1
|
Noda Y, Noguchi T, Nagano T, Aoki W, Ueda M. Bacterial removal using liposomes and an anionic adsorber. J Biosci Bioeng 2025; 139:249-256. [PMID: 39730254 DOI: 10.1016/j.jbiosc.2024.11.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2024] [Revised: 11/15/2024] [Accepted: 11/18/2024] [Indexed: 12/29/2024]
Abstract
Extracorporeal blood purification techniques using magnetic beads, which physically remove bacteria, fungi, viruses, and cytokines (disease agents) from the blood causing sepsis, have been studied. However, magnetic bead influx, which causes hemolysis and cytotoxicity, is an important issue. This study proposed a novel method for removing Escherichia coli from the blood using liposomes with high biocompatibility. To realize this method, a pegylated cationic liposome conjugated with antibodies (PCLA) that can simultaneously adsorb disease agents with the conjugated liposome antibodies and adhere to electrostatic absorbers was developed. E. coli was successfully adsorbed by PCLA in phosphate-buffered saline and electrostatically removed with a high removal efficiency of the antigen-antibody reaction (approximately 100 %). The removal efficiency of the antigen-antibody reaction in filtered bovine blood was approximately 50 %, demonstrating E. coli removal in the blood using the same method. Results suggested that this method can remove various disease agents from the blood by changing the antibody type.
Collapse
Affiliation(s)
- Yohei Noda
- Technology and Intellectual Property HQ, TDK Corporation, Ichikawa, Chiba 272-8558, Japan.
| | - Tomohiro Noguchi
- Technology and Intellectual Property HQ, TDK Corporation, Ichikawa, Chiba 272-8558, Japan
| | - Takashi Nagano
- Technology and Intellectual Property HQ, TDK Corporation, Ichikawa, Chiba 272-8558, Japan
| | - Wataru Aoki
- Division of Applied Life Sciences, Graduate School of Agriculture, Kyoto University, Sakyo-ku, Kyoto 606-8502, Japan
| | - Mitsuyoshi Ueda
- Division of Applied Life Sciences, Graduate School of Agriculture, Kyoto University, Sakyo-ku, Kyoto 606-8502, Japan
| |
Collapse
|
2
|
Tam NW, Becker A, Mangiarotti A, Cipitria A, Dimova R. Extracellular Vesicle Mobility in Collagen I Hydrogels Is Influenced by Matrix-Binding Integrins. ACS NANO 2024; 18:29585-29601. [PMID: 39400273 PMCID: PMC11526431 DOI: 10.1021/acsnano.4c07186] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Revised: 09/16/2024] [Accepted: 10/02/2024] [Indexed: 10/15/2024]
Abstract
Extracellular vesicles (EVs) are a diverse population of membrane structures produced and released by cells into the extracellular space for the intercellular trafficking of cargo molecules. They are implicated in various biological processes, including angiogenesis, immunomodulation, and cancer cell signaling. While much research has focused on their biogenesis or their effects on recipient cells, less is understood about how EVs are capable of traversing diverse tissue environments and crossing biological barriers. Their interactions with extracellular matrix components are of particular interest, as such interactions govern diffusivity and mobility, providing a potential basis for organotropism. To start to untangle how EV-matrix interactions affect diffusivity, we use high speed epifluorescence microscopy, single particle tracking, and confocal reflectance microscopy to analyze particle mobility and localization in extracellular matrix-mimicking hydrogels composed of collagen I. EVs are compared with synthetic liposomes and extruded plasma membrane vesicles to better understand the importance of membrane composition on these interactions. By treating EVs with trypsin to digest surface proteins, we determine that proteins are primarily responsible for EV immobilization in collagen I hydrogels. We next use a synthetic peptide competitive inhibitor to narrow down the identity of the proteins involved to argynylglycylaspartic acid (RGD) motif-binding integrins, which interact with unincorporated or denatured nonfibrillar collagen. Moreover, the effect of integrin inhibition with RGD peptides has strong implications for the use of RGD-peptide-based drugs to treat certain cancers, as integrin inhibition appears to increase EV mobility, improving their ability to infiltrate tissue-like environments.
Collapse
Affiliation(s)
- Nicky W. Tam
- Max
Planck Institute of Colloids and Interfaces, Science Park Golm, Potsdam 14476, Germany
| | | | - Agustín Mangiarotti
- Max
Planck Institute of Colloids and Interfaces, Science Park Golm, Potsdam 14476, Germany
| | - Amaia Cipitria
- Max
Planck Institute of Colloids and Interfaces, Science Park Golm, Potsdam 14476, Germany
- Group
of Bioengineering in Regeneration and Cancer, Biogipuzkoa Health Research
Institute, San Sebastián 20014, Spain
- IKERBASQUE,
Basque Foundation for Science, Bilbao 48009, Spain
| | - Rumiana Dimova
- Max
Planck Institute of Colloids and Interfaces, Science Park Golm, Potsdam 14476, Germany
| |
Collapse
|
3
|
Osmanagaoglu FH, Ekmekcioglu A, Ozcan B, Bayram Akcapinar G, Muftuoglu M. Preparation and Characterization of Hydrophobin 4-Coated Liposomes for Doxorubicin Delivery to Cancer Cells. Pharmaceuticals (Basel) 2024; 17:1422. [PMID: 39598333 PMCID: PMC11597365 DOI: 10.3390/ph17111422] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2024] [Revised: 10/18/2024] [Accepted: 10/22/2024] [Indexed: 11/29/2024] Open
Abstract
Background: The properties of nanoparticle surfaces are crucial in influencing their interaction with biological environments, as well as their stability, biocompatibility, targeting abilities, and cellular uptake. Hydrophobin 4 (HFB4) is a class II HFB protein produced by filamentous fungi that has a natural ability to self-assemble at hydrophobic-hydrophilic interfaces. The biocompatible, non-toxic, biodegradable, and amphipathic properties of HFB4 render it valuable for improving the solubility and bioavailability of hydrophobic drugs. We have investigated the physicochemical properties, cellular uptake, and anticancer effects of empty and Doxorubicin (Dox)-loaded HFB4 liposomes (HFB4L) and compared them to those of PEGylated liposomes (PPL). Methods: The Pichia pastoris KM71H strain was used for HFB4 purification. Liposomes were prepared through the thin film hydration method and characterized. The cytotoxic effects of free Dox, Dox-HFB4, and Dox-PPL were assessed in MCF7 cells using the SRB Assay. Results: All formulations showed good size homogeneity and a spherical shape. The HFB4 coating enhanced the physicochemical stability of Dox-HFB4L over 60 days at 4 °C without significantly affecting Dox release from HFB4L. It increased Dox release at pH 5.4 compared to pH 7.4, indicating higher delivery of drugs into acidic tumor environments, similar to Dox-PPL. While both formulations showed increased cellular uptake compared to free Dox, they exhibited a lower anticancer effect due to the sustained release of Dox. Notably, Dox-HFB4L displayed greater cytotoxicity than Dox-PPL in MCF7 cells. Conclusions: HFB4L may offer superior benefits in terms of delivering drugs to an acidic tumor environment in a stable, non-toxic, and sustained manner.
Collapse
Affiliation(s)
- Fatma Hande Osmanagaoglu
- Institute of Health Sciences, Department of Medical Biotechnology, Acibadem Mehmet Ali Aydinlar University, 34752 Istanbul, Turkey; (F.H.O.); (A.E.); (G.B.A.)
| | - Aysegul Ekmekcioglu
- Institute of Health Sciences, Department of Medical Biotechnology, Acibadem Mehmet Ali Aydinlar University, 34752 Istanbul, Turkey; (F.H.O.); (A.E.); (G.B.A.)
| | - Busel Ozcan
- Institute of Health Sciences, Department of Medical Biotechnology, Acibadem Mehmet Ali Aydinlar University, 34752 Istanbul, Turkey; (F.H.O.); (A.E.); (G.B.A.)
| | - Gunseli Bayram Akcapinar
- Institute of Health Sciences, Department of Medical Biotechnology, Acibadem Mehmet Ali Aydinlar University, 34752 Istanbul, Turkey; (F.H.O.); (A.E.); (G.B.A.)
| | - Meltem Muftuoglu
- Institute of Health Sciences, Department of Medical Biotechnology, Acibadem Mehmet Ali Aydinlar University, 34752 Istanbul, Turkey; (F.H.O.); (A.E.); (G.B.A.)
- Faculty of Engineering and Natural Sciences, Department of Molecular Biology and Genetics, Acibadem Mehmet Ali Aydinlar University, 34752 Istanbul, Turkey
| |
Collapse
|
4
|
Subramaniam S, Joyce P, Conn CE, Prestidge CA. Cellular uptake and in vitro antibacterial activity of lipid-based nanoantibiotics are influenced by protein corona. Biomater Sci 2024; 12:3411-3422. [PMID: 38809118 DOI: 10.1039/d4bm00608a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/30/2024]
Abstract
Bacteria have evolved survival mechanisms that enable them to live within host cells, triggering persistent intracellular infections that present significant clinical challenges due to the inability for conventional antibiotics to permeate cell membranes. In recent years, antibiotic nanocarriers or 'nanoantibiotics' have presented a promising strategy for overcoming intracellular infections by facilitating cellular uptake of antibiotics, thus improving targeting to the bacteria. However, prior to reaching host cells, nanocarriers experience interactions with proteins that form a corona and alter their physiological response. The influence of this protein corona on the cellular uptake, drug release and efficacy of nanoantibiotics for intracellular infections is poorly understood and commonly overlooked in preclinical studies. In this study, protein corona influence on cellular uptake was investigated for two nanoparticles; liposomes and cubosomes in macrophage and epithelial cells that are commonly infected with pathogens. Studies were conducted in presence of fetal bovine serum (FBS) to form a biologically relevant protein corona in an in vitro setting. Protein corona impact on cellular uptake was shown to be nanoparticle-dependent, where reduced internalization was observed for liposomes, the opposite was observed for cubosomes. Subsequently, vancomycin-loaded cubosomes were explored for their drug delivery performance against intracellular small colony variants of Staphylococcus aureus. We demonstrated improved bacterial killing in macrophages, with greater reduction in bacterial viability upon internalization of cubosomes mediated by the protein corona. However, no differences in efficacy were observed in epithelial cells. Thus, this study provides insights and evidence to the role of protein corona in modulating the performance of nanoparticles in a dynamic manner; these findings will facilitate improved understanding and translation of future investigations from in vitro to in vivo.
