficial for effective active targeting after formation of a protein corona which need to be identified. In this study trastuzumab was used as an active targeting ligand which was covalently attached to human serum albumin nanoparticles. For coupling reaction different molecular weight spacers were used and resulting physicochemical nanoparticle characteristics were evaluated. The in vitro cell association of the different nanoparticle formulations was tested in cell culture experiments with or without fetal bovine serum. For specific receptor-mediated cell interaction SK-BR-3 breast cancer cells with human epidermal growth factor receptor 2 (HER2) overexpression were used. MCF-7 breast cancer cells with normal HER2 expression served as control. Furthermore, serum protein adsorption on respective nanoparticles was characterized. The qualitative and quantitative composition of the protein corona was analyzed by SDS-PAGE and LC-MS/MS and the influence of protein adsorption on active targeting capability was determined.
Collapse
Affiliation(s)
- Christina Barth
- Institute of Pharmaceutical Technology and Biopharmacy, University of Muenster, Corrensstr. 48, 48149 Muenster, Germany
| | - Hendrik Spreen
- Institute of Pharmaceutical Technology and Biopharmacy, University of Muenster, Corrensstr. 48, 48149 Muenster, Germany
| | - Dennis Mulac
- Institute of Pharmaceutical Technology and Biopharmacy, University of Muenster, Corrensstr. 48, 48149 Muenster, Germany
| | - Lucas Keuter
- Institute of Food Chemistry, University of Muenster, Corrensstr. 45, 48149 Muenster, Germany
| | - Matthias Behrens
- Institute of Food Chemistry, University of Muenster, Corrensstr. 45, 48149 Muenster, Germany
| | - Hans-Ulrich Humpf
- Institute of Food Chemistry, University of Muenster, Corrensstr. 45, 48149 Muenster, Germany
| | - Klaus Langer
- Institute of Pharmaceutical Technology and Biopharmacy, University of Muenster, Corrensstr. 48, 48149 Muenster, Germany,To whom correspondence should be addressed.
| |
Collapse
|
152
|
Nandakumar A, Wei W, Siddiqui G, Li Y, Kakinen A, Wan X, Koppel K, Lin S, Davis TP, Leong DT, Creek DJ, Song Y, Ke PC. Dynamic Protein Corona of Gold Nanoparticles with an Evolving Morphology. ACS APPLIED MATERIALS & INTERFACES 2021; 13:58238-58251. [PMID: 34797630 PMCID: PMC8692073 DOI: 10.1021/acsami.1c19824] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/26/2023]
Abstract
Much has been learned about the protein coronae and their biological implications within the context of nanomedicine and nanotoxicology. However, no data is available about the protein coronae associated with nanoparticles undergoing spontaneous surface-energy minimization, a common phenomenon during the synthesis and shelf life of nanomaterials. Accordingly, here we employed gold nanoparticles (AuNPs) possessing the three initial states of spiky, midspiky, and spherical shapes and determined their acquisition of human plasma protein coronae with label-free mass spectrometry. The AuNPs collected coronal proteins that were different in abundance, physicochemical parameters, and interactive biological network. The size and structure of the coronal proteins matched the morphology of the AuNPs, where small globular proteins and large fibrillar proteins were enriched on spiky AuNPs, while large proteins were abundant on spherical AuNPs. Furthermore, the AuNPs induced endothelial leakiness to different degrees, which was partially negated by their protein coronae as revealed by confocal fluorescence microscopy, in vitro and ex vivo transwell assays, and signaling pathway assays. This study has filled a knowledge void concerning the dynamic protein corona of nanoparticles possessing an evolving morphology and shed light on their implication for future nanomedicine harnessing the paracellular pathway.
Collapse
Affiliation(s)
- Aparna Nandakumar
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, VIC 3052, Australia
| | - Wei Wei
- Key Laboratory of Luminescence Analysis and Molecular Sensing, Ministry of Education, College of Food Science, Southwest University, 2 Tiansheng Rd, Beibei District, Chongqing, 400715, China
| | - Ghizal Siddiqui
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, VIC 3052, Australia
| | - Yuhuan Li
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, VIC 3052, Australia
- Liver Cancer Institute, Zhongshan Hospital, Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Fudan University, Shanghai, 200032, China
| | - Aleksandr Kakinen
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane Qld 4072, Australia
| | - Xulin Wan
- Key Laboratory of Luminescence Analysis and Molecular Sensing, Ministry of Education, College of Food Science, Southwest University, 2 Tiansheng Rd, Beibei District, Chongqing, 400715, China
| | - Kairi Koppel
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, VIC 3052, Australia
| | - Sijie Lin
- College of Environmental Science and Engineering, Biomedical Multidisciplinary Innovation Research Institute, Shanghai East Hospital, Shanghai Institute of Pollution Control and Ecological Security, Tongji University, 1239 Siping Road, Shanghai 200092, China
| | - Thomas P. Davis
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, VIC 3052, Australia
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane Qld 4072, Australia
| | - David T. Leong
- Department of Chemical and Biomolecular Engineering, National University of Singapore, 4 Engineering Drive 4, Singapore 117585, Singapore
| | - Darren J. Creek
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, VIC 3052, Australia
| | - Yang Song
- Key Laboratory of Luminescence Analysis and Molecular Sensing, Ministry of Education, College of Pharmaceutical Sciences, Southwest University, 2 Tiansheng Rd, Beibei District, Chongqing 400715, China
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing, 100085, China
| | - Pu Chun Ke
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, VIC 3052, Australia
- The GBA National Institute for Nanotechnology Innovation, 136 Kaiyuan Avenue, Guangzhou, 510700, China
| |
Collapse
|
153
|
Andrian T, Pujals S, Albertazzi L. Quantifying the effect of PEG architecture on nanoparticle ligand availability using DNA-PAINT. NANOSCALE ADVANCES 2021; 3:6876-6881. [PMID: 34977461 PMCID: PMC8650147 DOI: 10.1039/d1na00696g] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Accepted: 11/01/2021] [Indexed: 06/14/2023]
Abstract
The importance of PEG architecture on nanoparticle (NP) functionality is known but still difficult to investigate, especially at a single particle level. Here, we apply DNA Point Accumulation for Imaging in Nanoscale Topography (DNA-PAINT), a super-resolution microscopy (SRM) technique, to study the surface functionality in poly(lactide-co-glycolide)-poly(ethylene glycol) (PLGA-PEG) NPs with different PEG structures. We demonstrated how the length of the PEG spacer can influence the accessibility of surface chemical functionality, highlighting the importance of SRM techniques to support the rational design of functionalized NPs.
Collapse
Affiliation(s)
- Teodora Andrian
- Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology Baldiri Reixac 15-21 08028 Barcelona Spain
| | - Silvia Pujals
- Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology Baldiri Reixac 15-21 08028 Barcelona Spain
| | - Lorenzo Albertazzi
- Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology Baldiri Reixac 15-21 08028 Barcelona Spain
- Department of Biomedical Engineering, Institute for Complex Molecular Systems (ICMS), Eindhoven University of Technology 5612AZ Eindhoven The Netherlands
| |
Collapse
|
154
|
The Interactions between Nanoparticles and the Innate Immune System from a Nanotechnologist Perspective. NANOMATERIALS 2021; 11:nano11112991. [PMID: 34835755 PMCID: PMC8621168 DOI: 10.3390/nano11112991] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Revised: 11/02/2021] [Accepted: 11/03/2021] [Indexed: 12/24/2022]
Abstract
The immune system contributes to maintaining the body’s functional integrity through its two main functions: recognizing and destroying foreign external agents (invading microorganisms) and identifying and eliminating senescent cells and damaged or abnormal endogenous entities (such as cellular debris or misfolded/degraded proteins). Accordingly, the immune system can detect molecular and cellular structures with a spatial resolution of a few nm, which allows for detecting molecular patterns expressed in a great variety of pathogens, including viral and bacterial proteins and bacterial nucleic acid sequences. Such patterns are also expressed in abnormal cells. In this context, it is expected that nanostructured materials in the size range of proteins, protein aggregates, and viruses with different molecular coatings can engage in a sophisticated interaction with the immune system. Nanoparticles can be recognized or passed undetected by the immune system. Once detected, they can be tolerated or induce defensive (inflammatory) or anti-inflammatory responses. This paper describes the different modes of interaction between nanoparticles, especially inorganic nanoparticles, and the immune system, especially the innate immune system. This perspective should help to propose a set of selection rules for nanosafety-by-design and medical nanoparticle design.
Collapse
|
155
|
Orellana N, Palma S, Torres E, Cordero ML, Vio V, Ruso JM, Juárez J, Topete A, Araya E, Vasquez-Contreras R, Kogan MJ, Hassan N. Study of the interaction of folic acid-modified gold nanorods and fibrinogen through microfluidics: implications for protein adsorption, incorporation and viability of cancer cells. NANOSCALE 2021; 13:17807-17821. [PMID: 34668502 DOI: 10.1039/d1nr03179a] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Gold nanoparticles (GNPs) are an attractive nanomaterial for potential applications in therapy and diagnostics due to their capability to direct toward specific sites in the organism. However, when exposed to plasma, GNPs can interact with different biomolecules that form a dynamic nano-bio interface called a "protein corona" (PC). Remarkably, the PC could affect multiple biological processes, such as cell targeting and uptake, cytotoxicity, and nanoparticle (NP) clearance. The interaction of nanomaterials with plasmatic proteins has been widely studied under bulk conditions, however, under dynamic conditions, it has just recently been explored. Thus, to mimic a dynamic natural environment found in arteries and veins, microfluidic devices were used. In this work, gold nanorods (GNRs) were synthesized and conjugated with polyethylene glycol (PEG) to reduce their interaction with plasma proteins and increase their biocompatibility. Then, GNRs were functionalized with folic acid, a targeting ligand typically used to recognize tumor cells. The resulting nanosystem was exposed to fibrinogen (FB) to study the development and biological impact of PC formation through two strategies: bulk and laminar flow conditions. The obtained nanosystems were characterized by absorption spectrophotometry, DLS, laser Doppler microelectrophoresis, neutron activation analysis, circular dichroism spectroscopy and TEM. Finally, cell viability and cellular uptake assays were performed to study the influence of the PC on the cell viability and delivery of nanosystems.
Collapse
Affiliation(s)
- Nacaroha Orellana
- Programa Institucional de Fomento a la I+D+I, Universidad Tecnológica Metropolitana, Ignacio Valdivieso 2409, San Joaquín, Chile.
| | - Sujey Palma
- Programa Institucional de Fomento a la I+D+I, Universidad Tecnológica Metropolitana, Ignacio Valdivieso 2409, San Joaquín, Chile.
- Departamento de Química Farmacológica y Toxicológica, Facultad de Ciencias Químicas y Farmacéuticas, Laboratorio de Nanobiotecnología, Universidad de Chile, Santos Dumont 964, Independencia, Chile.
- Advanced Center for Chronic Diseases (ACCDis), Santos Dumont 964, Independencia, Santiago, Chile
| | - Estefania Torres
- Programa Institucional de Fomento a la I+D+I, Universidad Tecnológica Metropolitana, Ignacio Valdivieso 2409, San Joaquín, Chile.
| | - María Luisa Cordero
- Departamento de Física, Facultad de Ciencias Físicas y Matemáticas, Universidad de Chile, Beauchef 850, Santiago, Chile
| | - Valentina Vio
- Departamento de Química Farmacológica y Toxicológica, Facultad de Ciencias Químicas y Farmacéuticas, Laboratorio de Nanobiotecnología, Universidad de Chile, Santos Dumont 964, Independencia, Chile.
- Advanced Center for Chronic Diseases (ACCDis), Santos Dumont 964, Independencia, Santiago, Chile
| | - Juan M Ruso
- Soft Matter and Molecular Biophysics Group, Department of Applied Physics, University of Santiago de Compostela, Santiago de Compostela 15782, Spain
| | - Josué Juárez
- Departamento de Física, Universidad de Sonora, Unidad Centro, Hermosillo, Sonora 83000, Mexico
| | - Antonio Topete
- Laboratorio de Inmunología, Departamento de Fisiología, Centro Universitario de Ciencias de la Salud (CUCS), Universidad de Guadalajara, Guadalajara 44340, Mexico
| | - Eyleen Araya
- Departamento de Ciencias Quimicas, Universidad Andres Bello, Republica 275, 8370146 Santiago, Chile
| | - Rodrigo Vasquez-Contreras
- Departamento de Química Farmacológica y Toxicológica, Facultad de Ciencias Químicas y Farmacéuticas, Laboratorio de Nanobiotecnología, Universidad de Chile, Santos Dumont 964, Independencia, Chile.
- Advanced Center for Chronic Diseases (ACCDis), Santos Dumont 964, Independencia, Santiago, Chile
- Departamento de Ciencias Quimicas, Universidad Andres Bello, Republica 275, 8370146 Santiago, Chile
| | - Marcelo J Kogan
- Departamento de Química Farmacológica y Toxicológica, Facultad de Ciencias Químicas y Farmacéuticas, Laboratorio de Nanobiotecnología, Universidad de Chile, Santos Dumont 964, Independencia, Chile.
- Advanced Center for Chronic Diseases (ACCDis), Santos Dumont 964, Independencia, Santiago, Chile
| | - Natalia Hassan
- Programa Institucional de Fomento a la I+D+I, Universidad Tecnológica Metropolitana, Ignacio Valdivieso 2409, San Joaquín, Chile.
- Advanced Center for Chronic Diseases (ACCDis), Santos Dumont 964, Independencia, Santiago, Chile
| |
Collapse
|
156
|
Investigation of morphology, micelle properties, drug encapsulation and release behavior of self-assembled PEG-PLA-PEG block copolymers: A coarse-grained molecular simulations study. Colloids Surf A Physicochem Eng Asp 2021. [DOI: 10.1016/j.colsurfa.2021.127445] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
|
157
|
Kaushal N, Chen ZS, Lin S. Double-Coated Poly(butyl Cyanoacrylate) Nanoparticles as a Potential Carrier for Overcoming P-Gp- and BCRP-Mediated Multidrug Resistance in Cancer Cells. FRONTIERS IN NANOTECHNOLOGY 2021. [DOI: 10.3389/fnano.2021.753857] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
The present study evaluates poly (butyl cyanoacrylate) nanoparticles (PBCA-NPs), double-coated with Tween 80 and polyethylene glycol (PEG) 20,000 as a potential carrier system for overcoming P-glycoprotein (P-gp) and breast cancer resistant protein (BCRP)-mediated multidrug resistance (MDR) in cancer cell lines. Doxorubicin-loaded PBCA-NPs were prepared by the anionic polymerization method and were successively double-coated with Tween 80 and PEG 20000 at varied concentrations. MDR reversing potential was investigated by cellular uptake in P-gp overexpressing cell line. And, the outcomes were verified by modified MTT assay in P-gp or BCRP overexpressing cell lines. The findings from the cell uptake study indicate that double-coated PBCA-NPs significantly enhanced doxorubicin accumulation within the cells. MTT assays revealed that double-coated PBCA-NPs significantly potentiated the sensitivity of doxorubicin in P-gp overexpressing cells, in comparison to free doxorubicin, single-, and un-coated PBCA-NPs, respectively. Moreover, further increase in concentration with Tween 80, double-coated PBCA-NPs significantly enhanced the sensitivity of doxorubicin in BCRP overexpressing cell line, in comparison to single- and double-coated formulations (with lower concentration of Tween 80). Hence, it could be concluded that double-coated PBCA-NPs can be used as a potential carrier for enhancing doxorubicin accumulation in MDR cancer cells.