Collapse
Affiliation(s)
- Santhni Subramaniam
- University of South Australia, UniSA Clinical and Health Sciences, Adelaide 5000, South Australia, Australia.
| | - Paul Joyce
- University of South Australia, UniSA Clinical and Health Sciences, Adelaide 5000, South Australia, Australia.
| | - Charlotte E Conn
- School of Science, College of Science, Engineering and Health, RMIT University, Melbourne 3000, Victoria, Australia
| | - Clive A Prestidge
- University of South Australia, UniSA Clinical and Health Sciences, Adelaide 5000, South Australia, Australia.
| |
Collapse
|
5
|
Flint Z, Grannemann H, Baffour K, Koti N, Taylor E, Grier E, Sutton C, Johnson D, Dandawate P, Patel R, Santra S, Banerjee T. Mechanistic Insights Behind the Self-Assembly of Human Insulin under the Influence of Surface-Engineered Gold Nanoparticles. ACS Chem Neurosci 2024; 15:2359-2371. [PMID: 38728258 PMCID: PMC11157486 DOI: 10.1021/acschemneuro.4c00226] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2024] [Revised: 04/30/2024] [Accepted: 05/01/2024] [Indexed: 05/12/2024] Open
Abstract
Elucidating the underlying principles of amyloid protein self-assembly at nanobio interfaces is extremely challenging due to the diversity in physicochemical properties of nanomaterials and their physical interactions with biological systems. It is, therefore, important to develop nanoscale materials with dynamic features and heterogeneities. In this work, through engineering of hierarchical polyethylene glycol (PEG) structures on gold nanoparticle (GNP) surfaces, tailored nanomaterials with different surface properties and conformations (GNPs-PEG) are created for modulating the self-assembly of a widely studied protein, insulin, under amyloidogenic conditions. Important biophysical studies including thioflavin T (ThT) binding, circular dichroism (CD), surface plasmon resonance (SPR), and atomic force microscopy (AFM) showed that higher-molecular weight GNPs-PEG triggered the formation of amyloid fibrils by promoting adsorption of proteins at nanoparticle surfaces and favoring primary nucleation rate. Moreover, the modulation of fibrillation kinetics reduces the overall toxicity of insulin oligomers and fibrils. In addition, the interaction between the PEG polymer and amyloidogenic insulin examined using MD simulations revealed major changes in the secondary structural elements of the B chain of insulin. The experimental findings provide molecular-level descriptions of how the PEGylated nanoparticle surface modulates protein adsorption and drives the self-assembly of insulin. This facile approach provides a new avenue for systematically altering the binding affinities on nanoscale surfaces by tailoring their topologies for examining adsorption-induced fibrillogenesis phenomena of amyloid proteins. Together, this study suggests the role of nanobio interfaces during surface-induced heterogeneous nucleation as a primary target for designing therapeutic interventions for amyloid-related neurodegenerative disorders.
Collapse
Affiliation(s)
- Zachary Flint
- Department
of Chemistry and Biochemistry, Missouri
State University, 901 S. National Avenue, Springfield, Missouri 65897, United States
| | - Haylee Grannemann
- Department
of Chemistry and Biochemistry, Missouri
State University, 901 S. National Avenue, Springfield, Missouri 65897, United States
| | - Kristos Baffour
- Department
of Chemistry and Biochemistry, Missouri
State University, 901 S. National Avenue, Springfield, Missouri 65897, United States
| | - Neelima Koti
- Department
of Chemistry and Biochemistry, Missouri
State University, 901 S. National Avenue, Springfield, Missouri 65897, United States
| | - Emma Taylor
- Department
of Chemistry and Biochemistry, Missouri
State University, 901 S. National Avenue, Springfield, Missouri 65897, United States
| | - Ethan Grier
- Department
of Chemistry and Biochemistry, Missouri
State University, 901 S. National Avenue, Springfield, Missouri 65897, United States
| | - Carissa Sutton
- Department
of Chemistry and Biochemistry, Missouri
State University, 901 S. National Avenue, Springfield, Missouri 65897, United States
| | - David Johnson
- Molecular
Graphics and Modeling Laboratory, University
of Kansas, 2034 Becker
Drive, Lawrence, Kansas 66018, United States
| | - Prasad Dandawate
- Department
of Cancer Biology, The University of Kansas
Medical Center, Kansas City, Kansas 66160, United States
| | - Rishi Patel
- Jordan
Valley Innovation Center, Missouri State
University, 542 N. Boonville
Avenue, Springfield, Missouri 65806, United States
| | - Santimukul Santra
- Department
of Chemistry and Biochemistry, Missouri
State University, 901 S. National Avenue, Springfield, Missouri 65897, United States
| | - Tuhina Banerjee
- Department
of Chemistry and Biochemistry, Missouri
State University, 901 S. National Avenue, Springfield, Missouri 65897, United States
| |
Collapse
|
6
|
Park HW, Lee W, Kim S, Jangid AK, Park J, Lee CE, Kim K. Optimized Design of Hyaluronic Acid-Lipid Conjugate Biomaterial for Augmenting CD44 Recognition of Surface-Engineered NK Cells. Biomacromolecules 2024; 25:1959-1971. [PMID: 38379131 DOI: 10.1021/acs.biomac.3c01373] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/22/2024]
Abstract
Triple-negative breast cancer (TNBC) presents treatment challenges due to a lack of detectable surface receptors. Natural killer (NK) cell-based adaptive immunotherapy is a promising treatment because of the characteristic anticancer effects of killing malignant cells directly by secreting cytokines and lytic granules. To maximize the cancer recognition ability of NK cells, biomaterial-mediated ex vivo cell surface engineering has been developed for sufficient cell membrane immobilization of tumor-targeting ligands via hydrophobic anchoring. In this study, we optimized amphiphilic balances of NK cell coating materials composed of CD44-targeting hyaluronic acid (HA)-poly(ethylene glycol) (PEG)-lipid to improve TNBC recognition and the anticancer effect. Changes in the modular design of our material by differentiating hydrophilic PEG length and incorporating lipid amount into HA backbones precisely regulated the amphiphilic nature of HA-PEG-lipid conjugates. The optimized biomaterial demonstrated improved anchoring into NK cell membranes and facilitating the surface presentation level of HA onto NK cell surfaces. This led to enhanced cancer targeting via increasing the formation of immune synapse, thereby augmenting the anticancer capability of NK cells specifically toward CD44-positive TNBC cells. Our approach addresses targeting ability of NK cell to solid tumors with a deficiency of surface tumor-specific antigens while offering a valuable material design strategy using amphiphilic balance in immune cell surface engineering techniques.
Collapse
Affiliation(s)
- Hee Won Park
- Department of Chemical & Biochemical Engineering, Dongguk University, Seoul 04620, Republic of Korea
| | - Wonjeong Lee
- Department of Chemical & Biochemical Engineering, Dongguk University, Seoul 04620, Republic of Korea
| | - Sungjun Kim
- Department of Chemical & Biochemical Engineering, Dongguk University, Seoul 04620, Republic of Korea
| | - Ashok Kumar Jangid
- Department of Chemical & Biochemical Engineering, Dongguk University, Seoul 04620, Republic of Korea
| | - Jaewon Park
- Department of Chemical & Biochemical Engineering, Dongguk University, Seoul 04620, Republic of Korea
| | - Chae Eun Lee
- Department of Chemical & Biochemical Engineering, Dongguk University, Seoul 04620, Republic of Korea
| | - Kyobum Kim
- Department of Chemical & Biochemical Engineering, Dongguk University, Seoul 04620, Republic of Korea
| |
Collapse
|
7
|
Tam NW, Schullian O, Cipitria A, Dimova R. Nonspecific membrane-matrix interactions influence diffusivity of lipid vesicles in hydrogels. Biophys J 2024; 123:638-650. [PMID: 38332584 PMCID: PMC10938137 DOI: 10.1016/j.bpj.2024.02.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2023] [Revised: 12/13/2023] [Accepted: 02/05/2024] [Indexed: 02/10/2024] Open
Abstract
The diffusion of extracellular vesicles and liposomes in vivo is affected by different tissue environmental conditions and is of great interest in the development of liposome-based therapeutics and drug-delivery systems. Here, we use a bottom-up biomimetic approach to better isolate and study steric and electrostatic interactions and their influence on the diffusivity of synthetic large unilamellar vesicles in hydrogel environments. Single-particle tracking of these extracellular vesicle-like particles in agarose hydrogels as an extracellular matrix model shows that membrane deformability and surface charge affect the hydrogel pore spaces that vesicles have access to, which determines overall diffusivity. Moreover, we show that passivation of vesicles with PEGylated lipids, as often used in drug-delivery systems, enhances diffusivity, but that this effect cannot be fully explained with electrostatic interactions alone. Finally, we compare our experimental findings with existing computational and theoretical work in the field to help explain the nonspecific interactions between diffusing particles and gel matrix environments.
Collapse
Affiliation(s)
- Nicky W Tam
- Max Planck Institute of Colloids and Interfaces, Science Park Golm, Potsdam, Germany
| | - Otto Schullian
- Max Planck Institute of Colloids and Interfaces, Science Park Golm, Potsdam, Germany; Free University of Berlin, Department of Physics, Berlin, Germany
| | - Amaia Cipitria
- Max Planck Institute of Colloids and Interfaces, Science Park Golm, Potsdam, Germany; Group of Bioengineering in Regeneration and Cancer, Biogipuzkoa Health Research Institute, San Sebastián, Spain; IKERBASQUE, Basque Foundation for Science, Bilbao, Spain
| | - Rumiana Dimova
- Max Planck Institute of Colloids and Interfaces, Science Park Golm, Potsdam, Germany.
| |
Collapse
|
8
|
Lustig DR, Buz E, Mulvey JT, Patterson JP, Kittilstved KR, Sambur JB. Characterizing the Ligand Shell Morphology of PEG-Coated ZnO Nanocrystals Using FRET Spectroscopy. J Phys Chem B 2023; 127:8961-8973. [PMID: 37802098 DOI: 10.1021/acs.jpcb.3c04900] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/08/2023]
Abstract
Poly(ethylene glycol) (PEG) ligands can inhibit proteins and other biomolecules from adhering to underlying surfaces, making them excellent surface ligands for nanocrystal (NC)-based drug carriers. Quantifying the PEG ligand shell morphology is important because its structure determines the permeability of biomolecules through the shell to the NC surface. However, few in situ analytical tools can reveal whether the PEG ligands form either an impenetrable barrier or a porous coating surrounding the NC. Here, we present a Förster resonance energy transfer (FRET) spectroscopy-based approach that can assess the permeability of molecules through PEG-coated ZnO NCs. In this approach, ZnO NCs serve as FRET donors, and freely diffusing molecules in the bulk solution are FRET acceptors. We synthesized a series of variable chain length PEG-silane-coated ZnO NCs such that the longest chain length ligands far exceed the Förster radius (R0), where the energy transfer (EnT) efficiency is 50%. We quantified the EnT efficiency as a function of the ligand chain length using time-resolved photoluminescence lifetime (TRPL) spectroscopy within the framework of FRET theory. Unexpectedly, the longest PEG-silane ligand showed equivalent EnT efficiency as that of bare, hydroxyl-passivated ZnO NCs. These results indicate that the "rigid shell" model fails and the PEG ligand shell morphology is more likely porous or in a patchy "mushroom state", consistent with transmission electron microscopy data. While the spectroscopic measurements and data analysis procedures discussed herein cannot directly visualize the ligand shell morphology in real space, the in situ spectroscopy approach can provide researchers with valuable information regarding the permeability of species through the ligand shell under practical biological conditions.
Collapse
Affiliation(s)
- Danielle R Lustig
- Department of Chemistry, Colorado State University, 200 West Lake Street, Fort Collins, Colorado 80523-1872, United States
| | - Enes Buz
- Department of Chemistry, University of Massachusetts Amherst, 710 North Pleasant Street, Amherst, Massachusetts 01003, United States
| | - Justin T Mulvey
- Center for Complex and Active Materials, University of California, Irvine, Irvine, California 92697-2025, United States
- Department of Materials Science and Engineering, University of California, Irvine, Irvine, California 92697-2025, United States
| | - Joseph P Patterson
- Center for Complex and Active Materials, University of California, Irvine, Irvine, California 92697-2025, United States
- Department of Chemistry, University of California, Irvine, Irvine, California 92697-2025, United States
- Department of Materials Science and Engineering, University of California, Irvine, Irvine, California 92697-2025, United States
| | - Kevin R Kittilstved
- Department of Chemistry, University of Massachusetts Amherst, 710 North Pleasant Street, Amherst, Massachusetts 01003, United States
| | - Justin B Sambur
- Department of Chemistry, Colorado State University, 200 West Lake Street, Fort Collins, Colorado 80523-1872, United States
| |
Collapse
|
9
|
Yin B, Zhang Q, Yan J, Huang Y, Li C, Chen J, Wen C, Wong SHD, Yang M. Nanomanipulation of Ligand Nanogeometry Modulates Integrin/Clathrin-Mediated Adhesion and Endocytosis of Stem Cells. NANO LETTERS 2023; 23:9160-9169. [PMID: 37494286 DOI: 10.1021/acs.nanolett.3c01757] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/28/2023]
Abstract
Nanosubstrate engineering can be a biomechanical approach for modulating stem cell differentiation in tissue engineering. However, the study of the effect of clathrin-mediated processes on manipulating this behavior is unexplored. Herein, we develop integrin-binding nanosubstrates with confined nanogeometries that regulate clathrin-mediated adhesion- or endocytosis-active signaling pathways for modulating stem fates. Isotropically presenting ligands on the nanoscale enhances the expression of clathrin in cells, thereby facilitating uptake of dexamethasone-loaded nanoparticles (NPs) to boost osteogenesis of stem cells. In contrast, anisotropic ligand nanogeometry suppresses this clathrin-mediated NP entry by strengthening the association between clathrin and adhesion spots to reinforce mechanotransduced signaling, which can be abrogated by the pharmacological inhibition of clathrin. Meanwhile, inhibiting focal adhesion formation hinders cell spreading and enables a higher endocytosis efficiency. Our findings reveal the crucial roles of clathrin in both endocytosis and mechanotransduction of stem cells and provide the parameter of ligand nanogeometry for the rational design of biomaterials for tissue engineering.
Collapse
Affiliation(s)
- Bohan Yin
- Department of Biomedical Engineering, The Hong Kong Polytechnic University, Kowloon, Hong Kong 999077, China
| | - Qin Zhang
- Department of Biomedical Engineering, The Hong Kong Polytechnic University, Kowloon, Hong Kong 999077, China
| | - Jiaxiang Yan
- Department of Biomedical Engineering, The Hong Kong Polytechnic University, Kowloon, Hong Kong 999077, China
| | - Yingying Huang
- Department of Biomedical Engineering, The Hong Kong Polytechnic University, Kowloon, Hong Kong 999077, China
| | - Chuanqi Li
- Department of Biomedical Engineering, The Hong Kong Polytechnic University, Kowloon, Hong Kong 999077, China
| | - Jiareng Chen
- Department of Biomedical Engineering, The Hong Kong Polytechnic University, Kowloon, Hong Kong 999077, China
| | - Chunyi Wen
- Department of Biomedical Engineering, The Hong Kong Polytechnic University, Kowloon, Hong Kong 999077, China
| | - Siu Hong Dexter Wong
- Department of Biomedical Engineering, The Hong Kong Polytechnic University, Kowloon, Hong Kong 999077, China
- Research Institute for Sports Science and Technology, The Hong Kong Polytechnic University, Kowloon, Hong Kong 999077, China
| | - Mo Yang
- Department of Biomedical Engineering, The Hong Kong Polytechnic University, Kowloon, Hong Kong 999077, China
- Research Institute for Sports Science and Technology, The Hong Kong Polytechnic University, Kowloon, Hong Kong 999077, China
| |
Collapse
|
10
|
Takatori SC, Son S, Lee DSW, Fletcher DA. Engineered molecular sensors for quantifying cell surface crowding. Proc Natl Acad Sci U S A 2023; 120:e2219778120. [PMID: 37186825 PMCID: PMC10214205 DOI: 10.1073/pnas.2219778120] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2022] [Accepted: 04/13/2023] [Indexed: 05/17/2023] Open
Abstract
Cells mediate interactions with the extracellular environment through a crowded assembly of transmembrane proteins, glycoproteins and glycolipids on their plasma membrane. The extent to which surface crowding modulates the biophysical interactions of ligands, receptors, and other macromolecules is poorly understood due to the lack of methods to quantify surface crowding on native cell membranes. In this work, we demonstrate that physical crowding on reconstituted membranes and live cell surfaces attenuates the effective binding affinity of macromolecules such as IgG antibodies in a surface crowding-dependent manner. We combine experiment and simulation to design a crowding sensor based on this principle that provides a quantitative readout of cell surface crowding. Our measurements reveal that surface crowding decreases IgG antibody binding by 2 to 20 fold in live cells compared to a bare membrane surface. Our sensors show that sialic acid, a negatively charged monosaccharide, contributes disproportionately to red blood cell surface crowding via electrostatic repulsion, despite occupying only ~1% of the total cell membrane by mass. We also observe significant differences in surface crowding for different cell types and find that expression of single oncogenes can both increase and decrease crowding, suggesting that surface crowding may be an indicator of both cell type and state. Our high-throughput, single-cell measurement of cell surface crowding may be combined with functional assays to enable further biophysical dissection of the cell surfaceome.
Collapse
Affiliation(s)
- Sho C. Takatori
- Department of Bioengineering, University of California, Berkeley, CA94720
- Department of Chemical Engineering, University of California, Santa Barbara, CA93106
| | - Sungmin Son
- Department of Bioengineering, University of California, Berkeley, CA94720
- Department of Bio and Brain Engineering, Korea Advanced Institute of Science and Technology, Daejeon, Republic of Korea
| | - Daniel S. W. Lee
- Department of Bioengineering, University of California, Berkeley, CA94720
| | - Daniel A. Fletcher
- Department of Bioengineering, University of California, Berkeley, CA94720
- University of California, Berkeley/University of California, San Francisco Graduate Group in Bioengineering, Berkeley, CA94720
- Division of Biological Systems and Engineering, Lawrence Berkeley National Laboratory, Berkeley, CA94720
- Chan Zuckerberg Biohub, San Francisco, CA94158
| |
Collapse
|
11
|
Fulton MD, Najahi-Missaoui W. Liposomes in Cancer Therapy: How Did We Start and Where Are We Now. Int J Mol Sci 2023; 24:ijms24076615. [PMID: 37047585 PMCID: PMC10095497 DOI: 10.3390/ijms24076615] [Citation(s) in RCA: 70] [Impact Index Per Article: 35.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 03/28/2023] [Accepted: 03/29/2023] [Indexed: 04/05/2023] Open
Abstract
Since their first discovery in the 1960s by Alec Bangham, liposomes have been shown to be effective drug delivery systems for treating various cancers. Several liposome-based formulations received approval by the U.S. Food and Drug Administration (FDA) and European Medicines Agency (EMA), with many others in clinical trials. Liposomes have several advantages, including improved pharmacokinetic properties of the encapsulated drug, reduced systemic toxicity, extended circulation time, and targeted disposition in tumor sites due to the enhanced permeability and retention (EPR) mechanism. However, it is worth noting that despite their efficacy in treating various cancers, liposomes still have some potential toxicity and lack specific targeting and disposition. This explains, in part, why their translation into the clinic has progressed only incrementally, which poses the need for more research to focus on addressing such translational limitations. This review summarizes the main properties of liposomes, their current status in cancer therapy, and their limitations and challenges to achieving maximal therapeutic efficacy.
Collapse
Affiliation(s)
- Melody D. Fulton
- Department of Chemistry, College of Arts and Sciences, Washington State University, Pullman, WA 99164, USA
| | - Wided Najahi-Missaoui
- Department of Pharmaceutical and Biomedical Sciences, College of Pharmacy, University of Georgia, Athens, GA 30602, USA
| |
Collapse
|
12
|
Current understanding of passive and active targeting nanomedicines to enhance tumor accumulation. Coord Chem Rev 2023. [DOI: 10.1016/j.ccr.2023.215051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
|
13
|
Kosaka T, Yamaguchi S, Izuta S, Yamahira S, Shibasaki Y, Tateno H, Okamoto A. Bioorthogonal Photoreactive Surfaces for Single-Cell Analysis of Intercellular Communications. J Am Chem Soc 2022; 144:17980-17988. [PMID: 36126284 DOI: 10.1021/jacs.2c07321] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Methods to construct single-cell pairs of heterogeneous cells attract attention because of their potential in cell biological and medical applications for analyzing individual intercellular communications such as immune and nerve synaptic interactions. Photoactivatable substrate surfaces for cell anchoring are promising tools to achieve single-cell pairing. However, conventional surfaces that photoactivate a single type of cell anchoring moiety restrict the combination of cell pair types and their applications. We developed a photoresponsive material comprising a bioorthogonal photoreactive moiety and non-cell adhesive hydrophilic polymer. The material-coated surface allows conjugation with various cell anchoring molecules in response to light at specific timings and consequently achieves light-induced anchoring of a variety of cells at defined regions. Using the platform surface, an array of cancer cell and natural-killer (NK) cell pairs was constructed on a flat substrate surface and the dynamic morphological changes of the cancer cells were monitored by cytotoxic interaction with NK cells at a single-cell level. The photoreactive surface is a useful tool for image-based investigation of the communications between a variety of cell types.