Collapse
|
158
|
Maisha N, Naik N, Okesola M, Coombs T, Zilberberg R, Pandala N, Lavik E. Engineering PEGylated Polyester Nanoparticles to Reduce Complement-Mediated Infusion Reaction. Bioconjug Chem 2021; 32:2154-2166. [PMID: 34499487 DOI: 10.1021/acs.bioconjchem.1c00339] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Translation of intravenously administered nanomaterials to the clinic is limited due to adverse infusion reactions. While these reactions are infrequent, with up to 10% prone to experiencing infusion reactions, the reactions can be severe and life-threatening. One of the innate immune pathways, the complement activation pathway, plays a significant role in mediating this response. Nanoparticle surface properties are a relevant design feature, as they control the blood proteins the nanoparticles interact with and allow the nanoparticles to evade the immune reaction. PEGylation of nanosurfaces is critical in improving the blood circulation of nanoparticles and reducing opsonization. Our goal was to understand whether modifying the surface architecture by varying the PEG density and architecture can impact the complement response in vitro. We utilized block copolymers of poly(lactic acid)-b-poly(ethylene glycol) prepared with poly(ethylene glycol) macroinitiators of molecular weights 3400 and 5000 Da. Tracking the complement biomarker C5a, we monitored the impact of changing PEGylation of the nanoparticles. We also investigated how the changing PEG length on the nanoparticle surface impacts further strengthening the stealth properties. Lastly, we determined which cytokines change upon blood incubation with nanoparticles in vitro to understand the extent to which inflammation may occur and the crosstalk between the complement and immune responses. Increasing PEGylation reduced the generation of complement-mediated anaphylatoxin C5a in vitro, with 5000 Da PEG more effectively reducing levels of C5a generated compared to 3400 Da PEG. The insights gathered regarding the impact of PEG density and PEG chain length would be critical in developing stealth nanoparticles that do not lead to infusion reactions upon intravenous administration.
Collapse
Affiliation(s)
- Nuzhat Maisha
- University of Maryland Baltimore County, Baltimore, MD Piscataway Territories 21250, United States
| | - Nidhi Naik
- University of Maryland Baltimore County, Baltimore, MD Piscataway Territories 21250, United States
| | - Mawuyon Okesola
- University of Maryland Baltimore County, Baltimore, MD Piscataway Territories 21250, United States
| | - Tobias Coombs
- University of Maryland Baltimore County, Baltimore, MD Piscataway Territories 21250, United States
| | - Rose Zilberberg
- University of Maryland Baltimore County, Baltimore, MD Piscataway Territories 21250, United States
| | - Narendra Pandala
- University of Maryland Baltimore County, Baltimore, MD Piscataway Territories 21250, United States
| | - Erin Lavik
- University of Maryland Baltimore County, Baltimore, MD Piscataway Territories 21250, United States
| |
Collapse
|
159
|
Baghban R, Afarid M, Soleymani J, Rahimi M. Were magnetic materials useful in cancer therapy? Biomed Pharmacother 2021; 144:112321. [PMID: 34656061 DOI: 10.1016/j.biopha.2021.112321] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2021] [Revised: 09/29/2021] [Accepted: 10/08/2021] [Indexed: 12/11/2022] Open
Abstract
Cancer is one of the major challenges fronting the biomedical basic researches in our time. The study and development of effective therapeutic strategies for cancer therapy are vital. Among the many probable core constituents of nanoparticles, magnetite-based nanoparticles have been widely studied for cancer therapy owing to their inherent magnetic features, multifunctional design, biodegradable and biocompatible properties. Magnetic nanoparticles have been also designed for utilizing as contrast enhancer agents for magnetic resonance imaging, drug delivery systems, and most recently as a therapeutic element in inducing cellular death in tumor ablation therapies. This review aimed to provide an overview of the various applications of magnetic nanoparticles and recent achievements in developing these advanced materials for cancer therapy.
Collapse
Affiliation(s)
- Roghayyeh Baghban
- Poostchi Ophthalmology Research Center, Department of Ophthalmology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran; Student Research Committee, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mehrdad Afarid
- Poostchi Ophthalmology Research Center, Department of Ophthalmology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Jafar Soleymani
- Pharmaceutical Analysis Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Mahdi Rahimi
- Lodz University of Technology, Institute of Polymer and Dye Technology, Stefanowskiego 16, 90-537 Lodz, Poland.
| |
Collapse
|
160
|
Stawicki CM, Rinker TE, Burns M, Tonapi SS, Galimidi RP, Anumala D, Robinson JK, Klein JS, Mallick P. Modular fluorescent nanoparticle DNA probes for detection of peptides and proteins. Sci Rep 2021; 11:19921. [PMID: 34620912 PMCID: PMC8497506 DOI: 10.1038/s41598-021-99084-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2021] [Accepted: 09/20/2021] [Indexed: 02/08/2023] Open
Abstract
Fluorescently labeled antibody and aptamer probes are used in biological studies to characterize binding interactions, measure concentrations of analytes, and sort cells. Fluorescent nanoparticle labels offer an excellent alternative to standard fluorescent labeling strategies due to their enhanced brightness, stability and multivalency; however, challenges in functionalization and characterization have impeded their use. This work introduces a straightforward approach for preparation of fluorescent nanoparticle probes using commercially available reagents and common laboratory equipment. Fluorescent polystyrene nanoparticles, Thermo Fisher Scientific FluoSpheres, were used in these proof-of-principle studies. Particle passivation was achieved by covalent attachment of amine-PEG-azide to carboxylated particles, neutralizing the surface charge from - 43 to - 15 mV. A conjugation-annealing handle and DNA aptamer probe were attached to the azide-PEG nanoparticle surface either through reaction of pre-annealed handle and probe or through a stepwise reaction of the nanoparticles with the handle followed by aptamer annealing. Nanoparticles functionalized with DNA aptamers targeting histidine tags and VEGF protein had high affinity (EC50s ranging from 3 to 12 nM) and specificity, and were more stable than conventional labels. This protocol for preparation of nanoparticle probes relies solely on commercially available reagents and common equipment, breaking down the barriers to use nanoparticles in biological experiments.
Collapse
Affiliation(s)
| | - Torri E Rinker
- Nautilus Biotechnology, 201 Industrial Rd #310, San Carlos, CA, 94070, USA.
| | - Markus Burns
- Nautilus Biotechnology, 201 Industrial Rd #310, San Carlos, CA, 94070, USA
| | - Sonal S Tonapi
- Nautilus Biotechnology, 201 Industrial Rd #310, San Carlos, CA, 94070, USA
| | - Rachel P Galimidi
- Nautilus Biotechnology, 201 Industrial Rd #310, San Carlos, CA, 94070, USA
| | - Deepthi Anumala
- Nautilus Biotechnology, 201 Industrial Rd #310, San Carlos, CA, 94070, USA
| | - Julia K Robinson
- Nautilus Biotechnology, 201 Industrial Rd #310, San Carlos, CA, 94070, USA
| | - Joshua S Klein
- Nautilus Biotechnology, 201 Industrial Rd #310, San Carlos, CA, 94070, USA
| | - Parag Mallick
- Nautilus Biotechnology, 201 Industrial Rd #310, San Carlos, CA, 94070, USA
| |
Collapse
|
161
|
Zhang W, Callmann CE, Mirkin CA. Controlling the Biological Fate of Liposomal Spherical Nucleic Acids Using Tunable Polyethylene Glycol Shells. ACS APPLIED MATERIALS & INTERFACES 2021; 13:46325-46333. [PMID: 34547202 PMCID: PMC8590845 DOI: 10.1021/acsami.1c12852] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Liposomal spherical nucleic acids (LSNAs) modified with polyethylene glycol (PEG) units are studied in an attempt to understand how the circulation time and biodistribution of the constructs can be manipulated. Specifically, the effect of (1) PEG molecular weight, (2) PEG shell stability, and (3) PEG modification method (PEG in both the core and shell versus PEG in the shell only) on LSNA blood circulation, biodistribution, and in vivo cell internalization in a syngeneic, orthotopic triple-negative breast cancer mouse model is studied. Generally, high PEG molecular weight extends blood circulation lifetime, and a more lipophilic anchor stabilizes the PEG shell and improves circulation and tumor accumulation but at the cost of cell uptake efficiency. The PEGylation strategy has a minor effect on in vitro properties of LSNAs but significantly alters in vivo cell uptake. For example, surface-only PEG in one design contributed to higher in vivo cell internalization than its counterpart with PEG both in the shell and core. Taken together, this work provides guidelines for designing LSNAs that exhibit maximal in vivo cancer cell uptake characteristics in the context of a breast cancer model.
Collapse
Affiliation(s)
- Wuliang Zhang
- Department of Chemistry, Northwestern University, 2145 Sheridan Road, Evanston, Illinois 60208, United States
- International Institute for Nanotechnology, Northwestern University, 2145 Sheridan Road, Evanston, Illinois 60208, United States
| | - Cassandra E Callmann
- Department of Chemistry, Northwestern University, 2145 Sheridan Road, Evanston, Illinois 60208, United States
- International Institute for Nanotechnology, Northwestern University, 2145 Sheridan Road, Evanston, Illinois 60208, United States
| | - Chad A Mirkin
- Department of Chemistry, Northwestern University, 2145 Sheridan Road, Evanston, Illinois 60208, United States
- International Institute for Nanotechnology, Northwestern University, 2145 Sheridan Road, Evanston, Illinois 60208, United States
| |
Collapse
|
162
|
Joseph A, Simo GM, Gao T, Alhindi N, Xu N, Graham DJ, Gamble LJ, Nance E. Surfactants influence polymer nanoparticle fate within the brain. Biomaterials 2021; 277:121086. [PMID: 34481289 PMCID: PMC8478896 DOI: 10.1016/j.biomaterials.2021.121086] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Revised: 08/12/2021] [Accepted: 08/23/2021] [Indexed: 12/20/2022]
Abstract
Drug delivery to the brain is limited by poor penetration of pharmaceutical agents across the blood-brain barrier (BBB), within the brain parenchyma, and into specific cells of interest. Nanotechnology can overcome these barriers, but its ability to do so is dependent on nanoparticle physicochemical properties including surface chemistry. Surface chemistry can be determined by a number of factors, including by the presence of stabilizing surfactant molecules introduced during the formulation process. Nanoparticles coated with poloxamer 188 (F68), poloxamer 407 (F127), and polysorbate 80 (P80) have demonstrated uptake in BBB endothelial cells and enhanced accumulation within the brain. However, the impact of surfactants on nanoparticle fate, and specifically on brain extracellular diffusion or intracellular targeting, must be better understood to design nanotherapeutics to efficiently overcome drug delivery barriers in the brain. Here, we evaluated the effect of the biocompatible and commonly used surfactants cholic acid (CHA), F68, F127, P80, and poly (vinyl alcohol) (PVA) on poly (lactic-co-glycolic acid)-poly (ethylene glycol) (PLGA-PEG) nanoparticle transport to and within the brain. The inclusion of these surfactant molecules decreases diffusive ability through brain tissue, reflecting the surfactant's role in encouraging cellular interaction at short length and time scales. After in vivo administration, PLGA-PEG/P80 nanoparticles demonstrated enhanced penetration across the BBB and subsequent internalization within neurons and microglia. Surfactants incorporated into the formulation of PLGA-PEG nanoparticles therefore represent an important design parameter for controlling nanoparticle fate within the brain.
Collapse
Affiliation(s)
- Andrea Joseph
- Department of Chemical Engineering, University of Washington, 98195, Seattle, WA, USA
| | - Georges Motchoffo Simo
- Department of Chemical Engineering, University of Washington, 98195, Seattle, WA, USA; Department of Biochemistry, University of Washington, 98195, Seattle, WA, USA
| | - Torahito Gao
- Department of Chemical Engineering, University of Washington, 98195, Seattle, WA, USA
| | - Norah Alhindi
- Department of Biochemistry, University of Washington, 98195, Seattle, WA, USA
| | - Nuo Xu
- Department of Chemical Engineering, University of Washington, 98195, Seattle, WA, USA
| | - Daniel J Graham
- National ESCA and Surface Analysis Center for Biomedical Problems, University of Washington, 98195, Seattle, WA, USA; Department of Bioengineering, University of Washington, 98195, Seattle, WA, USA
| | - Lara J Gamble
- National ESCA and Surface Analysis Center for Biomedical Problems, University of Washington, 98195, Seattle, WA, USA; Department of Bioengineering, University of Washington, 98195, Seattle, WA, USA
| | - Elizabeth Nance
- Department of Chemical Engineering, University of Washington, 98195, Seattle, WA, USA; Department of Bioengineering, University of Washington, 98195, Seattle, WA, USA; Center for Human Development and Disability, University of Washington, Seattle, WA, USA, 98195; Department of Radiology, University of Washington, 98195, Seattle, WA, USA.
| |
Collapse
|
163
|
Liu Q, Shaukat A, Kyllönen D, Kostiainen MA. Polyelectrolyte Encapsulation and Confinement within Protein Cage-Inspired Nanocompartments. Pharmaceutics 2021; 13:1551. [PMID: 34683843 PMCID: PMC8537137 DOI: 10.3390/pharmaceutics13101551] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Revised: 09/17/2021] [Accepted: 09/20/2021] [Indexed: 12/17/2022] Open
Abstract
Protein cages are nanocompartments with a well-defined structure and monodisperse size. They are composed of several individual subunits and can be categorized as viral and non-viral protein cages. Native viral cages often exhibit a cationic interior, which binds the anionic nucleic acid genome through electrostatic interactions leading to efficient encapsulation. Non-viral cages can carry various cargo, ranging from small molecules to inorganic nanoparticles. Both cage types can be functionalized at targeted locations through genetic engineering or chemical modification to entrap materials through interactions that are inaccessible to wild-type cages. Moreover, the limited number of constitutional subunits ease the modification efforts, because a single modification on the subunit can lead to multiple functional sites on the cage surface. Increasing efforts have also been dedicated to the assembly of protein cage-mimicking structures or templated protein coatings. This review focuses on native and modified protein cages that have been used to encapsulate and package polyelectrolyte cargos and on the electrostatic interactions that are the driving force for the assembly of such structures. Selective encapsulation can protect the payload from the surroundings, shield the potential toxicity or even enhance the intended performance of the payload, which is appealing in drug or gene delivery and imaging.
Collapse
Affiliation(s)
- Qing Liu
- Biohybrid Materials, Department of Bioproducts and Biosystems, Aalto University, 00076 Aalto, Finland; (Q.L.); (A.S.); (D.K.)
| | - Ahmed Shaukat
- Biohybrid Materials, Department of Bioproducts and Biosystems, Aalto University, 00076 Aalto, Finland; (Q.L.); (A.S.); (D.K.)
| | - Daniella Kyllönen
- Biohybrid Materials, Department of Bioproducts and Biosystems, Aalto University, 00076 Aalto, Finland; (Q.L.); (A.S.); (D.K.)
| | - Mauri A. Kostiainen
- Biohybrid Materials, Department of Bioproducts and Biosystems, Aalto University, 00076 Aalto, Finland; (Q.L.); (A.S.); (D.K.)