Collapse
Affiliation(s)
- Takahiro Kosaka
- Department of Chemistry & Biotechnology, Graduate School of Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8656, Japan
| | - Satoshi Yamaguchi
- Department of Chemistry & Biotechnology, Graduate School of Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8656, Japan.,PRESTO, Japan Science and Technology Agency (JST), 7, Gobancho, Chiyoda-ku, Tokyo 102-0076, Japan
| | - Shin Izuta
- Department of Chemistry & Biotechnology, Graduate School of Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8656, Japan
| | - Shinya Yamahira
- Center for Medical Sciences, St. Luke's International University, 9-1 Akashi-cho, Chuo-ku, Tokyo 104-8560, Japan
| | - Yoshikazu Shibasaki
- Research Center for Advanced Science and Technology (RCAST), The University of Tokyo, 4-6-1 Komaba, Meguro-ku, Tokyo 153-8904, Japan
| | - Hiroaki Tateno
- Cellular and Molecular Biotechnology Research Institute, National Institute of Advanced Industrial Science and Technology (AIST), 1-1-1 Higashi, Tsukuba, Ibaraki 305-8566, Japan
| | - Akimitsu Okamoto
- Department of Chemistry & Biotechnology, Graduate School of Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8656, Japan
| |
Collapse
|
14
|
Meewan J, Somani S, Almowalad J, Laskar P, Mullin M, MacKenzie G, Khadke S, Perrie Y, Dufès C. Preparation of Zein-Based Nanoparticles: Nanoprecipitation versus Microfluidic-Assisted Manufacture, Effects of PEGylation on Nanoparticle Characteristics and Cellular Uptake by Melanoma Cells. Int J Nanomedicine 2022; 17:2809-2822. [PMID: 35791309 PMCID: PMC9250780 DOI: 10.2147/ijn.s366138] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Accepted: 06/15/2022] [Indexed: 12/02/2022] Open
Abstract
Background The manufacture of nanoparticles using manual methods is hampered by its challenging scale-up and poor reproducibility. To overcome this issue, the production of zein nanoparticles entrapping a lipophilic drug model, coumarin-6, by using a microfluidic system was assessed in this study. The influence of PEG density and chain length on zein nanoparticle characteristics, as well as their uptake efficacy in melanoma cancer cells, was also evaluated. Methods Zein nanoparticles were prepared by both manual and microfluidic approaches to allow comparison between the two processes. PEGylated zein nanoparticles with various PEG densities and chain lengths were produced by nanoprecipitation and characterized. Their cellular uptake was evaluated on B16F10 melanoma cancer cells in vitro. Results Zein nanoparticles have successfully been produced by both manual and microfluidic approaches. Parameters such as total flow rate and flow rate ratio of the aqueous and organic phases in microfluidic process, as well as the method preparation and aqueous to organic phase volume ratio during nanoprecipitation, have been shown to strongly influence the characteristics of the resulting nanoparticles. Continuous microfluidics led to the production of nanoparticles with low yield and drug entrapment, unlike nanoprecipitation, which resulted in zein nanoparticles with an appropriate size and an optimal drug entrapment efficiency of 64%. The surface modification of the nanoparticles produced by nanoprecipitation, with lower PEG density and shorter PEG chain length made mPEG5K-zein (0.5:1) the most favorable formulation in our study, resulting in enhanced stability and higher coumarin-6 uptake by melanoma cancer cells. Conclusion mPEG5K-zein (0.5:1) nanoparticles prepared by nanoprecipitation were the most promising formulation in our study, exhibiting increased stability and enhancing coumarin-6 uptake by melanoma cancer cells.
Collapse
Affiliation(s)
- Jitkasem Meewan
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, G4 0RE, UK
| | - Sukrut Somani
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, G4 0RE, UK
| | - Jamal Almowalad
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, G4 0RE, UK
| | - Partha Laskar
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, G4 0RE, UK
| | - Margaret Mullin
- Glasgow Imaging Facility, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, G12 8QQ, UK
| | - Graeme MacKenzie
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, G4 0RE, UK
| | - Swapnil Khadke
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, G4 0RE, UK
| | - Yvonne Perrie
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, G4 0RE, UK
| | - Christine Dufès
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, G4 0RE, UK
| |
Collapse
|
15
|
Triantafyllopoulou E, Pippa N, Demetzos C. Protein-liposome interactions: the impact of surface charge and fluidisation effect on protein binding. J Liposome Res 2022; 33:77-88. [PMID: 35730463 DOI: 10.1080/08982104.2022.2071296] [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: 10/17/2022]
Abstract
At the dawn of a new nanotechnological era in the pharmaceutical field, it is very important to examine and understand all the aspects that influence in vivo behaviour of nanoparticles. In this point of view, the interactions between serum proteins and liposomes with incorporated anionic, cationic, and/or PEGylated lipids were investigated to elucidate the role of surface charge and bilayer fluidity in protein corona's formation. 1,2-dipalmitoyl-sn-glycero-3- phosphocholine (DPPC), hydrogenated soybean phosphatidylcholine (HSPC), and 1,2-dioctadecanoyl-sn-glycero-3-phosphocholine (DSPC) liposomes with the presence or absence of 1,2-dipalmitoyl-sn-glycero-3-phospho-(1'-rac-glycerol) (sodium salt) (DPPG), 1,2-di-(9Z-octadecenoyl)-3-trimethylammonium-propane (chloride salt) (DOTAP), and/or 1,2-dipalmitoylsn-glycero-3-phosphoethanolamine-N-[methoxy(polyethylene glycol)-5000] (DPPE-PEG 5000) lipids were prepared by the thin-film hydration method. The evaluation of their biophysical characteristics was enabled by differential scanning calorimetry and dynamic and electrophoretic light scattering. The physicochemical characteristics of mixed liposomes were compared before and after exposure to foetal bovine serum (FBS) and were correlated to calorimetric data. Our results indicate protein binding to all liposomal formulations. However, it is highlighted the importance of surface charge and fluidisation effect to the extent of protein adsorption. Additionally, considering the extensive use of cationic lipids for innovative delivery platforms, we deem PEGylation a key parameter, because even in a small proportion can reduce protein binding, and thus fast clearance and extreme toxicity without affecting positive charge. This study is a continuation of our previous work about protein-liposome interactions and fraction of stealthiness (Fs) parameter, and hopefully a design road map for drug and gene delivery.
Collapse
Affiliation(s)
- Efstathia Triantafyllopoulou
- Section of Pharmaceutical Technology, Department of Pharmacy, School of Health Sciences, National and Kapodistrian University of Athens, Athens, Greece
| | - Natassa Pippa
- Section of Pharmaceutical Technology, Department of Pharmacy, School of Health Sciences, National and Kapodistrian University of Athens, Athens, Greece
| | - Costas Demetzos
- Section of Pharmaceutical Technology, Department of Pharmacy, School of Health Sciences, National and Kapodistrian University of Athens, Athens, Greece
| |
Collapse
|
16
|
Li B, Çolak A, Blass J, Han M, Zhang J, Zheng Y, Jiang Q, Bennewitz R, Campo AD. Molecular stiffness cues of an interpenetrating network hydrogel for cell adhesion. Mater Today Bio 2022; 15:100323. [PMID: 35782598 PMCID: PMC9243155 DOI: 10.1016/j.mtbio.2022.100323] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2022] [Revised: 05/24/2022] [Accepted: 06/09/2022] [Indexed: 11/11/2022] Open
Abstract
Understanding cells' response to the macroscopic and nanoscale properties of biomaterials requires studies in model systems with the possibility to tailor their mechanical properties and different length scales. Here, we describe an interpenetrating network (IPN) design based on a stiff PEGDA host network interlaced within a soft 4-arm PEG-Maleimide/thiol (guest) network. We quantify the nano- and bulk mechanical behavior of the IPN and the single network hydrogels by single-molecule force spectroscopy and rheological measurements. The IPN presents different mechanical cues at the molecular scale, depending on which network is linked to the probe, but the same mechanical properties at the macroscopic length scale as the individual host network. Cells attached to the interpenetrating (guest) network of the IPN or to the single network (SN) PEGDA hydrogel modified with RGD adhesive ligands showed comparable attachment and spreading areas, but cells attached to the guest network of the IPN, with lower molecular stiffness, showed a larger number and size of focal adhesion complexes and a higher concentration of the Hippo pathway effector Yes-associated protein (YAP) than cells linked to the PEGDA single network. The observations indicate that cell adhesion to the IPN hydrogel through the network with lower molecular stiffness proceeds effectively as if a higher ligand density is offered. We claim that IPNs can be used to decipher how changes in ECM design and connectivity at the local scale affect the fate of cells cultured on biomaterials.
Collapse
|
17
|
Meewan J, Somani S, Laskar P, Irving C, Mullin M, Woods S, Roberts CW, Alzahrani AR, Ferro VA, McGill S, Weidt S, Burchmore R, Dufès C. Limited Impact of the Protein Corona on the Cellular Uptake of PEGylated Zein Micelles by Melanoma Cancer Cells. Pharmaceutics 2022; 14:pharmaceutics14020439. [PMID: 35214171 PMCID: PMC8877401 DOI: 10.3390/pharmaceutics14020439] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2022] [Revised: 02/13/2022] [Accepted: 02/14/2022] [Indexed: 12/17/2022] Open
Abstract
The formation of a protein layer “corona” on the nanoparticle surface upon entry into a biological environment was shown to strongly influence the interactions with cells, especially affecting the uptake of nanomedicines. In this work, we present the impact of the protein corona on the uptake of PEGylated zein micelles by cancer cells, macrophages, and dendritic cells. Zein was successfully conjugated with poly(ethylene glycol) (PEG) of varying chain lengths (5K and 10K) and assembled into micelles. Our results demonstrate that PEGylation conferred stealth effects to the zein micelles. The presence of human plasma did not impact the uptake levels of the micelles by melanoma cancer cells, regardless of the PEG chain length used. In contrast, it decreased the uptake by macrophages and dendritic cells. These results therefore make PEGylated zein micelles promising as potential drug delivery systems for cancer therapy.
Collapse
Affiliation(s)
- Jitkasem Meewan
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, 161 Cathedral Street, Glasgow G4 0RE, UK; (J.M.); (S.S.); (P.L.); (S.W.); (C.W.R.); (A.R.A.); (V.A.F.)
| | - Sukrut Somani
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, 161 Cathedral Street, Glasgow G4 0RE, UK; (J.M.); (S.S.); (P.L.); (S.W.); (C.W.R.); (A.R.A.); (V.A.F.)
| | - Partha Laskar
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, 161 Cathedral Street, Glasgow G4 0RE, UK; (J.M.); (S.S.); (P.L.); (S.W.); (C.W.R.); (A.R.A.); (V.A.F.)
- Department of Immunology and Microbiology, University of Texas Health Rio Grande Valley, 5300 North L Street 881 Madison, McAllen, TX 78504, USA
| | - Craig Irving
- Department of Pure and Applied Chemistry, University of Strathclyde, 295 Cathedral Street, Glasgow G1 1XL, UK;
| | - Margaret Mullin
- Glasgow Imaging Facility, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow G12 8QQ, UK;
| | - Stuart Woods
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, 161 Cathedral Street, Glasgow G4 0RE, UK; (J.M.); (S.S.); (P.L.); (S.W.); (C.W.R.); (A.R.A.); (V.A.F.)
| | - Craig W. Roberts
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, 161 Cathedral Street, Glasgow G4 0RE, UK; (J.M.); (S.S.); (P.L.); (S.W.); (C.W.R.); (A.R.A.); (V.A.F.)
| | - Abdullah R. Alzahrani
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, 161 Cathedral Street, Glasgow G4 0RE, UK; (J.M.); (S.S.); (P.L.); (S.W.); (C.W.R.); (A.R.A.); (V.A.F.)
- Department of Pharmacology & Toxicology, Faculty of Medicine, Umm Al-Qura University, Al-Abidiyah, P.O. Box 13578, Makkah 21955, Saudi Arabia
| | - Valerie A. Ferro
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, 161 Cathedral Street, Glasgow G4 0RE, UK; (J.M.); (S.S.); (P.L.); (S.W.); (C.W.R.); (A.R.A.); (V.A.F.)
| | - Suzanne McGill
- Glasgow Polyomics, Wolfson Wohl Cancer Research Centre, Garscube Campus, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow G61 1QH, UK; (S.M.); (S.W.); (R.B.)
| | - Stefan Weidt
- Glasgow Polyomics, Wolfson Wohl Cancer Research Centre, Garscube Campus, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow G61 1QH, UK; (S.M.); (S.W.); (R.B.)
| | - Richard Burchmore
- Glasgow Polyomics, Wolfson Wohl Cancer Research Centre, Garscube Campus, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow G61 1QH, UK; (S.M.); (S.W.); (R.B.)
| | - Christine Dufès
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, 161 Cathedral Street, Glasgow G4 0RE, UK; (J.M.); (S.S.); (P.L.); (S.W.); (C.W.R.); (A.R.A.); (V.A.F.)