- HYBER Center, Department of Applied Physics, Aalto University, 00076 Aalto, Finland
| |
Collapse
|
164
|
Understanding the Adsorption of Peptides and Proteins onto PEGylated Gold Nanoparticles. Molecules 2021; 26:molecules26195788. [PMID: 34641335 PMCID: PMC8510204 DOI: 10.3390/molecules26195788] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Revised: 09/18/2021] [Accepted: 09/20/2021] [Indexed: 11/25/2022] Open
Abstract
Polyethylene glycol (PEG) surface conjugations are widely employed to render passivating properties to nanoparticles in biological applications. The benefits of surface passivation by PEG are reduced protein adsorption, diminished non-specific interactions, and improvement in pharmacokinetics. However, the limitations of PEG passivation remain an active area of research, and recent examples from the literature demonstrate how PEG passivation can fail. Here, we study the adsorption amount of biomolecules to PEGylated gold nanoparticles (AuNPs), focusing on how different protein properties influence binding. The AuNPs are PEGylated with three different sizes of conjugated PEG chains, and we examine interactions with proteins of different sizes, charges, and surface cysteine content. The experiments are carried out in vitro at physiologically relevant timescales to obtain the adsorption amounts and rates of each biomolecule on AuNP-PEGs of varying compositions. Our findings are relevant in understanding how protein size and the surface cysteine content affect binding, and our work reveals that cysteine residues can dramatically increase adsorption rates on PEGylated AuNPs. Moreover, shorter chain PEG molecules passivate the AuNP surface more effectively against all protein types.
Collapse
|
165
|
Corrêa RLGQ, dos Santos R, Albuquerque LJC, de Araujo GLB, Edwards-Gayle CJC, Ferreira FF, Costa FN. Ciprofibrate-Loaded Nanoparticles Prepared by Nanoprecipitation: Synthesis, Characterization, and Drug Release. Polymers (Basel) 2021; 13:3158. [PMID: 34578074 PMCID: PMC8468397 DOI: 10.3390/polym13183158] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2021] [Revised: 09/14/2021] [Accepted: 09/15/2021] [Indexed: 11/17/2022] Open
Abstract
Ciprofibrate (CIP) is a highly lipophilic and poorly water-soluble drug, typically used for dyslipidemia treatment. Although it is already commercialized in capsules, no previous studies report its solid-state structure; thus, information about the correlation with its physicochemical properties is lacking. In parallel, recent studies have led to the improvement of drug administration, including encapsulation in polymeric nanoparticles (NPs). Here, we present CIP's crystal structure determined by PXRD data. We also propose an encapsulation method for CIP in micelles produced from Pluronic P123/F127 and PEO-b-PCL, aiming to improve its solubility, hydrophilicity, and delivery. We determined the NPs' physicochemical properties by DLS, SLS, ELS, SAXS and the loaded drug amount by UV-Vis spectroscopy. Micelles showed sizes around 10-20 nm for Pluronic and 35-45 nm for the PEO-b-PCL NPs with slightly negative surface charge and successful CIP loading, especially for the latter; a substantial reduction in ζ-potential may be evidenced. For Pluronic nanoparticles, we scanned different conditions for the CIP loading, and its encapsulation efficiency was reduced while the drug content increased in the nanoprecipitation protocol. We also performed in vitro release experiments; results demonstrate that probe release is driven by Fickian diffusion for the Pluronic NPs and a zero-order model for PEO-b-PCL NPs.
Collapse
Affiliation(s)
| | - Renan dos Santos
- Center for Natural and Human Sciences (CCNH), Federal University of ABC (UFABC), Santo André 09210-580, Brazil; (R.d.S.); (L.J.C.A.); (F.F.F.)
| | - Lindomar José Calumby Albuquerque
- Center for Natural and Human Sciences (CCNH), Federal University of ABC (UFABC), Santo André 09210-580, Brazil; (R.d.S.); (L.J.C.A.); (F.F.F.)
- Brazilian Synchrotron Light Laboratory (LNLS), Brazilian Center for Research in Energy and Materials (CNPEM), Campinas 13083-170, Brazil
| | | | | | - Fabio Furlan Ferreira
- Center for Natural and Human Sciences (CCNH), Federal University of ABC (UFABC), Santo André 09210-580, Brazil; (R.d.S.); (L.J.C.A.); (F.F.F.)
- Nanomedicine Research Unit (NANOMED), Federal University of ABC (UFABC), Santo André 09210-580, Brazil
| | - Fanny Nascimento Costa
- Center for Natural and Human Sciences (CCNH), Federal University of ABC (UFABC), Santo André 09210-580, Brazil; (R.d.S.); (L.J.C.A.); (F.F.F.)
- Diamond Light Source, Harwell Science & Innovation Campus, Didcot, Oxfordshire OX11 0DE, UK;
| |
Collapse
|
166
|
Shaumbwa VR, Liu D, Archer B, Li J, Su F. Preparation and application of magnetic chitosan in environmental remediation and other fields: A review. J Appl Polym Sci 2021. [DOI: 10.1002/app.51241] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Affiliation(s)
- Veino Risto Shaumbwa
- Jiangsu Key Laboratory of Atmospheric Environment Monitoring and Pollution Control, Collaborative Innovation Center of Atmospheric Environment and Equipment Technology, School of Environment Science & Engineering Nanjing University of Information Science & Technology Nanjing China
| | - Dagang Liu
- Jiangsu Key Laboratory of Atmospheric Environment Monitoring and Pollution Control, Collaborative Innovation Center of Atmospheric Environment and Equipment Technology, School of Environment Science & Engineering Nanjing University of Information Science & Technology Nanjing China
| | - Bright Archer
- Jiangsu Key Laboratory of Atmospheric Environment Monitoring and Pollution Control, Collaborative Innovation Center of Atmospheric Environment and Equipment Technology, School of Environment Science & Engineering Nanjing University of Information Science & Technology Nanjing China
| | - Jinlei Li
- Department of Chemical Engineering McMaster University Hamilton Ontario Canada
| | - Fan Su
- Jiangsu Key Laboratory of Atmospheric Environment Monitoring and Pollution Control, Collaborative Innovation Center of Atmospheric Environment and Equipment Technology, School of Environment Science & Engineering Nanjing University of Information Science & Technology Nanjing China
| |
Collapse
|
167
|
Shah N, Hussain M, Rehan T, Khan A, Khan ZU. Overview of polyethylene glycol-based materials with a special focus on core-shell particles for drug delivery application. Curr Pharm Des 2021; 28:352-367. [PMID: 34514984 DOI: 10.2174/1381612827666210910104333] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2021] [Revised: 07/10/2021] [Accepted: 08/13/2021] [Indexed: 11/22/2022]
Abstract
Polyethylene glycols (PEG) are water-soluble nonionic polymeric molecules. PEG and PEG-based materials are used for various important applications such as solvents, adhesives, adsorbents, drug delivery agents, tissue engineering scaffolds, etc. The coating of nanoparticles with PEG forms core-shell nanoparticles. The PEG-based core-shell nanoparticles are synthesized for the development of high-quality drug delivery systems. In the present review, we first explained the basics and various applications of PEGs and PEG-based composites materials and then concentrated on the PEG-based core-shell nanoparticles for biomedical applications specifically their use in drug delivery.
Collapse
Affiliation(s)
- Nasrullah Shah
- Department of Chemistry, Abdul Wali Khan University Mardan, Mardan, KP 23200. Pakistan
| | - Manzoor Hussain
- Department of Chemistry, Abdul Wali Khan University Mardan, Mardan, KP 23200. Pakistan
| | - Touseef Rehan
- Department of Biochemistry, Shaheed Benazir Bhutto Women University, Peshawar, KP 45000. Pakistan
| | - Abbas Khan
- Department of Chemistry, Abdul Wali Khan University Mardan, Mardan, KP 23200. Pakistan
| | - Zubair Ullah Khan
- Department of Chemistry, Abdul Wali Khan University Mardan, Mardan, KP 23200. Pakistan
| |
Collapse
|
168
|
Chen X, Zhuang Y, Rampal N, Hewitt R, Divitini G, O’Keefe CA, Liu X, Whitaker DJ, Wills JW, Jugdaohsingh R, Powell JJ, Yu H, Grey CP, Scherman OA, Fairen-Jimenez D. Formulation of Metal-Organic Framework-Based Drug Carriers by Controlled Coordination of Methoxy PEG Phosphate: Boosting Colloidal Stability and Redispersibility. J Am Chem Soc 2021; 143:13557-13572. [PMID: 34357768 PMCID: PMC8414479 DOI: 10.1021/jacs.1c03943] [Citation(s) in RCA: 49] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Indexed: 12/16/2022]
Abstract
Metal-organic framework nanoparticles (nanoMOFs) have been widely studied in biomedical applications. Although substantial efforts have been devoted to the development of biocompatible approaches, the requirement of tedious synthetic steps, toxic reagents, and limitations on the shelf life of nanoparticles in solution are still significant barriers to their translation to clinical use. In this work, we propose a new postsynthetic modification of nanoMOFs with phosphate-functionalized methoxy polyethylene glycol (mPEG-PO3) groups which, when combined with lyophilization, leads to the formation of redispersible solid materials. This approach can serve as a facile and general formulation method for the storage of bare or drug-loaded nanoMOFs. The obtained PEGylated nanoMOFs show stable hydrodynamic diameters, improved colloidal stability, and delayed drug-release kinetics compared to their parent nanoMOFs. Ex situ characterization and computational studies reveal that PEGylation of PCN-222 proceeds in a two-step fashion. Most importantly, the lyophilized, PEGylated nanoMOFs can be completely redispersed in water, avoiding common aggregation issues that have limited the use of MOFs in the biomedical field to the wet form-a critical limitation for their translation to clinical use as these materials can now be stored as dried samples. The in vitro performance of the addition of mPEG-PO3 was confirmed by the improved intracellular stability and delayed drug-release capability, including lower cytotoxicity compared with that of the bare nanoMOFs. Furthermore, z-stack confocal microscopy images reveal the colocalization of bare and PEGylated nanoMOFs. This research highlights a facile PEGylation method with mPEG-PO3, providing new insights into the design of promising nanocarriers for drug delivery.
Collapse
Affiliation(s)
- Xu Chen
- The
Adsorption & Advanced Materials Laboratory (AML),
Department of Chemical Engineering & Biotechnology, University of Cambridge, Philippa Fawcett Drive, Cambridge CB3 0AS, United
Kingdom
| | - Yunhui Zhuang
- The
Adsorption & Advanced Materials Laboratory (AML),
Department of Chemical Engineering & Biotechnology, University of Cambridge, Philippa Fawcett Drive, Cambridge CB3 0AS, United
Kingdom
| | - Nakul Rampal
- The
Adsorption & Advanced Materials Laboratory (AML),
Department of Chemical Engineering & Biotechnology, University of Cambridge, Philippa Fawcett Drive, Cambridge CB3 0AS, United
Kingdom
| | - Rachel Hewitt
- Biominerals
Research Laboratory & Cellular Imaging and Analysis Facility,
Department of Veterinary Medicine, University
of Cambridge, Madingley Road, Cambridge CB3 0ES, United Kingdom
| | - Giorgio Divitini
- Electron
Microscopy Group, Department of Materials Science and Metallurgy, University of Cambridge, 27 Charles Babbage Road, Cambridge CB3 0FS, United
Kingdom
| | - Christopher A. O’Keefe
- Department
of Chemistry, University of Cambridge, Lensfield Road, Cambridge CB2 1EW, United
Kingdom
| | - Xiewen Liu
- The
Adsorption & Advanced Materials Laboratory (AML),
Department of Chemical Engineering & Biotechnology, University of Cambridge, Philippa Fawcett Drive, Cambridge CB3 0AS, United
Kingdom
| | - Daniel J. Whitaker
- Department
of Chemistry, University of Cambridge, Lensfield Road, Cambridge CB2 1EW, United
Kingdom
| | - John W. Wills
- Biominerals
Research Laboratory & Cellular Imaging and Analysis Facility,
Department of Veterinary Medicine, University
of Cambridge, Madingley Road, Cambridge CB3 0ES, United Kingdom
| | - Ravin Jugdaohsingh
- Biominerals
Research Laboratory & Cellular Imaging and Analysis Facility,
Department of Veterinary Medicine, University
of Cambridge, Madingley Road, Cambridge CB3 0ES, United Kingdom
| | - Jonathan J. Powell
- Biominerals
Research Laboratory & Cellular Imaging and Analysis Facility,
Department of Veterinary Medicine, University
of Cambridge, Madingley Road, Cambridge CB3 0ES, United Kingdom
| | - Han Yu
- School
of Chemical and Environmental Engineering, Shanghai Institute of Technology, No. 100 Haiquan Road, Shanghai 201418, P. R. China
| | - Clare P. Grey
- Department
of Chemistry, University of Cambridge, Lensfield Road, Cambridge CB2 1EW, United
Kingdom
| | - Oren A. Scherman
- Department
of Chemistry, University of Cambridge, Lensfield Road, Cambridge CB2 1EW, United
Kingdom
| | - David Fairen-Jimenez
- The
Adsorption & Advanced Materials Laboratory (AML),
Department of Chemical Engineering & Biotechnology, University of Cambridge, Philippa Fawcett Drive, Cambridge CB3 0AS, United
Kingdom
| |
Collapse
|
169
|
Porcino M, Li X, Gref R, Martineau-Corcos C. Solid-state NMR spectroscopy as a powerful tool to investigate the location of fluorinated lipids in highly porous hybrid organic-inorganic nanoparticles. MAGNETIC RESONANCE IN CHEMISTRY : MRC 2021; 59:1038-1047. [PMID: 33709480 DOI: 10.1002/mrc.5148] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Revised: 03/08/2021] [Accepted: 03/08/2021] [Indexed: 06/12/2023]
Abstract
Nanosized metal-organic frameworks (nanoMOFs) have emerged as a new class of biodegradable and nontoxic nanomaterials of high interest for biomedical applications thanks to the possibility to load large amounts of a wide variety of therapeutic molecules in their porous structure. The surface of the highly porous nanoMOFs is usually engineered to increase their colloidal stability, to tune their interactions with the biological environment, and to allow targeting specific cells or organs. However, the atomic-scale analysis of these complex core-shell materials is highly challenging. In this study, we report the investigation of aluminum-based nanoMOFs containing two fluorinated lipids by solid-state NMR spectroscopy, including 27 Al, 1 H and 19 F MAS NMR. The ensemble of NMR data provides a better understanding of the localization and conformation of the fluorinated lipids inside the pores or on the nanoMOF surface.
Collapse
Affiliation(s)
| | - Xue Li
- Institut des Sciences Moléculaires d'Orsay, UMR CNRS 8214, Paris-Sud University, Université Paris Saclay, Orsay, France
| | - Ruxandra Gref
- Institut des Sciences Moléculaires d'Orsay, UMR CNRS 8214, Paris-Sud University, Université Paris Saclay, Orsay, France
| | - Charlotte Martineau-Corcos
- CEMHTI UPR CNRS 3079, Université d'Orléans, Orléans, France
- ILV UMR CNRS 8180, Université de Versailles St-Quentin en Yvelines, Université Paris Saclay, Versailles, France
- CortecNet, Les Ulis, France
| |
Collapse
|
170
|
Nanoparticle shell structural cues drive in vitro transport properties, tissue distribution and brain accessibility in zebrafish. Biomaterials 2021; 277:121085. [PMID: 34461457 DOI: 10.1016/j.biomaterials.2021.121085] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2021] [Revised: 07/26/2021] [Accepted: 08/23/2021] [Indexed: 01/14/2023]
Abstract
Zwitterion polymers with strong antifouling properties have been suggested as the prime alternative to polyethylene glycol (PEG) for drug nanocarriers surface coating. It is believed that PEG coating shortcomings, such as immune responses and incomplete protein repellency, could be overcome by zwitterionic polymers. However, no systematic study has been conducted so far to complete a comparative appraisal of PEG and zwitterionic-coating effects on nanoparticles (NPs) stealthness, cell uptake, cell barrier translocation and biodistribution in the context of nanocarriers brain targeting. Core-shell polymeric particles with identical cores and a shell of either PEG or poly(2-methacryloyloxyethyl phosphorylcholine (PMPC) were prepared by impinging jet mixer nanoprecipitation. NPs with similar size and surface potential were systematically compared using in vitro and in vivo assays. NPs behavior differences were rationalized based on their protein-particles interactions. PMPC-coated NPs were significantly more endocytosed by mouse macrophages or brain resident macrophages compared to PEGylated NPs but exhibited the remarkable ability to cross the blood-brain barrier in in vitro models. Nanoscale flow cytometry assays showed significantly more adsorbed proteins on PMPC-coated NPs than PEG-coated NPs. In vivo, distribution in zebrafish larvae, showed a strong propensity for PMPC-coated NPs to adhere to the vascular endothelium, while PEG-coated NPs were able to circulate for a longer time and escape the bloodstream to penetrate deep into the cerebral tissue. The stark differences between these two types of particles, besides their similarities in size and surface potential, points towards the paramount role of surface chemistry in controlling NPs fate likely via the formation of distinct protein corona for each coating.
Collapse
|
171
|
Razza N, Lavino AD, Fadda G, Lairez D, Impagnatiello A, Marchisio D, Sangermano M, Rizza G. Nanoprobes to investigate nonspecific interactions in lipid bilayers: from defect-mediated adhesion to membrane disruption. NANOSCALE ADVANCES 2021; 3:4979-4989. [PMID: 36132337 PMCID: PMC9418973 DOI: 10.1039/d1na00360g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Accepted: 07/08/2021] [Indexed: 06/15/2023]
Abstract
When a lipid membrane approaches a material/nanomaterial, nonspecific adhesion may occur. The interactions responsible for nonspecific adhesion can either preserve the membrane integrity or lead to its disruption. Despite the importance of the phenomenon, there is still a lack of clear understanding of how and why nonspecific adhesion may originate different resulting scenarios and how these interaction scenarios can be investigated. This work aims at bridging this gap by investigating the role of the interplay between cationic electrostatic and hydrophobic interactions in modulating the membrane stability during nonspecific adhesion phenomena. Here, the stability of the membrane has been studied employing anisotropic nanoprobes in zwitterionic lipid membranes with the support of coarse-grained molecular dynamics simulations to interpret the experimental observations. Lipid membrane electrical measurements and nanoscale visualization in combination with molecular dynamics simulations revealed the phenomena driving nonspecific adhesion. Any interaction with the lipidic bilayer is defect-mediated involving cationic electrostatically driven lipid extraction and hydrophobically-driven chain protrusion, whose interplay determines the existence of a thermodynamic optimum for the membrane structural integrity. These findings unlock unexplored routes to exploit nonspecific adhesion in lipid membranes. The proposed platform can act as a straightforward probing tool to locally investigate interactions between synthetic materials and lipid membranes for the design of antibacterials, antivirals, and scaffolds for tissue engineering.
Collapse
Affiliation(s)
- Nicolò Razza
- Department of Applied Science and Technology, Politecnico di Torino Torino Italy
| | - Alessio D Lavino
- Department of Applied Science and Technology, Politecnico di Torino Torino Italy
| | - Giulia Fadda
- CSPAT UMR 7244, Université Sorbonne Paris Nord 74 rue Marcel Cachin 93017 Bobigny France
- Laboratoire Léon Brillouin, CNRS, CEA, Université Paris-Saclay 91191 Gif-sur-Yvette Cedex France
| | - Didier Lairez
- Laboratoire Léon Brillouin, CNRS, CEA, Université Paris-Saclay 91191 Gif-sur-Yvette Cedex France
- Laboratoire des Solides Irradiés (LSI), Institut Polytechnique de Paris, CEA/DRF/IRAMIS, CNRS 91128 Palaiseau Cedex France
| | - Andrea Impagnatiello
- Laboratoire des Solides Irradiés (LSI), Institut Polytechnique de Paris, CEA/DRF/IRAMIS, CNRS 91128 Palaiseau Cedex France
| | - Daniele Marchisio
- Department of Applied Science and Technology, Politecnico di Torino Torino Italy
| | - Marco Sangermano
- Department of Applied Science and Technology, Politecnico di Torino Torino Italy
| | - Giancarlo Rizza
- Laboratoire des Solides Irradiés (LSI), Institut Polytechnique de Paris, CEA/DRF/IRAMIS, CNRS 91128 Palaiseau Cedex France
| |
Collapse
|
172
|
Becicka WM, Bielecki PA, Lorkowski ME, Moon TJ, Zhang Y, Atukorale PU, Covarrubias G, Karathanasis E. The effect of PEGylation on the efficacy and uptake of an immunostimulatory nanoparticle in the tumor immune microenvironment. NANOSCALE ADVANCES 2021; 3:4961-4972. [PMID: 34485818 PMCID: PMC8386411 DOI: 10.1039/d1na00308a] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Accepted: 07/23/2021] [Indexed: 05/28/2023]
Abstract
The efficacy of immunotherapies is often limited by the immunosuppressive tumor microenvironment, which is populated with dysfunctional innate immune cells. To reprogram the tumor-resident innate immune cells, we developed immunostimulatory silica mesoporous nanoparticles (immuno-MSN). The cargo of immuno-MSN is a Stimulator of Interferon Gene (STING) agonist, which activates innate immune cells leading to production of interferon (IFN) β. By proficiently trafficking its cargo into immune cells, the immuno-MSN induced a 9-fold increase of IFN-β secretion compared to free agonist. While an external PEG shield has historically been used to protect nanoparticles from immune recognition, a PEGylated immunostimulatory nanoparticle needs to strike a balance between immune evasion to avoid off-site accumulation and uptake by target immune cells in tumors. Using the 4T1 mouse model of metastatic breast cancer and flow cytometry, it was determined that the degree of PEGylation significantly influenced the uptake of 'empty' MSNs by tumor-resident innate immune cells. This was not the case for the agonist-loaded immuno-MSN variants. It should be noted the surface charge of the 'empty' MSNs was positive rather than neutral for the agonist-loaded immuno-MSNs. However, even though the cellular uptake was similar at 24 h after injection for the three immuno-MSN variants, we observed a significant beneficial effect on the activation and expansion of APCs especially in lung metastasis using the lightly PEGylated immuno-MSN variant.
Collapse
Affiliation(s)
- Wyatt M Becicka
- Department of Biomedical Engineering, Case Western Reserve University Cleveland OH USA
| | - Peter A Bielecki
- Department of Biomedical Engineering, Case Western Reserve University Cleveland OH USA
- Case Comprehensive Cancer Center, Case Western Reserve University Cleveland OH USA
| | - Morgan E Lorkowski
- Department of Biomedical Engineering, Case Western Reserve University Cleveland OH USA
| | - Taylor J Moon
- Department of Biomedical Engineering, Case Western Reserve University Cleveland OH USA
| | - Yahan Zhang
- Department of Biomedical Engineering, Case Western Reserve University Cleveland OH USA
| | - Prabhani U Atukorale
- Department of Biomedical Engineering, Case Western Reserve University Cleveland OH USA
- Case Comprehensive Cancer Center, Case Western Reserve University Cleveland OH USA
| | - Gil Covarrubias
- Department of Biomedical Engineering, Case Western Reserve University Cleveland OH USA
- Case Comprehensive Cancer Center, Case Western Reserve University Cleveland OH USA
| | - Efstathios Karathanasis
- Department of Biomedical Engineering, Case Western Reserve University Cleveland OH USA
- Case Comprehensive Cancer Center, Case Western Reserve University Cleveland OH USA
| |
Collapse
|
173
|
Liu Y, Choi CKK, Hong H, Xiao Y, Kwok ML, Liu H, Tian XY, Choi CHJ. Dopamine Receptor-Mediated Binding and Cellular Uptake of Polydopamine-Coated Nanoparticles. ACS NANO 2021; 15:13871-13890. [PMID: 34379407 DOI: 10.1021/acsnano.1c06081] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Polydopamine (PDA)-coated nanoparticles (NPs) are emerging carriers of therapeutic agents for nanomedicine applications due to their biocompatibility and abundant entry to various cell types, yet it remains unknown whether their cellular entry engages cell-surface receptors. As monomeric dopamine (DA) is an endogenous ligand of dopamine receptor and raw ingredient of PDA, we elucidate the interaction between polyethylene glycol-stabilized, PDA-coated gold NPs (Au@PDA@PEG NPs) and dopamine receptors, particularly D2 (D2DR). After proving the binding of Au@PDA@PEG NPs to recombinant and cellular D2DR, we employ antibody blocking, gene knockdown, and gene overexpression to establish the role of D2DR in the cellular uptake of Au@PDA@PEG NPs in vitro. By preparing a series of PEG-coated AuNPs that contain different structural analogues of DA (Au@PEG-X NPs), we demonstrate that catechol and amine groups collectively enhance the binding of NPs to D2DR and their cellular uptake. By intravenously injecting Au@PDA@PEG NPs to Balb/c mice, we reveal their in vivo binding to D2DR in the liver by competitive inhibition and immunohistochemistry together with their preferential association to D2DR-rich resident Kupffer cells by flow cytometry, a result consistent with the profuse expression of D2DR by resident Kupffer cells. Catechol and amine groups jointly contribute to the preferential association of NPs to D2DR-rich Kupffer cells. Our data highlight the importance of D2DR expression and DA-related functional groups in mediating the cell-nano interactions of PDA-based nanomedicines.
Collapse
|
174
|
Rajdev P, Ghosh S. Thermodynamic Insights into Protein Adsorption on Supramolecular Assemblies of π-Amphiphiles. J Phys Chem B 2021; 125:8981-8988. [PMID: 34324355 DOI: 10.1021/acs.jpcb.1c03283] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Nonspecific adsorption of proteins on the surface of nanocarriers plays a critical role in their cellular uptake and other biological functions. This article reports vesicular assemblies of two π-amphiphiles (NDI-1 and NDI-2) and thermodynamic aspects of their interaction with bovine serum albumin (BSA). Both contain a hydrophobic naphthalene-diimide (NDI) core and two oligo-oxyethylene (OE) wedges but differ by the presence of the hydrazide group in NDI-1. NDI-2 exhibits a constricted π-stacking and enthalpy-driven adsorption of BSA. In contrast, NDI-1 exhibits a stronger interaction due to enhanced entropy contribution. It is postulated that a tight packing of NDI chromophores in NDI-2 results in an inadequate space in the corona, leading to the dehydration of OE chains, which contributes to the observed enthalpy-driven binding. On the other hand, due to H-bonding along the direction of π-stacking in NDI-1, an enhanced interchromophoric distance provides more space in the shell, resulting in less dehydration of the OE chains, which results in an entropy gain from the BSA binding-induced release of water from the OE chains. Intercalation of an electron-rich pyrene in the electron-deficient NDI-1 stack further reduces the grafting density of the OE chains, resulting in negligible BSA adsorption, similar to a stealth polymer. A correlation can be seen between the thermodynamic landscape of the protein adsorption and the trend of their lower critical solution temperature (LCST), which follows the order NDI-1 + Py < NDI-1 < NDI-2.
Collapse
|
175
|
Guerra-Rebollo M, Stampa M, Lázaro MÁ, Cascante A, Fornaguera C, Borrós S. Electrostatic Coating of Viral Particles for Gene Delivery Applications in Muscular Dystrophies: Influence of Size on Stability and Antibody Protection. J Neuromuscul Dis 2021; 8:815-825. [PMID: 34366365 DOI: 10.3233/jnd-210662] [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: 11/15/2022]
Abstract
BACKGROUND Duchenne Muscular Dystrophy (DMD) is one of the most common muscular dystrophies, caused by mutated forms of the dystrophin gene. Currently, the only treatment available is symptoms management. Novel approximations are trying to treat these patients with gene therapy, namely, using viral vectors. However, these vectors can be recognized by the immune system decreasing their therapeutic activity and making impossible a multidose treatment due to the induction of the humoral immunity following the first dose. OBJECTIVE Our objective is to demonstrate the feasibility of using a hybrid vector to avoid immune clearance, based on the electrostatic coating of adeno-associated virus (AAVs) vectors with our proprietary polymers. METHODS We coated model adeno-associated virus vectors by electrostatic interaction of our cationic poly (beta aminoester) polymers with the viral anionic capsid and characterized biophysical properties. Once the nanoformulations were designed, we studied their in vivo biodistribution by bioluminescence analysis and we finally studied the capacity of the polymers as potential coatings to avoid antibody neutralization. RESULTS We tested two polymer combinations and we demonstrated the need for poly(ethylene glycol) addition to avoid vector aggregation after coating. In vivo biodistribution studies demonstrated that viral particles are located in the liver (short times) and also in muscles (long times), the target organ. However, we did not achieve complete antibody neutralization shielding using this electrostatic coating. CONCLUSIONS The null hypothesis stands: although it is feasible to coat viral particles by electrostatic interaction with a proprietary polymer, this strategy is not appropriate for AAVs due to their small size, so other alternatives are required as a novel treatment for DMD patients.
Collapse
Affiliation(s)
- Marta Guerra-Rebollo
- Grup d'Enginyeria de Materials (Gemat), Institut Químic de Sarriá (IQS), Universitat Ramon Llull (URL), Via Augusta, Barcelona, Spain.,Sagetis Biotech, Via Augusta, Barcelona, Spain
| | - María Stampa
- Grup d'Enginyeria de Materials (Gemat), Institut Químic de Sarriá (IQS), Universitat Ramon Llull (URL), Via Augusta, Barcelona, Spain
| | | | - Anna Cascante
- Grup d'Enginyeria de Materials (Gemat), Institut Químic de Sarriá (IQS), Universitat Ramon Llull (URL), Via Augusta, Barcelona, Spain.,Sagetis Biotech, Via Augusta, Barcelona, Spain
| | - Cristina Fornaguera
- Grup d'Enginyeria de Materials (Gemat), Institut Químic de Sarriá (IQS), Universitat Ramon Llull (URL), Via Augusta, Barcelona, Spain.,Sagetis Biotech, Via Augusta, Barcelona, Spain
| | - Salvador Borrós
- Grup d'Enginyeria de Materials (Gemat), Institut Químic de Sarriá (IQS), Universitat Ramon Llull (URL), Via Augusta, Barcelona, Spain.,Sagetis Biotech, Via Augusta, Barcelona, Spain
| |
Collapse
|
176
|
Zhu X, Chen X, Huo D, Cen J, Jia Z, Liu Y, Liu J. A hybrid nanozymes in situ oxygen supply synergistic photothermal/chemotherapy of cancer management. Biomater Sci 2021; 9:5330-5343. [PMID: 34190241 DOI: 10.1039/d1bm00667c] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Hypoxia in the solid tumor microenvironment (TME) can easily induce tumor recurrence, metastasis, and drug resistance. The use of man-made nanozymes is considered to be an effective strategy for regulating hypoxia in the TME. Herein, Ru@MnO2 nanozymes were constructed via an in situ reduction method, and they showed excellent photothermal conversion efficiency and catalytic activity. The anti-tumor drug DOX with fluorescence was loaded on the Ru@MnO2 nanozymes, and an erythrocyte membrane was further coated on the surface of the Ru@MnO2 nanozymes to construct nanozymes with on-demand release abilities. The erythrocyte membrane (RBCm) enhances the biocompatibility of the Ru@MnO2 nanozymes and prolongs their circulation time in the blood. Ru@MnO2 nanozymes can catalyze endogenous H2O2 to produce O2 to relieve hypoxia in the TME to enhance the efficacy of the photothermal therapy/chemotherapy of cancer. In vitro studies confirmed that the Ru@MnO2 nanozymes showed good tumor penetration abilities and a synergistic anti-tumor effect. Importantly, both in vivo and in vitro studies have confirmed that the oxygen supply in situ enhanced the efficacy of the PTT/chemotherapy of cancer. Accordingly, this study demonstrated that Ru@MnO2 nanozymes can be used as an effective integrated system allowing catalysis, photothermal therapy, and chemotherapy for cancer management.