- Correspondence: ; Tel.: +44-141-548-3796
| |
Collapse
|
18
|
Nguyen TA, Lindner JR. Contrast-Enhanced Ultrasound Molecular Imaging in Atherosclerosis Research. Methods Mol Biol 2022; 2419:801-808. [PMID: 35238002 DOI: 10.1007/978-1-0716-1924-7_48] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
The management of cardiovascular conditions will likely be improved by noninvasive in vivo molecular imaging technologies that can provide earlier or more accurate diagnosis. These techniques are already having a positive impact in preclinical research by providing insight into disease pathobiology or efficacy of new therapies. Contrast enhanced ultrasound (CEU) molecular imaging is a technique that relies on the ultrasound detection of targeted microbubble contrast agents to examine molecular or cellular events that occur at the blood pool-endothelial interface. For the most part, targeted contrast agents are composed of encapsulated gas microbubbles (MBs) that are 2-4 μm in diameter, or other acoustically active micro- or nanoparticles. These agents bear several tens of thousands of binding molecules per particle. Because nonadhered agent is cleared rapidly, CEU molecular imaging can be performed in a matter of minutes. MBs are detected using contrast-specific techniques that generate and receive nonlinear signals produced by MB cavitation, thereby increasing signal-to-noise ratio. Dedicated kinetic models for molecular imaging have been generated that permit the elimination of signal from nonadherent agent.
Collapse
|
19
|
Hage M, Khelissa S, Akoum H, Chihib NE, Jama C. Cold plasma surface treatments to prevent biofilm formation in food industries and medical sectors. Appl Microbiol Biotechnol 2022; 106:81-100. [PMID: 34889984 PMCID: PMC8661349 DOI: 10.1007/s00253-021-11715-y] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Revised: 11/19/2021] [Accepted: 11/24/2021] [Indexed: 10/28/2022]
Abstract
Environmental conditions in food and medical fields enable the bacteria to attach and grow on surfaces leading to resistant bacterial biofilm formation. Indeed, the first step in biofilm formation is the bacterial irreversible adhesion. Controlling and inhibiting this adhesion is a passive approach to fight against biofilm development. This strategy is an interesting path in the inhibition of biofilm formation since it targets the first step of biofilm development. Those pathogenic structures are responsible for several foodborne diseases and nosocomial infections. Therefore, to face this public health threat, researchers employed cold plasma technologies in coating development. In this review, the different factors influencing the bacterial adhesion to a substrate are outlined. The goal is to present the passive coating strategies aiming to prevent biofilm formation via cold plasma treatments, highlighting antiadhesive elaborated surfaces. General aspects of surface treatment, including physico-chemical modification and application of cold plasma technologies, were also presented. KEY POINTS: • Factors surrounding pathogenic bacteria influence biofilm development. • Controlling bacterial adhesion prevents biofilm formation. • Materials can be coated via cold plasma to inhibit bacterial adhesion.
Collapse
Affiliation(s)
- Mayssane Hage
- UMR 8207 - UMET - Unité Matériaux Et Transformations, Univ. Lille, CNRS, INRAE, Centrale Lille, 59000, Lille, France
- Laboratoire d'analyses Chimiques Et Microbiologiques, Faculté de Santé Publique - Université Libanaise, Saida, Lebanon
| | - Simon Khelissa
- UMR 8207 - UMET - Unité Matériaux Et Transformations, Univ. Lille, CNRS, INRAE, Centrale Lille, 59000, Lille, France
| | - Hikmat Akoum
- Laboratoire d'analyses Chimiques Et Microbiologiques, Faculté de Santé Publique - Université Libanaise, Saida, Lebanon
| | - Nour-Eddine Chihib
- UMR 8207 - UMET - Unité Matériaux Et Transformations, Univ. Lille, CNRS, INRAE, Centrale Lille, 59000, Lille, France
| | - Charafeddine Jama
- UMR 8207 - UMET - Unité Matériaux Et Transformations, Univ. Lille, CNRS, INRAE, Centrale Lille, 59000, Lille, France.
| |
Collapse
|
20
|
Challenges of Current Anticancer Treatment Approaches with Focus on Liposomal Drug Delivery Systems. Pharmaceuticals (Basel) 2021; 14:ph14090835. [PMID: 34577537 PMCID: PMC8466509 DOI: 10.3390/ph14090835] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Revised: 08/13/2021] [Accepted: 08/20/2021] [Indexed: 12/13/2022] Open
Abstract
According to a 2020 World Health Organization report (Globocan 2020), cancer was a leading cause of death worldwide, accounting for nearly 10 million deaths in 2020. The aim of anticancer therapy is to specifically inhibit the growth of cancer cells while sparing normal dividing cells. Conventional chemotherapy, radiotherapy and surgical treatments have often been plagued by the frequency and severity of side effects as well as severe patient discomfort. Cancer targeting by drug delivery systems, owing to their selective targeting, efficacy, biocompatibility and high drug payload, provides an attractive alternative treatment; however, there are technical, therapeutic, manufacturing and clinical barriers that limit their use. This article provides a brief review of the challenges of conventional anticancer therapies and anticancer drug targeting with a special focus on liposomal drug delivery systems.
Collapse
|
21
|
Aghajani M, Esmaeili F. Anti-biofouling assembly strategies for protein & cell repellent surfaces: a mini-review. JOURNAL OF BIOMATERIALS SCIENCE-POLYMER EDITION 2021; 32:1770-1789. [PMID: 34085909 DOI: 10.1080/09205063.2021.1932357] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
The protein/cell interactions with the surface at the blood-biomaterial interface generally control the efficiency of biomedical devices. A wide range of active processes and slow kinetics occur simultaneously with many biomaterials in healthcare applications, leading to multiple biological reactions and reduced clinical functions. In this work, we present a brief review of studies as the interface between proteins and biomaterials. These include mechanisms of resistance to proteins, protein-rejecting polyelectrolyte multilayers, and coatings of hydrophilic, polysaccharide and phospholipid nature. The mechanisms required to attain surfaces that resist adhesion include steric exclusion, water-related effects, and volume effects. Also, approaches in the use of hydrophilic, highly hydrated, and electrically neutral coatings have demonstrated a good ability to decrease cell adhesion. Moreover, amongst the available methods, the approach of layer-by-layer deposition has been known as an interesting process to manipulate protein and cell adhesion behavior.
Collapse
Affiliation(s)
- Mahdi Aghajani
- Department of Medical Nanotechnology, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran.,School of Advanced Technologies in Medicine, Golestan University of Medical Sciences, Gorgan, Iran
| | - Fariba Esmaeili
- Department of Medical Nanotechnology, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
22
|
Clapis JR, Fan MJ, Kovarik ML. Supported bilayer membranes for reducing cell adhesion in microfluidic devices. ANALYTICAL METHODS : ADVANCING METHODS AND APPLICATIONS 2021; 13:1535-1540. [PMID: 33690748 DOI: 10.1039/d0ay01992e] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
The high surface area-to-volume ratio of microfluidic channels makes them susceptible to fouling and clogging when used for biological analyses, including cell-based assays. We evaluated the role of electrostatic and van der Waals interactions in cell adhesion in PDMS microchannels coated with supported lipid bilayers and identified conditions that resulted in minimal cell adhesion. For low ionic strength buffer, optimum results were obtained for a zwitterionic coating of pure egg phosphatidylcholine; for a rich growth medium, the best results were obtained for zwitterionic bilayers or those with slight negative or moderate positive charge from the incorporation of 5-10 mol% egg phosphatidylglycerol or 30 mol% ethylphosphocholine. In both solutions, the presence of 10 g L-1 glucose in the cell suspension reduced cell adhesion. Under optimum conditions, all cells were consistently removed from the channels, demonstrating the utility of these coatings for whole-cell microfluidic assays. These results provide practical information for immediate application and suggest future research areas on cell-lipid interactions.
Collapse
Affiliation(s)
- Julia R Clapis
- Department of Chemistry, Trinity College, 300 Summit St., Hartford, CT 06106, USA.
| | | | | |
Collapse
|
23
|
Li M, Jiang S, Simon J, Paßlick D, Frey ML, Wagner M, Mailänder V, Crespy D, Landfester K. Brush Conformation of Polyethylene Glycol Determines the Stealth Effect of Nanocarriers in the Low Protein Adsorption Regime. NANO LETTERS 2021; 21:1591-1598. [PMID: 33560851 PMCID: PMC8023711 DOI: 10.1021/acs.nanolett.0c03756] [Citation(s) in RCA: 106] [Impact Index Per Article: 26.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/12/2023]
Abstract
For nanocarriers with low protein affinity, we show that the interaction of nanocarriers with cells is mainly affected by the density, the molecular weight, and the conformation of polyethylene glycol (PEG) chains bound to the nanocarrier surface. We achieve a reduction of nonspecific uptake of ovalbumin nanocarriers by dendritic cells using densely packed PEG chains with a "brush" conformation instead of the collapsed "mushroom" conformation. We also control to a minor extent the dysopsonin adsorption by tailoring the conformation of attached PEG on the nanocarriers. The brush conformation of PEG leads to a stealth behavior of the nanocarriers with inhibited uptake by phagocytic cells, which is a prerequisite for successful in vivo translation of nanomedicine to achieve long blood circulation and targeted delivery. We can clearly correlate the brush conformation of PEG with inhibited phagocytic uptake of the nanocarriers. This study shows that, in addition to the surface's chemistry, the conformation of polymers controls cellular interactions of the nanocarriers.
Collapse
Affiliation(s)
- Mengyi Li
- Max
Planck Institute for Polymer Research, Ackermannweg 10, 55128 Mainz, Germany
| | - Shuai Jiang
- Max
Planck Institute for Polymer Research, Ackermannweg 10, 55128 Mainz, Germany
| | - Johanna Simon
- Max
Planck Institute for Polymer Research, Ackermannweg 10, 55128 Mainz, Germany
- Department
of Dermatology, Johannes-Gutenberg University, 55131 Mainz, Germany
| | - David Paßlick
- Max
Planck Institute for Polymer Research, Ackermannweg 10, 55128 Mainz, Germany
- Department
of Dermatology, Johannes-Gutenberg University, 55131 Mainz, Germany
| | - Marie-Luise Frey
- Max
Planck Institute for Polymer Research, Ackermannweg 10, 55128 Mainz, Germany
| | - Manfred Wagner
- Max
Planck Institute for Polymer Research, Ackermannweg 10, 55128 Mainz, Germany
| | - Volker Mailänder
- Max
Planck Institute for Polymer Research, Ackermannweg 10, 55128 Mainz, Germany
- Department
of Dermatology, Johannes-Gutenberg University, 55131 Mainz, Germany
| | - Daniel Crespy
- Department
of Materials Science and Engineering, School of Molecular Science
and Engineering, Vidyasirimedhi Institute
of Science and Technology (VISTEC), Rayong 21210, Thailand
| | - Katharina Landfester
- Max
Planck Institute for Polymer Research, Ackermannweg 10, 55128 Mainz, Germany
| |
Collapse
|
24
|
Bunker A, Róg T. Mechanistic Understanding From Molecular Dynamics Simulation in Pharmaceutical Research 1: Drug Delivery. Front Mol Biosci 2020; 7:604770. [PMID: 33330633 PMCID: PMC7732618 DOI: 10.3389/fmolb.2020.604770] [Citation(s) in RCA: 56] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Accepted: 11/02/2020] [Indexed: 12/12/2022] Open
Abstract
In this review, we outline the growing role that molecular dynamics simulation is able to play as a design tool in drug delivery. We cover both the pharmaceutical and computational backgrounds, in a pedagogical fashion, as this review is designed to be equally accessible to pharmaceutical researchers interested in what this new computational tool is capable of and experts in molecular modeling who wish to pursue pharmaceutical applications as a context for their research. The field has become too broad for us to concisely describe all work that has been carried out; many comprehensive reviews on subtopics of this area are cited. We discuss the insight molecular dynamics modeling has provided in dissolution and solubility, however, the majority of the discussion is focused on nanomedicine: the development of nanoscale drug delivery vehicles. Here we focus on three areas where molecular dynamics modeling has had a particularly strong impact: (1) behavior in the bloodstream and protective polymer corona, (2) Drug loading and controlled release, and (3) Nanoparticle interaction with both model and biological membranes. We conclude with some thoughts on the role that molecular dynamics simulation can grow to play in the development of new drug delivery systems.