Collapse
Affiliation(s)
- Xufeng Zhu
- Shenzhen Longhua Maternity and Child Healthcare Hospital, Shenzhen, 518110, China. and Department of Chemistry, College of Chemistry and Materials Science, Jinan University, Guangzhou 511436, China.
| | - Xu Chen
- Shenzhen Longhua Maternity and Child Healthcare Hospital, Shenzhen, 518110, China. and Department of Chemistry, College of Chemistry and Materials Science, Jinan University, Guangzhou 511436, China.
| | - Dongliang Huo
- Shenzhen Longhua Maternity and Child Healthcare Hospital, Shenzhen, 518110, China. and Department of Chemistry, College of Chemistry and Materials Science, Jinan University, Guangzhou 511436, China.
| | - Jieqiong Cen
- Shenzhen Longhua Maternity and Child Healthcare Hospital, Shenzhen, 518110, China. and Department of Chemistry, College of Chemistry and Materials Science, Jinan University, Guangzhou 511436, China.
| | - Zhi Jia
- Shenzhen Longhua Maternity and Child Healthcare Hospital, Shenzhen, 518110, China. and Department of Chemistry, College of Chemistry and Materials Science, Jinan University, Guangzhou 511436, China.
| | - Yanan Liu
- Shenzhen Longhua Maternity and Child Healthcare Hospital, Shenzhen, 518110, China.
| | - Jie Liu
- Shenzhen Longhua Maternity and Child Healthcare Hospital, Shenzhen, 518110, China. and Department of Chemistry, College of Chemistry and Materials Science, Jinan University, Guangzhou 511436, China.
| |
Collapse
|
177
|
Srivastava P, Gunawan C, Soeriyadi A, Amal R, Hoehn K, Marquis C. In vitro coronal protein signatures and biological impact of silver nanoparticles synthesized with different natural polymers as capping agents. NANOSCALE ADVANCES 2021; 3:4424-4439. [PMID: 36133466 PMCID: PMC9418127 DOI: 10.1039/d0na01013h] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Accepted: 05/16/2021] [Indexed: 06/15/2023]
Abstract
Biopolymer-capped particles, sodium alginate-, gelatin- and reconstituted silk fibroin-capped nanosilver (AgNPs), were synthesized with an intention to study, simultaneously, their in vitro and in vivo haemocompatibility, one of the major safety factors in biomedical applications. Solid state characterization showed formation of spherical nanoparticles with 5 to 30 nm primary sizes (transmission electron microscopy) and X-ray photoelectron spectroscopy analysis of particles confirmed silver bonding with the biopolymer moieties. The degree of aggregation of the biopolymer-capped AgNPs in the synthesis medium (ultrapure water) is relatively low, with comparable hydrodynamic size with those of the control citrate-stabilized NPs, and remained relatively unchanged even after 6 weeks. The polymer-capped nanoparticles showed different degrees of aggregation in biologically relevant media - PBS (pH 7.4) and 2% human blood plasma - with citrate- (control) and alginate-capped particles showing the highest aggregation, while gelatin- and silk fibroin-capped particles revealed better stability and less aggregation in these media. In vitro cytotoxicity studies revealed that the polymer-capped particles exhibited both concentration and (hydrodynamic) size-dependent haemolytic activity, the extent of which was higher (up to 100% in some cases) in collected whole blood samples of healthy human volunteers when compared to that in the washed erythrocytes. This difference is thought to result from the detected protein corona formation on the nanoparticle surface in the whole blood system, which was associated with reduced particle aggregation, causing more severe cytotoxic effects. At the tested particle concentration range in vitro, we observed a negligible haemolysis effect in vivo (Balb/c mice). Polymer-capped particles did accumulate in organs, with the highest levels detected in the liver (up to 422 μg per g tissue), yet no adverse behavioural effects were observed in the mice during the duration of the nanoparticle exposure.
Collapse
Affiliation(s)
- Priyanka Srivastava
- School of Biotechnology and Biomolecular Sciences, University of New South Wales Sydney NSW 2052 Australia
- School of Biosciences and Technology, Vellore Institute of Technology Vellore Tamil Nadu 632014 India
| | - Cindy Gunawan
- i3 Institute, University of Technology Sydney NSW 2006 Australia
| | - Alexander Soeriyadi
- School of Chemistry, University of New South Wales Sydney NSW 2052 Australia
- Mochtar Riady Institute for Nanotechnology Tangerang 15810 Indonesia
| | - Rose Amal
- School of Chemical Engineering, University of New South Wales Sydney NSW 2052 Australia
| | - Kyle Hoehn
- School of Biotechnology and Biomolecular Sciences, University of New South Wales Sydney NSW 2052 Australia
| | - Christopher Marquis
- School of Biotechnology and Biomolecular Sciences, University of New South Wales Sydney NSW 2052 Australia
| |
Collapse
|
178
|
Ramírez García G, d'Orlyé F, Richard C, Mignet N, Varenne A. Electrokinetic elucidation of the interactions between persistent luminescent nanoprobes and the binary apolipoprotein-E/albumin protein system. Analyst 2021; 146:5245-5254. [PMID: 34296726 DOI: 10.1039/d1an00781e] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
The affinity between functional nanoparticles (NPs) and proteins could determine the efficacy of nanoprobes, nanosensors, nanocarriers, and many other devices for biomedical applications. Therefore, it is necessary to develop analytical strategies to accurately evaluate the magnitude of these protein corona interactions in physiological media. In this work, different electrokinetic strategies were implemented to accurately determine the interactions between PEGylated ZnGa1.995Cr0.005O4 persistent luminescent NPs (ZGO-PEG) and two important serum proteins: human serum albumin (HSA), the most abundant serum protein, and apolipoprotein-E (ApoE), associated with the active transport of NPs through the blood-brain barrier. Firstly, the injection of ZGO-PEG in a background electrolyte (BGE) containing individual proteins allowed an affinity study to separately characterize each NP-protein system. Then, the same procedure was applied in a buffer containing a mixture of the two proteins at different molar ratios. Finally, the NPs were pre-incubated with one protein and thereafter electrokinetically separated in a BGE containing the second protein. These analytical strategies revealed the mechanisms (comparative, cooperative or competitive systems) and the magnitude of their interactions, resulting in all cases in notably higher affinity and stability between ZGO-PEG and ApoE (Ka = 1.96 ± 0.25 × 1010 M-M) compared to HSA (Ka = 4.60 ± 0.41 × 106 M-M). For the first time, the inter-protein ApoE/HSA interactions with ZGO-PEG were also demonstrated, highlighting the formation of a ternary ZGO-PEG/ApoE/HSA nanocomplex. These results open the way for a deeper understanding of the protein corona formation, and the development of versatile optical imaging applications for ZGO-PEG and other systemically delivered nanoprobes ideally vectorized to the brain.
Collapse
Affiliation(s)
- Gonzalo Ramírez García
- Centro de Física Aplicada y Tecnología Avanzada, Universidad Nacional Autónoma de México, 3001, Blvd. Juriquilla, 76230, Querétaro, Mexico
| | | | | | | | | |
Collapse
|
179
|
Lu C, Han L, Wang J, Wan J, Song G, Rao J. Engineering of magnetic nanoparticles as magnetic particle imaging tracers. Chem Soc Rev 2021; 50:8102-8146. [PMID: 34047311 DOI: 10.1039/d0cs00260g] [Citation(s) in RCA: 61] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Magnetic particle imaging (MPI) has recently emerged as a promising non-invasive imaging technique because of its signal linearly propotional to the tracer mass, ability to generate positive contrast, low tissue background, unlimited tissue penetration depth, and lack of ionizing radiation. The sensitivity and resolution of MPI are highly dependent on the properties of magnetic nanoparticles (MNPs), and extensive research efforts have been focused on the design and synthesis of tracers. This review examines parameters that dictate the performance of MNPs, including size, shape, composition, surface property, crystallinity, the surrounding environment, and aggregation state to provide guidance for engineering MPI tracers with better performance. Finally, we discuss applications of MPI imaging and its challenges and perspectives in clinical translation.
Collapse
Affiliation(s)
- Chang Lu
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, Changsha 410082, P. R. China.
| | - Linbo Han
- College of Health Science and Environmental Engineering, Shenzhen Technology University, Shenzhen 518118, P. R. China
| | - Joanna Wang
- Molecular Imaging Program at Stanford, Department of Radiology, Stanford University School of Medicine, 1201 Welch Road, Stanford, California 94305-5484, USA.
| | - Jiacheng Wan
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, Changsha 410082, P. R. China.
| | - Guosheng Song
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, Changsha 410082, P. R. China.
| | - Jianghong Rao
- Molecular Imaging Program at Stanford, Department of Radiology, Stanford University School of Medicine, 1201 Welch Road, Stanford, California 94305-5484, USA.
| |
Collapse
|
180
|
Li H, Wang Y, Tang Q, Yin D, Tang C, He E, Zou L, Peng Q. The protein corona and its effects on nanoparticle-based drug delivery systems. Acta Biomater 2021; 129:57-72. [PMID: 34048973 DOI: 10.1016/j.actbio.2021.05.019] [Citation(s) in RCA: 112] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Revised: 04/25/2021] [Accepted: 05/18/2021] [Indexed: 02/04/2023]
Abstract
In most cases, once nanoparticles (NPs) enter the blood, their surface is covered by biological molecules, especially proteins, forming a so-called protein corona (PC). As a result, what the cells of the body "see" is not the NPs as formulated by the chemists, but the PC. In this way, the PC can influence the effects of the NPs and even mask the desired effects of the NP components. While this can argue for trying to inhibit protein-nanomaterial interactions, encapsulating NPs in an endogenous PC may increase their clinical usefulness. In this review, we briefly introduce the concept of the PC, its formation and its effects on the behavior of NPs. We also discuss how to reduce the formation of PCs or exploit them to enhance NP functions. Studying the interactions between proteins and NPs will provide insights into their clinical activity in health and disease. STATEMENT OF SIGNIFICANCE: The formation of protein corona (PC) will affect the operation of nanoparticles (NPs) in vivo. Since there are many proteins in the blood, it is impossible to completely overcome the formation of PC. Therefore, the use of PCs to deliver drug is the best choice. De-opsonins adsorbed on NPs can reduce macrophage phagocytosis and cytotoxicity of NPs, and prolong their circulation in blood. Albumin, apolipoprotein and transferrin are typical de-opsonins. In present review, we mainly discuss how to optimize the delivery of nanoparticles through the formation of albumin corona, transferrin corona and apolipoprotein corona in vivo or in vitro.
Collapse
Affiliation(s)
- Hanmei Li
- Key Laboratory of Coarse Cereal Processing (Ministry of Agriculture and Rural Affairs), School of Food and Biological Engineering, Chengdu university, Chengdu 610106, China
| | - Yao Wang
- Sichuan Industrial Institute of Antibiotics, School of Pharmacy, Chengdu university, Chengdu 610106, China
| | - Qi Tang
- Sichuan Industrial Institute of Antibiotics, School of Pharmacy, Chengdu university, Chengdu 610106, China
| | - Dan Yin
- Sichuan Industrial Institute of Antibiotics, School of Pharmacy, Chengdu university, Chengdu 610106, China
| | - Chuane Tang
- School of Mechanical Engineering, Chengdu university, Chengdu 610106, China
| | - En He
- Key Laboratory of Coarse Cereal Processing (Ministry of Agriculture and Rural Affairs), School of Food and Biological Engineering, Chengdu university, Chengdu 610106, China
| | - Liang Zou
- Key Laboratory of Coarse Cereal Processing (Ministry of Agriculture and Rural Affairs), School of Food and Biological Engineering, Chengdu university, Chengdu 610106, China.
| | - Qiang Peng
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China.
| |
Collapse
|
181
|
Yazdian-Robati R, Hedayati N, Dehghani S, Ramezani M, Alibolandi M, Saeedi M, Abnous K, Taghdisi SM. Application of the catalytic activity of gold nanoparticles for development of optical aptasensors. Anal Biochem 2021; 629:114307. [PMID: 34273317 DOI: 10.1016/j.ab.2021.114307] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2021] [Revised: 06/04/2021] [Accepted: 07/11/2021] [Indexed: 01/15/2023]
Abstract
Biosensor technology is considered to be a great alternative in analytical techniques over the conventional methods. Among many recently developed techniques and devices, aptasensors are interesting because of their high specificity, selectivity and sensitivity. Combining aptamer as a biological recognition element with gold nanoparticles (AuNPs) as probe, are becoming more general owing to their beneficial properties, including low cost and ability to analyze specific targets on-site and using naked eye. Hydrogen bonds, nucleic acid hybridization, aptamer-target and antigen-antibody binding, Raman signature, enzyme inhibition, and enzyme-mimicking activity are main different sensing strategies exploited in AuNPs-based optical aptasensors. In this review article, we discussed the recent advances in optical aptasensors with a special emphasis on the catalytic activity property of AuNPs.
Collapse
Affiliation(s)
- Rezvan Yazdian-Robati
- Molecular and Cell Biology Research Center, Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
| | - Narges Hedayati
- Department of Pharmacodynamics and Toxicology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Shahrzad Dehghani
- Department of Pharmaceutical Biotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mohammad Ramezani
- Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mona Alibolandi
- Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Majid Saeedi
- Department of Pharmaceutics, Faculty of Pharmacy, Mazandaran University of Medical Sciences, Sari, Iran
| | - Khalil Abnous
- Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Medicinal Chemistry, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran.
| | - Seyed Mohammad Taghdisi
- Targeted Drug Delivery Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
182
|
Stanicki D, Larbanoix L, Boutry S, Vangijzegem T, Ternad I, Garifo S, Muller RN, Laurent S. Impact of the chain length on the biodistribution profiles of PEGylated iron oxide nanoparticles: a multimodal imaging study. J Mater Chem B 2021; 9:5055-5068. [PMID: 34132320 DOI: 10.1039/d1tb00573a] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Bimodal sub-5 nm superparamagnetic iron oxide nanoparticles (SPIO-5) coated with polyethylene glycol of different chain lengths (i.e. PEG-800, -2000 and -5000) have been prepared and characterized. Fluorescence properties have been obtained by mean of the grafting of a near-infrared-emitting dye (NIR-dye) onto the surface of the oxide, thanks to the carboxylic acid functions introduced towards an organosilane coating. Such modification allowed us to follow in vivo their biodistribution and elimination pathways by T1-w and T2-w high-field magnetic resonance imaging (MRI), as well as by optical and optoacoustic imaging. Interestingly, it has been highlighted that for a given composition, the thickness of the coating strongly influences the pharmacokinetic properties of the administrated SPIO-5.
Collapse
Affiliation(s)
- Dimitri Stanicki
- General, Organic and Biomedical Chemistry Units, NMR and Molecular Imaging Laboratory, University of Mons, B-7000 Mons, Belgium.
| | | | | | | | | | | | | | | |
Collapse
|
183
|
Gorbet MJ, Singh A, Mao C, Fiering S, Ranjan A. Using nanoparticles for in situ vaccination against cancer: mechanisms and immunotherapy benefits. Int J Hyperthermia 2021; 37:18-33. [PMID: 33426995 DOI: 10.1080/02656736.2020.1802519] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Immunotherapy to treat cancer is now an established clinical approach. Immunotherapy can be applied systemically, as done with checkpoint blockade antibodies, but it can also be injected directly into identified tumors, in a strategy of in situ vaccination (ISV). ISV is designed to stimulate a strong local antitumor immune response involving both innate and adaptive immune cells, and through this generate a systemic antitumor immune response against metastatic tumors. A variety of ISVs have been utilized to generate an immunostimulatory tumor microenvironment (TME). These include attenuated microorganisms, recombinant proteins, small molecules, physical disruptors of TME (alternating magnetic and focused ultrasound heating, photothermal therapy, and radiotherapy), and more recently nanoparticles (NPs). NPs are attractive and unique since they can load multiple drugs or other reagents to influence immune and cancer cell functions in the TME, affording a unique opportunity to stimulate antitumor immunity. Here, we describe the NP-ISV therapeutic mechanisms, review chemically synthesized NPs (i.e., liposomes, polymeric, chitosan-based, inorganic NPs, etc.), biologically derived NPs (virus and bacteria-based NPs), and energy-activated NP-ISVs in the context of their use as local ISV. Data suggests that NP-ISVs can enhance outcomes of immunotherapeutic regimens including those utilizing tumor hyperthermia and checkpoint blockade therapies.
Collapse
Affiliation(s)
| | - Akansha Singh
- College of Veterinary Medicine, Oklahoma State University, Stillwater, OK, USA
| | - Chenkai Mao
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Hanover, NH, USA
| | - Steven Fiering
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Hanover, NH, USA.,Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Norris Cotton Cancer Center at Dartmouth and Dartmouth Hitchcock, Lebanon, NH, USA
| | - Ashish Ranjan
- College of Veterinary Medicine, Oklahoma State University, Stillwater, OK, USA
| |
Collapse
|
184
|
Mihalik NE, Wen S, Driesschaert B, Eubank TD. Formulation and In Vitro Characterization of PLGA/PLGA-PEG Nanoparticles Loaded with Murine Granulocyte-Macrophage Colony-Stimulating Factor. AAPS PharmSciTech 2021; 22:191. [PMID: 34169366 DOI: 10.1208/s12249-021-02049-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2021] [Accepted: 05/11/2021] [Indexed: 02/06/2023] Open
Abstract
Granulocyte-macrophage colony-stimulating factor (GM-CSF) has demonstrated notable clinical activity in cancer immunotherapy, but it is limited by systemic toxicities, poor bioavailability, rapid clearance, and instability in vivo. Nanoparticles (NPs) may overcome these limitations and provide a mechanism for passive targeting of tumors. This study aimed to develop GM-CSF-loaded PLGA/PLGA-PEG NPs and evaluate them in vitro as a potential candidate for in vivo administration. NPs were created by a phase-separation technique that did not require toxic/protein-denaturing solvents or harsh agitation techniques and encapsulated GM-CSF in a more stable precipitated form. NP sizes were within 200 nm for enhanced permeability and retention (EPR) effect with negative zeta potentials, spherical morphology, and high entrapment efficiencies. The optimal formulation was identified by sustained release of approximately 70% of loaded GM-CSF over 24 h, alongside an average size of 143 ± 35 nm and entrapment efficiency of 84 ± 5%. These NPs were successfully freeze-dried in 5% (w/v) hydroxypropyl-β-cyclodextrin for long-term storage and further characterized. Bioactivity of released GM-CSF was determined by observing GM-CSF receptor activation on murine monocytes and remained fully intact. NPs were not cytotoxic to murine bone marrow-derived macrophages (BMDMs) at concentrations up to 1 mg/mL as determined by MTT and trypan blue exclusion assays. Lastly, NP components generated no significant transcription of inflammation-regulating genes from BMDMs compared to IFNγ+LPS "M1" controls. This report lays the preliminary groundwork to validate in vivo studies with GM-CSF-loaded PLGA/PEG-PLGA NPs for tumor immunomodulation. Overall, these data suggest that in vivo delivery will be well tolerated.
Collapse
|
185
|
Song J, Ju Y, Amarasena TH, Lin Z, Mettu S, Zhou J, Rahim MA, Ang CS, Cortez-Jugo C, Kent SJ, Caruso F. Influence of Poly(ethylene glycol) Molecular Architecture on Particle Assembly and Ex Vivo Particle-Immune Cell Interactions in Human Blood. ACS NANO 2021; 15:10025-10038. [PMID: 34009935 DOI: 10.1021/acsnano.1c01642] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/28/2023]
Abstract
Poly(ethylene glycol) (PEG) is widely used in particle assembly to impart biocompatibility and stealth-like properties in vivo for diverse biomedical applications. Previous studies have examined the effect of PEG molecular weight and PEG coating density on the biological fate of various particles; however, there are few studies that detail the fundamental role of PEG molecular architecture in particle engineering and bio-nano interactions. Herein, we engineered PEG particles using a mesoporous silica (MS) templating method and investigated how the PEG building block architecture impacted the physicochemical properties (e.g., surface chemistry and mechanical characteristics) of the PEG particles and subsequently modulated particle-immune cell interactions in human blood. Varying the PEG architecture from 3-arm to 4-arm, 6-arm, and 8-arm generated PEG particles with a denser, stiffer structure, with increasing elastic modulus from 1.5 to 14.9 kPa, inducing an increasing level of immune cell association (from 15% for 3-arm to 45% for 8-arm) with monocytes. In contrast, the precursor PEG particles with the template intact (MS@PEG) were stiffer and generally displayed higher levels of immune cell association but showed the opposite trend-immune cell association decreased with increasing PEG arm numbers. Proteomics analysis demonstrated that the biomolecular corona that formed on the PEG particles minimally influenced particle-immune cell interactions, whereas the MS@PEG particle-cell interactions correlated with the composition of the corona that was abundant in histidine-rich glycoproteins. Our work highlights the role of PEG architecture in the design of stealth PEG-based particles, thus providing a link between the synthetic nature of particles and their biological behavior in blood.
Collapse
Affiliation(s)
- Jiaying Song
- ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, and the Department of Chemical Engineering, The University of Melbourne, Parkville, Victoria 3010, Australia
| | - Yi Ju
- ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, and the Department of Chemical Engineering, The University of Melbourne, Parkville, Victoria 3010, Australia
| | - Thakshila H Amarasena
- ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, and Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Parkville, Victoria 3010, Australia
| | - Zhixing Lin
- ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, and the Department of Chemical Engineering, The University of Melbourne, Parkville, Victoria 3010, Australia
| | - Srinivas Mettu
- Department of Chemical Engineering, The University of Melbourne, Parkville, Victoria 3010, Australia
| | - Jiajing Zhou
- ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, and the Department of Chemical Engineering, The University of Melbourne, Parkville, Victoria 3010, Australia
| | - Md Arifur Rahim
- ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, and the Department of Chemical Engineering, The University of Melbourne, Parkville, Victoria 3010, Australia
| | - Ching-Seng Ang
- Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Parkville, Victoria 3010, Australia
| | - Christina Cortez-Jugo
- ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, and the Department of Chemical Engineering, The University of Melbourne, Parkville, Victoria 3010, Australia
| | - Stephen J Kent
- ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, and Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Parkville, Victoria 3010, Australia
| | - Frank Caruso
- ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, and the Department of Chemical Engineering, The University of Melbourne, Parkville, Victoria 3010, Australia
| |
Collapse
|
186
|
Zhang Z, Wen Y, Song X, Zhu J, Li J. Nonviral DNA Delivery System with Supramolecular PEGylation Formed by Host-Guest Pseudo-Block Copolymers. ACS APPLIED BIO MATERIALS 2021; 4:5057-5070. [PMID: 35007054 DOI: 10.1021/acsabm.1c00306] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
A cationic supramolecular system based on host-guest pseudoblock copolymers was developed for nonviral DNA delivery. In this system, the macromolecular host was a cationic star-shaped polymer composed of a β-cyclodextrin (β-CD) core and multiple poly(2-(dimethylamino)ethyl methacrylate) (PDMAEMA) chains grafted on the core, while the macromolecular guest was a linear adamantyl-ended poly(ethylene glycol) (mPEG-Ad). Pseudoblock copolymers were self-assembled from the polymeric host-guest pairs (typically, 1:1 molar ratio) in aqueous media through the inclusion of an adamantyl group at the end of guest polymer into the β-CD cavity of host polymers. Through such an approach, the resultant supramolecular system was integrated with not only a superior DNA condensing ability due to the host polymer but also an outstanding polyplex-stabilizing ability as well as biocompatibility due to the guest polymer. The cationic star-shaped host polymers alone were capable of condensing plasmid DNA efficiently into nanoparticles (70-100 nm) with positive surface charge. They showed obviously lower cytotoxicity than PEI 25K (commercial branched polyethylenimine with a molecular weight around 25 kDa) in cell lines of L929, MB231, and Hela under high dose. In serum-free or serum-containing culture conditions, these host polymers exhibited either higher or lower in vitro DNA transfection efficiency as compared with PEI 25K in the three cell lines under study, which was dependent on the N/P ratios and PDMAEMA arm length. Upon incorporation of the PEG block through host-guest complexation with mPEG-Ad (i.e., supramolecular PEGylation), the resulting host-guest supramolecular systems exhibited even lower cytotoxicity than the host polymers alone. The polyplexes between plasmid DNA (pDNA) and the host-guest systems showed significantly improved stability in BSA-PBS buffer solution (pH 7.4) and enhanced in vitro DNA transfection efficiency in the cases of higher N/P ratios or longer PDMAEMA arms in all tested cell lines under both serum-free and serum-containing culture conditions, as compared with the corresponding polyplexes without supramolecular PEGylation. Further, through forming pseudoblock copolymer, the DNA transfection ability of the supramolecular system can be easily modulated and optimized either by changing the ratio between the guest and host or by using different hosts with varied PDMAEMA arm lengths.
Collapse
Affiliation(s)
- Zhongxing Zhang
- Department of Biomedical Engineering, Faculty of Engineering, National University of Singapore, Singapore 117574, Singapore
| | - Yuting Wen
- Department of Biomedical Engineering, Faculty of Engineering, National University of Singapore, Singapore 117574, Singapore
| | - Xia Song
- Department of Biomedical Engineering, Faculty of Engineering, National University of Singapore, Singapore 117574, Singapore
| | - Jingling Zhu
- Department of Biomedical Engineering, Faculty of Engineering, National University of Singapore, Singapore 117574, Singapore
| | - Jun Li
- Department of Biomedical Engineering, Faculty of Engineering, National University of Singapore, Singapore 117574, Singapore
| |
Collapse
|
187
|
An L, Tao Q, Wu Y, Wang N, Liu Y, Wang F, Zhang L, Shi A, Zhou X, Yu S, Zhang J. Synthesis of SPIO Nanoparticles and the Subsequent Applications in Stem Cell Labeling for Parkinson's Disease. NANOSCALE RESEARCH LETTERS 2021; 16:107. [PMID: 34128153 PMCID: PMC8203769 DOI: 10.1186/s11671-021-03540-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/24/2020] [Accepted: 04/28/2021] [Indexed: 06/12/2023]
Abstract
Parkinson's disease (PD) is characterized by the progressive loss of dopaminergic neurons in the midbrain, and the stem cell transplantation method provides a promising strategy for the treatment. In these studies, tracking the biological behaviors of the transplanted cells in vivo is essential for a basic understanding of stem cell function and evaluation of clinical effectiveness. In the present study, we developed a novel ultrasmall superparamagnetic iron oxide nanoparticles coating with the polyacrylic acid (PAA) and methoxypolyethylene glycol amine (PEG) by thermal decomposition method and a two-step modification. The USPIO-PAA/PEG NPs have a uniform diameter of 10.07 ± 0.55 nm and proper absorption peak of the corresponding ligands, as showed by TEM and FTIR. MTT showed that the survival of cells incubated with USPIO-PAA/PEG NPs remained above 96%. The synthesized USPIO-PAA/PEG had a good relaxation rate of 84.65 s-1 Mm-1, indicating that they could be efficiently uptake and traced by MRI. Furthermore, the primary human adipose-derived stem cells (HADSCs) were characterized, labeled with USPIO-PAA/PEG and transplanted into the striatum of 6-hydroxydopamine (6-OHDA)-induced PD rat models. The labeled cells could be traced by MRI for up to 3 weeks after the transplantation surgery; moreover, transplantation with the labeled HADSCs significantly attenuated apomorphine-induced rotations in PD models and increased the number of the dopaminergic neurons in the substania nigra. Overall, the development of USPIO-PAA/PEG NPs provides a promising tool for in vivo tracing technique of cell therapy and identifies a novel strategy to track stem cells with therapeutic potential in PD.
Collapse
Affiliation(s)
- Li An
- Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Sciences, No. 88 Keling Road, Suzhou New District, Suzhou, 215163, China
| | - Qing Tao
- Department of Radiology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Yue Wu
- Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Sciences, No. 88 Keling Road, Suzhou New District, Suzhou, 215163, China
| | - Nana Wang
- Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Sciences, No. 88 Keling Road, Suzhou New District, Suzhou, 215163, China
| | - Yan Liu
- Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Sciences, No. 88 Keling Road, Suzhou New District, Suzhou, 215163, China
- Zhengzhou Institute of Engineering and Technology Affiliated with SIBET, Zhengzhou, 450001, China
| | - Feifei Wang
- Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Sciences, No. 88 Keling Road, Suzhou New District, Suzhou, 215163, China
- Zhengzhou Institute of Engineering and Technology Affiliated with SIBET, Zhengzhou, 450001, China
| | - Lixing Zhang
- Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Sciences, No. 88 Keling Road, Suzhou New District, Suzhou, 215163, China
- Zhengzhou Institute of Engineering and Technology Affiliated with SIBET, Zhengzhou, 450001, China
| | - Aihua Shi
- Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Sciences, No. 88 Keling Road, Suzhou New District, Suzhou, 215163, China
- Zhengzhou Institute of Engineering and Technology Affiliated with SIBET, Zhengzhou, 450001, China
| | - Xiumin Zhou
- Department of Oncology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Shuang Yu
- Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Sciences, No. 88 Keling Road, Suzhou New District, Suzhou, 215163, China.
| | - Jingzhong Zhang
- Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Sciences, No. 88 Keling Road, Suzhou New District, Suzhou, 215163, China.