Collapse
Affiliation(s)
- Alex Bunker
- Division of Pharmaceutical Biosciences, Drug Research Program, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland
| | - Tomasz Róg
- Department of Physics, University of Helsinki, Helsinki, Finland
| |
Collapse
|
25
|
Zhang P, Pham T, Liu C, Leon Plata P, Kalkowski J, Cheng G, Bu W, Lin B, Liu Y. Impeded Molecular Reorganization by Polyethylene Glycol Conjugation Revealed by X-ray Reflectivity and Diffraction Measurements. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2020; 36:7573-7581. [PMID: 32501715 DOI: 10.1021/acs.langmuir.0c01202] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Polyethylene glycol (PEG) coatings have been widely applied in pharmaceutical and biomedical systems to prevent nonspecific protein absorption, increase vesicle blood circulation time, and sustain drug release. This study systematically investigated the planar interfacial organization of phospholipid monolayers containing various amounts of PEG conjugations before and after enzyme-catalyzed degradation of the lipids using X-ray reflectivity and grazing incidence X-ray diffraction techniques. Results showed that attaching PEG to the headgroup of the lipids up to 15 mol % had limited effects on molecular packing of the lipid monolayers in the condensed phase at the gas-liquid interface and negligible effects on the enzyme adsorption to the interface. After enzyme-catalyzed degradation, equimolar fatty acids and lyso PC were generated. The fatty acids together with the subphase Ca2+ self-assembled into highly organized multilayer domains at the interface. The X-ray measurements unambiguously revealed that the densely packed PEG markedly hindered microphase separation and formation of the palmitic acid-Ca2+ complexes.
Collapse
Affiliation(s)
- Pin Zhang
- Department of Chemical Engineering, University of Illinois at Chicago, Chicago, Illinois 60608, United States
| | - Tiep Pham
- Department of Chemical Engineering, University of Illinois at Chicago, Chicago, Illinois 60608, United States
| | - Chang Liu
- Department of Chemical Engineering, University of Illinois at Chicago, Chicago, Illinois 60608, United States
| | - Paola Leon Plata
- Department of Chemical Engineering, University of Illinois at Chicago, Chicago, Illinois 60608, United States
| | - Joseph Kalkowski
- Department of Chemical Engineering, University of Illinois at Chicago, Chicago, Illinois 60608, United States
| | - Gang Cheng
- Department of Chemical Engineering, University of Illinois at Chicago, Chicago, Illinois 60608, United States
| | - Wei Bu
- NSF's ChemMatCARS, University of Chicago, Chicago, Illinois 60637, United States
| | - Binhua Lin
- NSF's ChemMatCARS, University of Chicago, Chicago, Illinois 60637, United States
| | - Ying Liu
- Department of Chemical Engineering, University of Illinois at Chicago, Chicago, Illinois 60608, United States
- Richard & Loan Hill Department of Bioengineering, University of Illinois at Chicago, Chicago, Illinois 60607, United States
| |
Collapse
|
26
|
Lee H. Molecular Simulations of PEGylated Biomolecules, Liposomes, and Nanoparticles for Drug Delivery Applications. Pharmaceutics 2020; 12:E533. [PMID: 32531886 PMCID: PMC7355693 DOI: 10.3390/pharmaceutics12060533] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2020] [Revised: 06/04/2020] [Accepted: 06/08/2020] [Indexed: 12/17/2022] Open
Abstract
Since the first polyethylene glycol (PEG)ylated protein was approved by the FDA in 1990, PEGylation has been successfully applied to develop drug delivery systems through experiments, but these experimental results are not always easy to interpret at the atomic level because of the limited resolution of experimental techniques. To determine the optimal size, structure, and density of PEG for drug delivery, the structure and dynamics of PEGylated drug carriers need to be understood close to the atomic scale, as can be done using molecular dynamics simulations, assuming that these simulations can be validated by successful comparisons to experiments. Starting with the development of all-atom and coarse-grained PEG models in 1990s, PEGylated drug carriers have been widely simulated. In particular, recent advances in computer performance and simulation methodologies have allowed for molecular simulations of large complexes of PEGylated drug carriers interacting with other molecules such as anticancer drugs, plasma proteins, membranes, and receptors, which makes it possible to interpret experimental observations at a nearly atomistic resolution, as well as help in the rational design of drug delivery systems for applications in nanomedicine. Here, simulation studies on the following PEGylated drug topics will be reviewed: proteins and peptides, liposomes, and nanoparticles such as dendrimers and carbon nanotubes.
Collapse
Affiliation(s)
- Hwankyu Lee
- Department of Chemical Engineering, Dankook University, Yongin 16890, Korea
| |
Collapse
|
27
|
Murthy NS, Wang W, Sommerfeld SD, Vaknin D, Kohn J. Temperature-Activated PEG Surface Segregation Controls the Protein Repellency of Polymers. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2019; 35:9769-9776. [PMID: 31257890 PMCID: PMC6686177 DOI: 10.1021/acs.langmuir.9b00702] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
Poly(ethylene glycol) (PEG) is widely used to modulate the hydration states of biomaterials and is often applied to produce nonfouling surfaces. Here, we present X-ray scattering data, which show that it is the surface segregation of PEG, not just its presence in the bulk, that makes this happen by influencing the hydrophilicity of PEG-containing substrates. We demonstrate a temperature-dependent trigger that transforms a PEG-containing substrate from a protein-adsorbing to a protein-repelling state. On films of poly(desaminotyrosyl-tyrosine-co-PEG carbonate) with high (20 wt %) PEG content, in which very little protein adsorption is expected, quartz crystal microbalance data showed significant adsorption of fibrinogen and bovine serum albumin at 8 °C. The surface became protein-repellent at 37.5 °C. When the same polymer was iodinated, the polymer was protein-adsorbent, even when 37 wt % PEG was incorporated into the polymer backbone. This demonstrates that high PEG content by itself is not sufficient to repel proteins. By inhibiting phase separation either with iodine or by lowering the temperature, we show that PEG must phase-separate and bloom to the surface to create an antifouling surface. These results suggest an opportunity to design materials with high PEG content that can be switched from a protein-attractant to a protein-repellent state by inducing phase separation through brief exposure to temperatures above their glass transition temperature.
Collapse
Affiliation(s)
- N Sanjeeva Murthy
- New Jersey Center for Biomaterials , Rutgers - The State University of New Jersey , Piscataway , New Jersey 08854 , United States
| | - Wenjie Wang
- Ames Laboratory , Iowa State University , Ames , Iowa 50011 , United States
| | - Sven D Sommerfeld
- New Jersey Center for Biomaterials , Rutgers - The State University of New Jersey , Piscataway , New Jersey 08854 , United States
| | - David Vaknin
- Ames Laboratory , Iowa State University , Ames , Iowa 50011 , United States
| | - Joachim Kohn
- Ames Laboratory , Iowa State University , Ames , Iowa 50011 , United States
| |
Collapse
|
28
|
Liposome and immune system interplay: Challenges and potentials. J Control Release 2019; 305:194-209. [DOI: 10.1016/j.jconrel.2019.05.030] [Citation(s) in RCA: 76] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2019] [Revised: 05/15/2019] [Accepted: 05/17/2019] [Indexed: 01/20/2023]
|
29
|
Yan Q, Wang C, Wang J, Pan L, Zhang Z. Controllable and flexible cellular network for virus cell-to-cell spread. CHINESE CHEM LETT 2019. [DOI: 10.1016/j.cclet.2019.03.028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
|
30
|
Schmidt ST, Olsen CL, Franzyk H, Wørzner K, Korsholm KS, Rades T, Andersen P, Foged C, Christensen D. Comparison of two different PEGylation strategies for the liposomal adjuvant CAF09: Towards induction of CTL responses upon subcutaneous vaccine administration. Eur J Pharm Biopharm 2019; 140:29-39. [PMID: 31055066 DOI: 10.1016/j.ejpb.2019.04.020] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2018] [Revised: 02/20/2019] [Accepted: 04/30/2019] [Indexed: 01/28/2023]
Abstract
Using subunit vaccines, e.g., based on peptide or protein antigens, to teach the immune system to kill abnormal host cells via induction of cytotoxic T lymphocytes (CTL) is a promising strategy against intracellular infections and cancer. However, customized adjuvants are required to potentiate antigen-specific cellular immunity. One strong CTL-inducing adjuvant is the liposomal cationic adjuvant formulation (CAF)09, which is composed of dimethyldioctadecylammonium (DDA) bromide, monomycoloyl glycerol (MMG) analogue 1 and polyinosinic:polycytidylic acid [poly(I:C)]. However, this strong CTL induction requires intraperitoneal administration because the vaccine forms a depot at the site of injection (SOI) after subcutaneous (s.c.) or intramuscular (i.m.) injection, and depot formation impedes the crucial vaccine targeting to the cross-presenting dendritic cells (DCs) residing in the lymph nodes (LNs). The purpose of the present study was to investigate the effect of polyethylene glycol (PEG) grafting of CAF09 on the ability of the vaccine to induce antigen-specific CTL responses after s.c. administration. We hypothesized that steric stabilization and charge shielding of CAF09 by PEGylation may reduce depot formation at the SOI and enhance passive drainage to the LNs, eventually improving CTL induction. Hence, the vaccine (antigen/CAF09) was post-grafted with a novel type of anionic PEGylated peptides based on GDGDY repeats, which were end-conjugated with one or two PEG1000 moieties, resulting in mono- and bis-PEG-peptides of different lengths (10, 15 and 20 amino acid residues). For comparison, CAF09 was also grafted by inclusion of 1,2-distearoyl-sn-glycero-3-phosphoethanolamine-N-methoxy(PEG)-2000 (DSPE-PEG2000) in the bilayer structure during preparation. Grafting of CAF09 with either type of PEG resulted in charge shielding, evident from a reduced surface charge. Upon s.c. immunization of mice with the model antigen ovalbumin (OVA) adjuvanted with PEGylated CAF09, stronger CTL responses were induced as compared to immunization of mice with unadjuvanted OVA. Biodistribution studies confirmed that grafting of CAF09 with DSPE-PEG2000 improved the passive drainage of the vaccine to LNs, because a higher dose fraction was recovered in DCs present in the draining LNs, as compared to the dose fraction detected for non-PEGylated CAF09. In conclusion, PEGylation of CAF09 may be a useful strategy for the design of an adjuvant, which induces CTL responses after s.c. and i.m. administration. In the present studies, CAF09 grafted with 10 mol% DSPE-PEG2000 is the most promising of the tested adjuvants, but additional studies are required to further elucidate the potential of the strategy.