- Zhengzhou Institute of Engineering and Technology Affiliated with SIBET, Zhengzhou, 450001, China.
- Tianjin Guokeyigong Science and Technology Development Company Limited, Tianjin, 300399, China.
| |
Collapse
|
188
|
Frutiger A, Tanno A, Hwu S, Tiefenauer RF, Vörös J, Nakatsuka N. Nonspecific Binding-Fundamental Concepts and Consequences for Biosensing Applications. Chem Rev 2021; 121:8095-8160. [PMID: 34105942 DOI: 10.1021/acs.chemrev.1c00044] [Citation(s) in RCA: 118] [Impact Index Per Article: 29.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Nature achieves differentiation of specific and nonspecific binding in molecular interactions through precise control of biomolecules in space and time. Artificial systems such as biosensors that rely on distinguishing specific molecular binding events in a sea of nonspecific interactions have struggled to overcome this issue. Despite the numerous technological advancements in biosensor technologies, nonspecific binding has remained a critical bottleneck due to the lack of a fundamental understanding of the phenomenon. To date, the identity, cause, and influence of nonspecific binding remain topics of debate within the scientific community. In this review, we discuss the evolution of the concept of nonspecific binding over the past five decades based upon the thermodynamic, intermolecular, and structural perspectives to provide classification frameworks for biomolecular interactions. Further, we introduce various theoretical models that predict the expected behavior of biosensors in physiologically relevant environments to calculate the theoretical detection limit and to optimize sensor performance. We conclude by discussing existing practical approaches to tackle the nonspecific binding challenge in vitro for biosensing platforms and how we can both address and harness nonspecific interactions for in vivo systems.
Collapse
Affiliation(s)
- Andreas Frutiger
- Laboratory of Biosensors and Bioelectronics, Institute for Biomedical Engineering, ETH Zürich, Zürich CH-8092, Switzerland
| | - Alexander Tanno
- Laboratory of Biosensors and Bioelectronics, Institute for Biomedical Engineering, ETH Zürich, Zürich CH-8092, Switzerland
| | - Stephanie Hwu
- Laboratory of Biosensors and Bioelectronics, Institute for Biomedical Engineering, ETH Zürich, Zürich CH-8092, Switzerland
| | - Raphael F Tiefenauer
- Laboratory of Biosensors and Bioelectronics, Institute for Biomedical Engineering, ETH Zürich, Zürich CH-8092, Switzerland
| | - János Vörös
- Laboratory of Biosensors and Bioelectronics, Institute for Biomedical Engineering, ETH Zürich, Zürich CH-8092, Switzerland
| | - Nako Nakatsuka
- Laboratory of Biosensors and Bioelectronics, Institute for Biomedical Engineering, ETH Zürich, Zürich CH-8092, Switzerland
| |
Collapse
|
189
|
Miyazawa T, Itaya M, Burdeos GC, Nakagawa K, Miyazawa T. A Critical Review of the Use of Surfactant-Coated Nanoparticles in Nanomedicine and Food Nanotechnology. Int J Nanomedicine 2021; 16:3937-3999. [PMID: 34140768 PMCID: PMC8203100 DOI: 10.2147/ijn.s298606] [Citation(s) in RCA: 75] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2021] [Accepted: 03/31/2021] [Indexed: 12/12/2022] Open
Abstract
Surfactants, whose existence has been recognized as early as 2800 BC, have had a long history with the development of human civilization. With the rapid development of nanotechnology in the latter half of the 20th century, breakthroughs in nanomedicine and food nanotechnology using nanoparticles have been remarkable, and new applications have been developed. The technology of surfactant-coated nanoparticles, which provides new functions to nanoparticles for use in the fields of nanomedicine and food nanotechnology, is attracting a lot of attention in the fields of basic research and industry. This review systematically describes these "surfactant-coated nanoparticles" through various sections in order: 1) surfactants, 2) surfactant-coated nanoparticles, application of surfactant-coated nanoparticles to 3) nanomedicine, and 4) food nanotechnology. Furthermore, current progress and problems of the technology using surfactant-coated nanoparticles through recent research reports have been discussed.
Collapse
Affiliation(s)
- Taiki Miyazawa
- New Industry Creation Hatchery Center (NICHe), Tohoku University, Sendai, Miyagi, Japan
| | - Mayuko Itaya
- Food and Biodynamic Chemistry Laboratory, Graduate School of Agricultural Science, Tohoku University, Sendai, Miyagi, Japan
| | - Gregor C Burdeos
- Institute for Animal Nutrition and Physiology, Christian Albrechts University Kiel, Kiel, Germany
| | - Kiyotaka Nakagawa
- Food and Biodynamic Chemistry Laboratory, Graduate School of Agricultural Science, Tohoku University, Sendai, Miyagi, Japan
| | - Teruo Miyazawa
- New Industry Creation Hatchery Center (NICHe), Tohoku University, Sendai, Miyagi, Japan
| |
Collapse
|
190
|
Characterizing block-copolymer micelles used in nanomedicines via solution static scattering techniques. Polym J 2021. [DOI: 10.1038/s41428-021-00489-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
AbstractBlock copolymers are well recognized as excellent nanotools for delivering hydrophobic drugs. The formulation of such delivery nanoparticles requires robust characterization and clarification of the critical quality attributes correlating with the safety and efficacy of the drug before applying to regulatory authorities for approval. Static solution scattering from block copolymers is one such technique. This paper first outlines the theoretical background and current models for analyzing this scattering and then presents an overview of our recent studies on block copolymers.
Collapse
|
191
|
Zahiri M, Taghdisi SM, Abnous K, Ramezani M, Alibolandi M. Fabrication of versatile targeted lipopolymersomes for improved camptothecin efficacy against colon adenocarcinoma in vitro and in vivo. Expert Opin Drug Deliv 2021; 18:1309-1322. [PMID: 33970721 DOI: 10.1080/17425247.2021.1928631] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
BACKGROUND Hybrid vesicular systems (lipopolymersomes) are promising platforms for minimizing the liposomes and polymersomes disadvantages in terms of chemotherapeutic transportation. In this regard, lipopolymersome has been designed to integrate the advantage of both polymersomes and liposomes to enable better structural integrity of the bilayer after encapsulation of hydrophobic drugs while maintaining the soft nature of liposomes, superior serum stability, and high encapsulation efficiency of cargos in the bilayer segment. RESEARCH DESIGN AND METHODS In the present study, we reported preparation and characterization of five camptothecin (CPT)-loaded lipopolymersomal formulations composed of poly (ethylene glycol)-poly (lactic acid) (PEG-PLA) and dipalmitoylphosphatidylcholine (DPPC) at different molar ratios using film rehydration method. Afterward, the preferred formulation was tagged with AS1411 DNA aptamer in order to evaluate the therapeutic index using nucleolin-positive colon cancer cell lines (HT29 and C26). RESULTS The obtained data indicated that the prepared CPT-loaded lipopolymersome at a PEG-PLA: DPPC ratio of 75:25 exhibited superior stability and high loading capacity compared to other systems. Moreover, high cytotoxicity of the aptamer-targeted lipopolymersome and increased tumor accumulation were observed in comparison with non-targeted one. CONCLUSIONS The designed polymer-rich lipopolymersomal platform offers bright future for the development of potent nanomedicine against cancer.
Collapse
Affiliation(s)
- Mahsa Zahiri
- Department of Pharmaceutical Nanotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran.,Student Research Committee, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Seyed Mohammad Taghdisi
- Targeted Drug Delivery Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran.,Department of Pharmaceutical Biotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Khalil Abnous
- Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran.,Department of Medicinal Chemistry, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mohammad Ramezani
- Department of Pharmaceutical Biotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran.,Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mona Alibolandi
- Department of Pharmaceutical Biotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran.,Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
192
|
Characterization and biodistribution of Au nanoparticles loaded in PLGA nanocarriers using an original encapsulation process. Colloids Surf B Biointerfaces 2021; 205:111875. [PMID: 34058691 DOI: 10.1016/j.colsurfb.2021.111875] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Revised: 05/05/2021] [Accepted: 05/19/2021] [Indexed: 11/20/2022]
Abstract
Due to their imaging and radiosensitizing properties, ultrasmall gadolinium chelate-coated gold nanoparticles (AuNP) represent a promising approach in the diagnosis and the treatment of tumors. However, their poor pharmacokinetic profile, especially their rapid renal clearance prevents from an efficient exploitation of their potential for medical applications. The present study focuses on a strategy which resides in the encapsulation of AuNP in large polymeric NP to avoid the glomerular filtration and then to prolong the vascular residence time. An original encapsulation procedure using the polyethyleneimine (PEI) was set up to electrostatically entrap AuNP in biodegradable poly(lactic-co-glycolic acid) (PLGA) and polyethylene glycol -PLGA (PLGA-PEG) NP. Hydrodynamic diameters of NP were dependent of the PEI/Au ratio and comprised between 115 and 196 nm for ratios equal or superior to 4. Encapsulation yield was close to 90 % whereas no loading was observed without PEI. No toxicity was observed after 24 h exposure in hepatocyte cell-lines. Entrapement of AuNP in polymeric nanocarriers facilitated the passive uptake in cancer cells after only 2 h incubation. In healthy rat, the encapsulation allowed increasing the gold concentration in the blood within the first hour after intravenous administration. Polymeric nanoparticles were sequestered in the liver and the spleen rather than the kidneys. T1-weighted magnetic resonance demonstrated that encapsulation process did not alter the contrast agent properties of gadolinium. The encapsulation of the gold nanoparticles in PLGA particles paves the way to innovative imaging-guided anticancer therapies in personalized medicine.
Collapse
|
193
|
Sousa de Almeida M, Susnik E, Drasler B, Taladriz-Blanco P, Petri-Fink A, Rothen-Rutishauser B. Understanding nanoparticle endocytosis to improve targeting strategies in nanomedicine. Chem Soc Rev 2021; 50:5397-5434. [PMID: 33666625 PMCID: PMC8111542 DOI: 10.1039/d0cs01127d] [Citation(s) in RCA: 484] [Impact Index Per Article: 121.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2020] [Indexed: 12/19/2022]
Abstract
Nanoparticles (NPs) have attracted considerable attention in various fields, such as cosmetics, the food industry, material design, and nanomedicine. In particular, the fast-moving field of nanomedicine takes advantage of features of NPs for the detection and treatment of different types of cancer, fibrosis, inflammation, arthritis as well as neurodegenerative and gastrointestinal diseases. To this end, a detailed understanding of the NP uptake mechanisms by cells and intracellular localization is essential for safe and efficient therapeutic applications. In the first part of this review, we describe the several endocytic pathways involved in the internalization of NPs and we discuss the impact of the physicochemical properties of NPs on this process. In addition, the potential challenges of using various inhibitors, endocytic markers and genetic approaches to study endocytosis are addressed along with the principal (semi) quantification methods of NP uptake. The second part focuses on synthetic and bio-inspired substances, which can stimulate or decrease the cellular uptake of NPs. This approach could be interesting in nanomedicine where a high accumulation of drugs in the target cells is desirable and clearance by immune cells is to be avoided. This review contributes to an improved understanding of NP endocytic pathways and reveals potential substances, which can be used in nanomedicine to improve NP delivery.
Collapse
Affiliation(s)
- Mauro Sousa de Almeida
- Adolphe Merkle Institute, University of FribourgChemin des Verdiers 41700 FribourgSwitzerland
| | - Eva Susnik
- Adolphe Merkle Institute, University of FribourgChemin des Verdiers 41700 FribourgSwitzerland
| | - Barbara Drasler
- Adolphe Merkle Institute, University of FribourgChemin des Verdiers 41700 FribourgSwitzerland
| | | | - Alke Petri-Fink
- Adolphe Merkle Institute, University of FribourgChemin des Verdiers 41700 FribourgSwitzerland
- Department of Chemistry, University of FribourgChemin du Musée 91700 FribourgSwitzerland
| | | |
Collapse
|
194
|
Kumar R, Santa Chalarca CF, Bockman MR, Bruggen CV, Grimme CJ, Dalal RJ, Hanson MG, Hexum JK, Reineke TM. Polymeric Delivery of Therapeutic Nucleic Acids. Chem Rev 2021; 121:11527-11652. [PMID: 33939409 DOI: 10.1021/acs.chemrev.0c00997] [Citation(s) in RCA: 198] [Impact Index Per Article: 49.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The advent of genome editing has transformed the therapeutic landscape for several debilitating diseases, and the clinical outlook for gene therapeutics has never been more promising. The therapeutic potential of nucleic acids has been limited by a reliance on engineered viral vectors for delivery. Chemically defined polymers can remediate technological, regulatory, and clinical challenges associated with viral modes of gene delivery. Because of their scalability, versatility, and exquisite tunability, polymers are ideal biomaterial platforms for delivering nucleic acid payloads efficiently while minimizing immune response and cellular toxicity. While polymeric gene delivery has progressed significantly in the past four decades, clinical translation of polymeric vehicles faces several formidable challenges. The aim of our Account is to illustrate diverse concepts in designing polymeric vectors towards meeting therapeutic goals of in vivo and ex vivo gene therapy. Here, we highlight several classes of polymers employed in gene delivery and summarize the recent work on understanding the contributions of chemical and architectural design parameters. We touch upon characterization methods used to visualize and understand events transpiring at the interfaces between polymer, nucleic acids, and the physiological environment. We conclude that interdisciplinary approaches and methodologies motivated by fundamental questions are key to designing high-performing polymeric vehicles for gene therapy.
Collapse
Affiliation(s)
- Ramya Kumar
- Department of Chemistry, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | | | - Matthew R Bockman
- Department of Chemistry, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Craig Van Bruggen
- Department of Chemistry, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Christian J Grimme
- Department of Chemical Engineering and Materials Science, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Rishad J Dalal
- Department of Chemistry, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Mckenna G Hanson
- Department of Chemistry, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Joseph K Hexum
- Department of Chemistry, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Theresa M Reineke
- Department of Chemistry, University of Minnesota, Minneapolis, Minnesota 55455, United States
| |
Collapse
|
195
|
Ruan S, Zhou Y, Jiang X, Gao H. Rethinking CRITID Procedure of Brain Targeting Drug Delivery: Circulation, Blood Brain Barrier Recognition, Intracellular Transport, Diseased Cell Targeting, Internalization, and Drug Release. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2021; 8:2004025. [PMID: 33977060 PMCID: PMC8097396 DOI: 10.1002/advs.202004025] [Citation(s) in RCA: 109] [Impact Index Per Article: 27.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Revised: 12/03/2020] [Indexed: 05/06/2023]
Abstract
The past decades have witnessed great progress in nanoparticle (NP)-based brain-targeting drug delivery systems, while their therapeutic potentials are yet to be fully exploited given that the majority of them are lost during the delivery process. Rational design of brain-targeting drug delivery systems requires a deep understanding of the entire delivery process along with the issues that they may encounter. Herein, this review first analyzes the typical delivery process of a systemically administrated NPs-based brain-targeting drug delivery system and proposes a six-step CRITID delivery cascade: circulation in systemic blood, recognizing receptor on blood-brain barrier (BBB), intracellular transport, diseased cell targeting after entering into parenchyma, internalization by diseased cells, and finally intracellular drug release. By dissecting the entire delivery process into six steps, this review seeks to provide a deep understanding of the issues that may restrict the delivery efficiency of brain-targeting drug delivery systems as well as the specific requirements that may guarantee minimal loss at each step. Currently developed strategies used for troubleshooting these issues are reviewed and some state-of-the-art design features meeting these requirements are highlighted. The CRITID delivery cascade can serve as a guideline for designing more efficient and specific brain-targeting drug delivery systems.