Collapse
Affiliation(s)
- Signe Tandrup Schmidt
- Department of Pharmacy, Faculty of Health and Medical Sciences, University of Copenhagen, Universitetsparken 2, 2100 Copenhagen Ø, Denmark; Statens Serum Institut, Department of Infectious Disease Immunology, Artillerivej 5, 2300 Copenhagen S, Denmark
| | - Camilla Line Olsen
- Department of Pharmacy, Faculty of Health and Medical Sciences, University of Copenhagen, Universitetsparken 2, 2100 Copenhagen Ø, Denmark; Statens Serum Institut, Department of Infectious Disease Immunology, Artillerivej 5, 2300 Copenhagen S, Denmark
| | - Henrik Franzyk
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Jagtvej 162, 2100 Copenhagen Ø, Denmark
| | - Katharina Wørzner
- Department of Pharmacy, Faculty of Health and Medical Sciences, University of Copenhagen, Universitetsparken 2, 2100 Copenhagen Ø, Denmark; Statens Serum Institut, Department of Infectious Disease Immunology, Artillerivej 5, 2300 Copenhagen S, Denmark
| | - Karen Smith Korsholm
- Statens Serum Institut, Department of Infectious Disease Immunology, Artillerivej 5, 2300 Copenhagen S, Denmark
| | - Thomas Rades
- Department of Pharmacy, Faculty of Health and Medical Sciences, University of Copenhagen, Universitetsparken 2, 2100 Copenhagen Ø, Denmark
| | - Peter Andersen
- Statens Serum Institut, Department of Infectious Disease Immunology, Artillerivej 5, 2300 Copenhagen S, Denmark
| | - Camilla Foged
- Department of Pharmacy, Faculty of Health and Medical Sciences, University of Copenhagen, Universitetsparken 2, 2100 Copenhagen Ø, Denmark
| | - Dennis Christensen
- Statens Serum Institut, Department of Infectious Disease Immunology, Artillerivej 5, 2300 Copenhagen S, Denmark.
| |
Collapse
|
31
|
Nemoto R, Fujieda K, Hiruta Y, Hishida M, Ayano E, Maitani Y, Nagase K, Kanazawa H. Liposomes with temperature-responsive reversible surface properties. Colloids Surf B Biointerfaces 2019; 176:309-316. [DOI: 10.1016/j.colsurfb.2019.01.007] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2018] [Revised: 12/28/2018] [Accepted: 01/02/2019] [Indexed: 10/27/2022]
|
32
|
Sun L, Xu H, Shao Y, Liu J, Fan LJ. Preparation and evaluation of fluorescent poly(p-phenyleneethylene) covalently coated microspheres with reactive sites for bioconjugation. J Colloid Interface Sci 2019; 540:362-370. [PMID: 30660793 DOI: 10.1016/j.jcis.2019.01.009] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2018] [Revised: 01/02/2019] [Accepted: 01/03/2019] [Indexed: 10/27/2022]
Abstract
Fluorescent microspheres with reactive sites for interacting with biomolecules are greatly demanded in flow cytometry based suspension array. Aiming to develop a new method for preparing fluorescent microspheres, two poly(p-phenyleneethylene) (PPE) conjugated polymers (CPs) with pedant carboxylic groups were synthesized via Sonogashira coupling and followed with hydrolysis of ester groups; then the conjugated polymers were immobilized onto monodispersed amino-modified porous poly(glycidylmethacrylate) (APGMA) microspheres via coupling reaction between carboxylic and amino groups to give APGMA-CP fluorescent microspheres. The fluorescent microspheres were found to have good photo- and thermal stability as well as negligible influence from rigorous washing. The emission was uniform all across the inner and surface of the spheres. To evaluate the effectiveness of bioconjugation on the fluorescent microspheres, fluorescein isothiocyanate isomer I (FITC) labeled bovine serum albumin (BSA) (BSA-FITC) was chosen as the representative biomolecule to react with the fluorescent microspheres to give APGMA-CP-BSA-FITC. In the flow cytometry study, fluorescence compensation between the V500 and FITC detectors (receiving signals from fluorophores excited by 405 nm and 488 nm, respectively), to remove the interference between the emission of FITC and CPs, was realized using singly-stained microspheres. Finally, APGMA-CP-BSA-FITC microspheres were found to be double positive for CP and FITC with very high percentage (>95%), suggesting the bioconjugation is very effective. This study provides a facile method for simultaneous introduction of fluorescence and reactive sites onto the microspheres, which is very promising to be used as general strategy for fabricating fluorescence microspheres for application in high-throughput technology.
Collapse
Affiliation(s)
- Lijuan Sun
- Department of Polymer Science and Engineering, College of Chemistry, Chemical Engineering and Materials Science, Soochow University, Suzhou, Jiangsu 215123, PR China
| | - Haibo Xu
- Department of Polymer Science and Engineering, College of Chemistry, Chemical Engineering and Materials Science, Soochow University, Suzhou, Jiangsu 215123, PR China
| | - Ya Shao
- Department of Polymer Science and Engineering, College of Chemistry, Chemical Engineering and Materials Science, Soochow University, Suzhou, Jiangsu 215123, PR China
| | - Jiangxin Liu
- Department of Polymer Science and Engineering, College of Chemistry, Chemical Engineering and Materials Science, Soochow University, Suzhou, Jiangsu 215123, PR China
| | - Li-Juan Fan
- Department of Polymer Science and Engineering, College of Chemistry, Chemical Engineering and Materials Science, Soochow University, Suzhou, Jiangsu 215123, PR China.
| |
Collapse
|
33
|
Abstract
PURPOSE OF REVIEW Non-invasive molecular imaging is currently used as a research technique to better understand disease pathophysiology. There are also many potential clinical applications where molecular imaging may provide unique information that allows either earlier or more definitive diagnosis, or can guide precision medicine-based decisions on therapy. Contrast-enhanced ultrasound (CEU) with targeted microbubble contrast agents is one such technique that has been developed that has the unique properties of providing rapid information and revealing information only on events that occur within the vascular space. RECENT FINDINGS CEU molecular probes have been developed for a wide variety of disease states including atherosclerosis, vascular inflammation, thrombosis, tumor neovascularization, and ischemic injury. While the technique has not yet been adapted to clinical use, it has been used to reveal pathological processes, to identify new therapeutic targets, and to test the efficacy of novel treatments. This review will explore the physical basis for CEU molecular imaging, its strengths and limitations compared to other molecular imaging modalities, and the pre-clinical translational research experience.
Collapse
Affiliation(s)
- Eran Brown
- Department of Biomedical Engineering, Oregon Health & Science University, Portland, OR, USA.,Knight Cardiovascular Institute, UHN-62, Oregon Health & Science University, 3181 SW Sam Jackson Park Rd., Portland, OR, 97239, USA
| | - Jonathan R Lindner
- Knight Cardiovascular Institute, UHN-62, Oregon Health & Science University, 3181 SW Sam Jackson Park Rd., Portland, OR, 97239, USA. .,Oregon National Primate Research Center (J.R.L.), Oregon Health & Science University, Portland, OR, USA.
| |
Collapse
|
34
|
Jin Q, Deng Y, Chen X, Ji J. Rational Design of Cancer Nanomedicine for Simultaneous Stealth Surface and Enhanced Cellular Uptake. ACS NANO 2019; 13:954-977. [PMID: 30681834 DOI: 10.1021/acsnano.8b07746] [Citation(s) in RCA: 71] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/20/2023]
Abstract
Owing to the complex and still not fully understood physiological environment, the development of traditional nanosized drug delivery systems is very challenging for precision cancer therapy. It is very difficult to control the in vivo distribution of nanoparticles after intravenous injection. The ideal drug nanocarriers should not only have stealth surface for prolonged circulation time but also possess enhanced cellular internalization in tumor sites. Unfortunately, the stealth surface and enhanced cellular uptake seem contradictory to each other. How to integrate the two opposite aspects into one system is a very herculean but meaningful task. As an alternative drug delivery strategy, chameleon-like drug delivery systems were developed to achieve long circulation time while maintaining enhanced cancer cell uptake. Such drug nanocarriers can "turn off" their internalization ability during circulation. However, the enhanced cellular uptake can be readily activated upon arriving at tumor tissues. In this way, stealth surface and enhanced uptake are of dialectical unity in drug delivery. In this review, we focus on the surface engineering of drug nanocarriers to obtain simultaneous stealth surfaces in circulation and enhanced uptake in tumors. The current strategies and ongoing developments, including programmed tumor-targeting strategies and some specific zwitterionic surfaces, will be discussed in detail.
Collapse
Affiliation(s)
- Qiao Jin
- MOE Key Laboratory of Macromolecule Synthesis and Functionalization of Ministry of Education, Department of Polymer Science and Engineering , Zhejiang University , Hangzhou 310027 , Zhejiang Province , P.R. China
| | - Yongyan Deng
- MOE Key Laboratory of Macromolecule Synthesis and Functionalization of Ministry of Education, Department of Polymer Science and Engineering , Zhejiang University , Hangzhou 310027 , Zhejiang Province , P.R. China
| | - Xiaohui Chen
- MOE Key Laboratory of Macromolecule Synthesis and Functionalization of Ministry of Education, Department of Polymer Science and Engineering , Zhejiang University , Hangzhou 310027 , Zhejiang Province , P.R. China
| | - Jian Ji
- MOE Key Laboratory of Macromolecule Synthesis and Functionalization of Ministry of Education, Department of Polymer Science and Engineering , Zhejiang University , Hangzhou 310027 , Zhejiang Province , P.R. China
| |
Collapse
|
35
|
Omolo CA, Kalhapure RS, Agrawal N, Jadhav M, Rambharose S, Mocktar C, Govender T. A hybrid of mPEG-b-PCL and G1-PEA dendrimer for enhancing delivery of antibiotics. J Control Release 2018; 290:112-128. [PMID: 30312719 DOI: 10.1016/j.jconrel.2018.10.005] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2018] [Revised: 09/17/2018] [Accepted: 10/06/2018] [Indexed: 01/02/2023]
|
36
|
|
37
|
Bernhard C, Roeters SJ, Franz J, Weidner T, Bonn M, Gonella G. Repelling and ordering: the influence of poly(ethylene glycol) on protein adsorption. Phys Chem Chem Phys 2018; 19:28182-28188. [PMID: 29022982 DOI: 10.1039/c7cp05445a] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Development of new materials for drug delivery and biosensing requires the fine-tuning of interfacial properties. We report here the influence of the poly(ethylene glycol) (PEG) grafting density in model phospholipid monolayers on the adsorption behavior of bovine serum albumin and human fibrinogen, not only with respect to the amount of adsorbed protein, but also its orientational ordering on the surface. As expected, with increasing interfacial PEG density, the amount of adsorbed protein decreases up to the point where complete protein repellency is reached. However, at intermediate concentrations, the net orientation of adsorbed fibrinogen is highest. The different proteins respond differently to PEG, not only in the amount of protein adsorbed, but also in the manner that proteins adsorb. The results show that for specific cases, tuning the interfacial PEG concentration allows to guide the protein adsorption configuration, a feature sought after in materials for both biosensing and biomedical applications.