Collapse
Affiliation(s)
- Shaobo Ruan
- Key laboratory of Drug Targeting and Drug Delivery Systems of the Education MinistrySichuan Engineering Laboratory for Plant‐sourced Drug and Sichuan Research Center for Drug Precision Industrial TechnologyWest China School of PharmacySichuan UniversityChengdu610041China
- Department of PharmaceuticsCollege of PharmacyUniversity of FloridaGainesvilleFlorida32610USA
| | - Yang Zhou
- Key laboratory of Drug Targeting and Drug Delivery Systems of the Education MinistrySichuan Engineering Laboratory for Plant‐sourced Drug and Sichuan Research Center for Drug Precision Industrial TechnologyWest China School of PharmacySichuan UniversityChengdu610041China
| | - Xinguo Jiang
- Key laboratory of Smart Drug DeliveryMinistry of EducationSchool of PharmacyFudan UniversityShanghai201203China
| | - Huile Gao
- Key laboratory of Drug Targeting and Drug Delivery Systems of the Education MinistrySichuan Engineering Laboratory for Plant‐sourced Drug and Sichuan Research Center for Drug Precision Industrial TechnologyWest China School of PharmacySichuan UniversityChengdu610041China
| |
Collapse
|
196
|
de Castro RR, do Carmo FA, Martins C, Simon A, de Sousa VP, Rodrigues CR, Cabral LM, Sarmento B. Clofazimine functionalized polymeric nanoparticles for brain delivery in the tuberculosis treatment. Int J Pharm 2021; 602:120655. [PMID: 33915184 DOI: 10.1016/j.ijpharm.2021.120655] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Revised: 03/31/2021] [Accepted: 04/23/2021] [Indexed: 02/06/2023]
Abstract
Central nervous system tuberculosis (CNS-TB) is the most severe form of the disease especially due to the inability of therapeutics to cross the blood-brain barrier (BBB). Clofazimine (CFZ) stands out for presenting high in vitro activity against multi-drug resistant strains of Mycobacterium tuberculosis, however, CFZ physicochemical and pharmacokinetics properties limit drug penetration into the CNS and, consequently, its clinical use. The aim of this work was to develop polymeric nanoparticles (NPs) of poly(lactic-co-glycolic acid) (PLGA) and polyethylene glycol (PEG) loaded with CFZ and functionalized with a transferrin receptor (TfR)-binding peptide, aiming brain drug delivery for CNS-TB treatment by the intravenous route. The poor water solubility and high lipophilicity of CFZ was overcome through its entrapment into PLGA-PEG NPs manufactured by both conventional and microfluidic techniques using the nanoprecipitation principle. In vitro studies in brain endothelial hCMEC/D3 cells demonstrated that CFZ incorporation into the NPs was advantageous to reduce drug cytotoxicity. The TfR-binding peptide-functionalized NPs showed superior cell interaction and higher CFZ permeability across hCMEC/D3 cell monolayers compared to the non-functionalized NP control, thus indicating the efficacy of the functionalization strategy on providing CFZ transport through the BBB in vitro. The functionalized NPs demonstrate suitability for CFZ biological administration, suggested with low plasma protein binding, off-target biodistribution and precise delivery of CFZ towards the brain parenchyma.
Collapse
Affiliation(s)
- Renata Ribeiro de Castro
- Department of Drugs and Pharmaceutics, Faculty of Pharmacy, Universidade Federal do Rio de Janeiro, Avenida Carlos Chagas Filho 373, 21941-902 Rio de Janeiro, Brazil; Laboratory of Molecular Pharmacology, Institute of Drug Technology (Farmanguinhos), Oswaldo Cruz Foundation, Rua Sizenando Nabuco 100, 21041-250 Rio de Janeiro, Brazil
| | - Flavia Almada do Carmo
- Department of Drugs and Pharmaceutics, Faculty of Pharmacy, Universidade Federal do Rio de Janeiro, Avenida Carlos Chagas Filho 373, 21941-902 Rio de Janeiro, Brazil
| | - Cláudia Martins
- INEB-Instituto de Engenharia Biomédica, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal; i3S-Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal; ICBAS - Instituto Ciências Biomédicas Abel Salazar, Universidade do Porto, Rua de Jorge Viterbo Ferreira 228, 4050-313 Porto, Portugal
| | - Alice Simon
- Department of Drugs and Pharmaceutics, Faculty of Pharmacy, Universidade Federal do Rio de Janeiro, Avenida Carlos Chagas Filho 373, 21941-902 Rio de Janeiro, Brazil
| | - Valeria Pereira de Sousa
- Department of Drugs and Pharmaceutics, Faculty of Pharmacy, Universidade Federal do Rio de Janeiro, Avenida Carlos Chagas Filho 373, 21941-902 Rio de Janeiro, Brazil
| | - Carlos Rangel Rodrigues
- Department of Drugs and Pharmaceutics, Faculty of Pharmacy, Universidade Federal do Rio de Janeiro, Avenida Carlos Chagas Filho 373, 21941-902 Rio de Janeiro, Brazil
| | - Lucio Mendes Cabral
- Department of Drugs and Pharmaceutics, Faculty of Pharmacy, Universidade Federal do Rio de Janeiro, Avenida Carlos Chagas Filho 373, 21941-902 Rio de Janeiro, Brazil
| | - Bruno Sarmento
- INEB-Instituto de Engenharia Biomédica, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal; i3S-Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal; CESPU - Instituto de Investigação e Formação Avançada em Ciências e Tecnologias da Saúde and Instituto Universitário de Ciências da Saúde, Rua Central de Gandra 1317, 4585-116 Gandra, Portugal.
| |
Collapse
|
197
|
Wang N, Huang Z, Wang S, Lang M, Zhang X. Minocycline hydrochloride loaded mPEG-PCLA membranes: Preparation and in vitro evaluation for periodontitis therapy. J BIOACT COMPAT POL 2021. [DOI: 10.1177/0883911521992795] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
This study was aimed at alleviating shortcomings in the treatment of periodontitis by preparation of a biopolymer membrane loaded with minocycline hydrochloride (MH) inserted into periodontal pockets to treat infections. Monomethoxy-poly (ethylene glycol)-poly (ε-caprolactone-co-L-lactide) (mPEG-PCLA) is a biocompatible and biodegradable amphiphilic block copolymer. It, therefore, has attracted considerable attention in drug delivery systems and periodontal treatment. We chose it as a membrane material for MH-drug loading. The MH-loaded membranes were prepared by the solvent casting technique with the content of 5, 8 and 10 wt.%, respectively. Fourier transform infrared spectra (FTIR) revealed no interaction between MH and polymer. The drug-loaded membrane surface morphology was investigated by scanning electron microscopy (SEM). In vitro release studies showed that the initial drug release exceeded 40% within 24 h, followed by a sustained release for up to 2 weeks, which would enable the therapeutic level to maintain over a longer time. The antibacterial activity studies in vitro demonstrated a positive effect on the periodontal pathogen. MH drug-loaded membranes have no adverse effect on the growth of periodontal ligament fibroblasts in the MTT test. The study suggests that mPEG-PCLA membranes containing MH are a potential antibacterial drug delivery system for local treatment of periodontitis.
Collapse
Affiliation(s)
- Ningtao Wang
- Department of 2nd Dental Center, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine; College of Stomatology, Shanghai Jiao Tong University; National Center for Stomatology; National Clinical Research Center for Oral Diseases; Shanghai Key Laboratory of Stomatology, Shanghai, China
| | - Zhengmei Huang
- Department of Stomatology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Shenchun Wang
- School of Materials Science and Engineering, East China University of Science and Technology, Shanghai, China
| | - Meidong Lang
- School of Materials Science and Engineering, East China University of Science and Technology, Shanghai, China
| | - Xiuyin Zhang
- Department of Prosthodontics, Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
198
|
Zhang L, Chen C, Tay SS, Wen S, Cao C, Biro M, Jin D, Stenzel MH. Optimizing the Polymer Cloak for Upconverting Nanoparticles: An Evaluation of Bioactivity and Optical Performance. ACS APPLIED MATERIALS & INTERFACES 2021; 13:16142-16154. [PMID: 33787198 DOI: 10.1021/acsami.1c01922] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
The ability of upconversion nanoparticles (UCNPs) to convert low-energy near-infrared (NIR) light into high-energy visible-ultraviolet light has resulted in their development as novel contrast agents for biomedical imaging. However, UCNPs often succumb to poor colloidal stability in aqueous media, which can be conquered by decorating the nanoparticle surface with polymers. The polymer cloak, therefore, plays an instrumental role in ensuring good stability in biological media. This study aims to understand the relationship between the length and grafting density of the polymer shell on the physicochemical and biological properties of these core-shell UCNPs. Poly(ethylene glycol) methyl ether methacrylate block ethylene glycol methacrylate phosphate (PPEGMEMAn-b-PEGMP3) with different numbers of PEGMEMA repeating units (26, 38, and 80) was prepared and attached to the UCNPs via the phosphate ligand of the poly(ethylene glycol methacrylate phosphate) (PEGMP) block at different polymer densities. The in vitro and in vivo protein corona, cellular uptake in two-dimensional (2D) monolayer and three-dimensional (3D) multicellular tumor spheroid (MCTS) models, and in vivo biodistribution in mice were evaluated. Furthermore, the photoluminescence of single-polymer-coated UCNPs was compared in solid state and cancer cells using laser scanning confocal microscopy (LSCM). Our results showed that the bioactivity and luminescence properties are chain length and grafting density dependent. The UCNPs coated with the longest PPEGMEMA chain, grafted at low brush density, were able to reduce the formation of the protein corona in vitro and in vivo, while these UCNPs also showed the brightest upconversion luminescence in the solid state. Moreover, these particular polymer-coated UCNPs showed enhanced cellular uptake, extended in vivo blood circulation time, and more accumulation in the liver, brain, and heart.
Collapse
Affiliation(s)
- Lin Zhang
- Cluster for Advanced Macromolecular Design (CAMD), School of Chemistry, University of New South Wales, Sydney, 2052 NSW, Australia
| | - Chaohao Chen
- Institute for Biomedical Materials and Devices, Faculty of Science, University of Technology Sydney, Sydney, 2006 NSW, Australia
| | - Szun S Tay
- EMBL Australia, Single Molecule Science Node, School of Medical Sciences, University of New South Wales, Sydney, 2052 NSW, Australia
| | - Shihui Wen
- Institute for Biomedical Materials and Devices, Faculty of Science, University of Technology Sydney, Sydney, 2006 NSW, Australia
| | - Cheng Cao
- Cluster for Advanced Macromolecular Design (CAMD), School of Chemistry, University of New South Wales, Sydney, 2052 NSW, Australia
| | - Maté Biro
- EMBL Australia, Single Molecule Science Node, School of Medical Sciences, University of New South Wales, Sydney, 2052 NSW, Australia
| | - Dayong Jin
- Institute for Biomedical Materials and Devices, Faculty of Science, University of Technology Sydney, Sydney, 2006 NSW, Australia
| | - Martina H Stenzel
- Cluster for Advanced Macromolecular Design (CAMD), School of Chemistry, University of New South Wales, Sydney, 2052 NSW, Australia
| |
Collapse
|
199
|
Huang W, Xiao G, Zhang Y, Min W. Research progress and application opportunities of nanoparticle-protein corona complexes. Biomed Pharmacother 2021; 139:111541. [PMID: 33848776 DOI: 10.1016/j.biopha.2021.111541] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2020] [Revised: 02/22/2021] [Accepted: 03/23/2021] [Indexed: 12/22/2022] Open
Abstract
Nanoparticles (NPs) can be used to design for nanomedicines with different chemical surface properties owing to their size advantages and the capacity of specific delivery to targeted sites in organisms. The discovery of the presence of protein corona (PC) has changed our classical view of NPs, stimulating researchers to investigate the in vivo fate of NPs as they enter biological systems. Both NPs and PC have their specificity but complement each other, so they should be considered as a whole. The formation and characterization of NP-PC complexes provide new insights into the design, functionalization, and application of nanocarriers. Based on progress of recent researches, we reviewed the formation, characterization, and composition of the PC, and introduced those critical factors influencing PC, simultaneously expound the effect of PC on the biological function of NPs. Especially we put forward the opportunities and challenges when NP-PC as a novel nano-drug carrier for targeted applications. Furthermore, we discussed the pros versus cons of the PC, as well as how to make better PC in the future application of NPs.
Collapse
Affiliation(s)
- Wei Huang
- Department of Pharmacy, The First People's Hospital of Jiande, Jiande 311600, China; Department of immunology, School of Basic Medical Sciences and School of Pharmacy, Nanchang University, Nanchang 330006, China
| | - Gao Xiao
- College of Environment and Resources, Fuzhou University, Fuzhou 350108, China
| | - Yujuan Zhang
- Department of immunology, School of Basic Medical Sciences and School of Pharmacy, Nanchang University, Nanchang 330006, China.
| | - Weiping Min
- Department of immunology, School of Basic Medical Sciences and School of Pharmacy, Nanchang University, Nanchang 330006, China
| |
Collapse
|
200
|
Erlichman JS, Leiter JC. Complexity of the Nano-Bio Interface and the Tortuous Path of Metal Oxides in Biological Systems. Antioxidants (Basel) 2021; 10:antiox10040547. [PMID: 33915992 PMCID: PMC8066112 DOI: 10.3390/antiox10040547] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2021] [Revised: 03/11/2021] [Accepted: 03/23/2021] [Indexed: 01/12/2023] Open
Abstract
Metal oxide nanoparticles (NPs) have received a great deal of attention as potential theranostic agents. Despite extensive work on a wide variety of metal oxide NPs, few chemically active metal oxide NPs have received Food and Drug Administration (FDA) clearance. The clinical translation of metal oxide NP activity, which often looks so promising in preclinical studies, has not progressed as rapidly as one might expect. The lack of FDA approval for metal oxide NPs appears to be a consequence of the complex transformation of NP chemistry as any given NP passes through multiple extra- and intracellular environments and interacts with a variety of proteins and transport processes that may degrade or transform the chemical properties of the metal oxide NP. Moreover, the translational models frequently used to study these materials do not represent the final therapeutic environment well, and studies in reduced preparations have, all too frequently, predicted fundamentally different physico-chemical properties from the biological activity observed in intact organisms. Understanding the evolving pharmacology of metal oxide NPs as they interact with biological systems is critical to establish translational test systems that effectively predict future theranostic activity.
Collapse
Affiliation(s)
- Joseph S. Erlichman
- Department of Biology, St. Lawrence University, Canton, NY 13617, USA
- Correspondence: ; Tel.: +1-(315)-229-5639
| | - James C. Leiter
- White River Junction VA Medical Center, White River Junction, VT 05009, USA;
| |
Collapse
|