Collapse
Affiliation(s)
- Christoph Bernhard
- Max Planck Institute for Polymer Research, Ackermannweg 10, D-55128 Mainz, Germany.
| | | | | | | | | | | |
Collapse
|
38
|
Ganji N, Khan IA, Bothun GD. Surface Activity of Poly(ethylene glycol)-Coated Silver Nanoparticles in the Presence of a Lipid Monolayer. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2018; 34:2039-2045. [PMID: 29309159 DOI: 10.1021/acs.langmuir.7b03743] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
We have investigated the surface activity of poly(ethylene glycol) (PEG)-coated silver nanoparticles (Ag-PEG) in the presence or absence of lipid monolayers comprised of monounsaturated dioleoylphosphocholine and dioleoylphosphoglycerol (DOPC/DOPG; 1:1 mol ratio). Dynamic measurements of surface pressure demonstrated that Ag-PEG were surface-active at the air/water interface. Surface excess concentrations suggested that at high Ag-PEG subphase concentrations, Ag-PEG assembled as densely packed monolayers in the presence and absence of a lipid monolayer. The presence of a lipid monolayer led to only a slight decrease in the excess surface concentration of Ag-PEG. Surface pressure-area isotherms showed that in the absence of lipids Ag-PEG increased the surface pressure up to 45 mN m-1 upon compression before the Ag-PEG surface layer collapsed. Our results suggest that surface activity of Ag-PEG was due to hydrophobic interactions imparted by a combination of the amphiphilic polymer coating and the hydrophobic dodecanethiol ligands bound to the Ag-PEG surface. With lipid present, Ag-PEG + lipid surface pressure-area (π-A) isotherms reflected Ag-PEG incorporation within the lipid monolayers. At high Ag-PEG concentrations, the π-A isotherms of the Ag-PEG + lipid films closely resembled that of Ag-PEG alone with a minimal contribution from the lipids present. Analysis of the subphase silver (Ag) and phosphorus (P) concentrations revealed that most of the adsorbed material remained at the air/lipid/water interface and was not forced into the aqueous subphase upon compression, confirming the presence of a composite Ag-PEG + lipid film. While interactions between "water-soluble" nanoparticles and lipids are often considered to be dominated by electrostatic interactions, these results provide further evidence that the amphiphilic character of a nanoparticle coating can also play a significant role.
Collapse
Affiliation(s)
- Nasim Ganji
- Department of Chemical Engineering, University of Rhode Island , 51 Lower College Road, Kinston, Rhode Island 02881, United States
| | - Iftheker A Khan
- Department of Chemical Engineering, University of Rhode Island , 51 Lower College Road, Kinston, Rhode Island 02881, United States
| | - Geoffrey D Bothun
- Department of Chemical Engineering, University of Rhode Island , 51 Lower College Road, Kinston, Rhode Island 02881, United States
| |
Collapse
|
39
|
Zhang DY, Hao Q, Liu J, Shi YS, Zhu J, Su L, Wang Y. Antifouling polyimide membrane with grafted silver nanoparticles and zwitterion. Sep Purif Technol 2018. [DOI: 10.1016/j.seppur.2017.10.018] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
|
40
|
Wei Y, Wang Y, Xia D, Guo S, Wang F, Zhang X, Gan Y. Thermosensitive Liposomal Codelivery of HSA-Paclitaxel and HSA-Ellagic Acid Complexes for Enhanced Drug Perfusion and Efficacy Against Pancreatic Cancer. ACS APPLIED MATERIALS & INTERFACES 2017; 9:25138-25151. [PMID: 28696100 DOI: 10.1021/acsami.7b07132] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
Fibrotic stroma and tumor-promoting pancreatic stellate cells (PSCs), critical characters in the pancreatic ductal adenocarcinoma (PDA) microenvironment, promote a tumor-facilitating environment that simultaneously prevents drug penetration into tumor foci and stimulates tumor growth. Nab-PTX, a human serum albumin (HSA) nanoparticle of paclitaxel (PTX), indicates enhanced matrix penetration in PDA probably due to its small size in vivo and high affinity of HSA with secreted protein acidic and rich in cysteine (SPARC), overexpressed in the PDA stroma. However, this HSA nanoparticle shows poor drug blood retention because of its weak colloidal stability in vivo, thus resulting in insufficient drug accumulation within tumor. Encapsulating HSA nanoparticles into the internal aqueous phase of ordinary liposomes improves their blood retention and the following tumor accumulation, but the large 200 nm size and shielding of HSA in the interior might make it difficult for this hybrid nanomedicine to penetrate the fibrotic PDA matrix and promote bioavailability of the payload. In our current work, we prepared ∼9 nm HSA complexes with an antitumor drug (PTX) and an anti-PSC drug (ellagic acid, EA), and these two HSA-drug complexes were further coencapsulated into thermosensitive liposomes (TSLs). This nanomedicine was named TSL/HSA-PE. The use of TSL/HSA-PE could improve drug blood retention, and upon reaching locally heated tumors, these TSLs can rapidly release their payloads (HSA-drug complexes) to facilitate their further tumor accumulation and matrix penetration. With superior tumor accumulation, impressive matrix penetration, and simultaneous action upon tumor cells and PSCs to disrupt PSCs-PDA interaction, TSL/HSA-PE treatment combined with heat exhibited strong tumor growth inhibition and apoptosis in vivo.
Collapse
Affiliation(s)
- Yan Wei
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences , 501 Haike Road, Shanghai 201203, China
| | - Yuxi Wang
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences , 501 Haike Road, Shanghai 201203, China
- Nano Science and Technology Institute, University of Science and Technology of China , 166 Renai Road, Suzhou, Jiangsu 215123, China
| | - Dengning Xia
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences , 501 Haike Road, Shanghai 201203, China
| | - Shiyan Guo
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences , 501 Haike Road, Shanghai 201203, China
| | - Feng Wang
- Shanghai Institute of Pharmaceutical Industry , 285 Gebaini Road, Shanghai 201203, China
| | - Xinxin Zhang
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences , 501 Haike Road, Shanghai 201203, China
| | - Yong Gan
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences , 501 Haike Road, Shanghai 201203, China
| |
Collapse
|
41
|
Balikov DA, Crowder SW, Boire TC, Lee JB, Gupta MK, Fenix AM, Lewis HN, Ambrose CM, Short PA, Kim CS, Burnette DT, Reilly MA, Murthy NS, Kang ML, Kim WS, Sung HJ. Tunable Surface Repellency Maintains Stemness and Redox Capacity of Human Mesenchymal Stem Cells. ACS APPLIED MATERIALS & INTERFACES 2017; 9:22994-23006. [PMID: 28621931 PMCID: PMC5687519 DOI: 10.1021/acsami.7b06103] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/06/2023]
Abstract
Human bone marrow derived mesenchymal stem cells (hMSCs) hold great promise for regenerative medicine due to their multipotent differentiation capacity and immunomodulatory capabilities. Substantial research has elucidated mechanisms by which extracellular cues regulate hMSC fate decisions, but considerably less work has addressed how material properties can be leveraged to maintain undifferentiated stem cells. Here, we show that synthetic culture substrates designed to exhibit moderate cell-repellency promote high stemness and low oxidative stress-two indicators of naïve, healthy stem cells-in commercial and patient-derived hMSCs. Furthermore, the material-mediated effect on cell behavior can be tuned by altering the molar percentage (mol %) and/or chain length of poly(ethylene glycol) (PEG), the repellant block linked to hydrophobic poly(ε-caprolactone) (PCL) in the copolymer backbone. Nano- and angstrom-scale characterization of the cell-material interface reveals that PEG interrupts the adhesive PCL domains in a chain-length-dependent manner; this prevents hMSCs from forming mature focal adhesions and subsequently promotes cell-cell adhesions that require connexin-43. This study is the first to demonstrate that intrinsic properties of synthetic materials can be tuned to regulate the stemness and redox capacity of hMSCs and provides new insight for designing highly scalable, programmable culture platforms for clinical translation.
Collapse
Affiliation(s)
- Daniel A. Balikov
- Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee 37235, United States
- Center for Stem Cell Biology, Vanderbilt University Medical Center, Nashville, Tennessee 37232, United States
| | - Spencer W. Crowder
- Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee 37235, United States
- Center for Stem Cell Biology, Vanderbilt University Medical Center, Nashville, Tennessee 37232, United States
| | - Timothy C. Boire
- Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee 37235, United States
| | - Jung Bok Lee
- Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee 37235, United States
| | - Mukesh K. Gupta
- Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee 37235, United States
| | - Aidan M. Fenix
- Department of Cell and Developmental Biology, Vanderbilt University Medical Center, Nashville, Tennessee 37232, United States
| | - Holley N. Lewis
- Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee 37235, United States
| | - Caitlyn M. Ambrose
- Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee 37235, United States
| | - Philip A. Short
- Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee 37235, United States
| | - Chang Soo Kim
- Department of Materials Science and Engineering, University of Wisconsin, Milwaukee, Wisconsin 53211, United States
| | - Dylan T. Burnette
- Department of Cell and Developmental Biology, Vanderbilt University Medical Center, Nashville, Tennessee 37232, United States
| | - Matthew A. Reilly
- Department of Biomedical Engineering, The Ohio State University, Columbus, Ohio 43210, United States
| | - N. Sanjeeva Murthy
- Department of Biomedical Engineering, Rutgers University, Piscataway, New Jersey 08854, United States
| | - Mi-Lan Kang
- Severance Biomedical Science Institute, College of Medicine, Yonsei University, Seoul 120-752, Republic of Korea
| | - Won Shik Kim
- Department of Otorhinolaryngology, College of Medicine, Yonsei University, Seoul 120-752, Republic of Korea
| | - Hak-Joon Sung
- Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee 37235, United States
- Center for Stem Cell Biology, Vanderbilt University Medical Center, Nashville, Tennessee 37232, United States
- Severance Biomedical Science Institute, College of Medicine, Yonsei University, Seoul 120-752, Republic of Korea
| |
Collapse
|
42
|
McCallen J, Prybylski J, Yang Q, Lai SK. Cross-Reactivity of Select PEG-Binding Antibodies to Other Polymers Containing a C-C-O Backbone. ACS Biomater Sci Eng 2017; 3:1605-1615. [DOI: 10.1021/acsbiomaterials.7b00147] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Affiliation(s)
- Justin McCallen
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy; ‡UNC/NCSU Joint Department of Biomedical Engineering; and §Department of Microbiology & Immunology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States
| | - John Prybylski
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy; ‡UNC/NCSU Joint Department of Biomedical Engineering; and §Department of Microbiology & Immunology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States
| | - Qi Yang
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy; ‡UNC/NCSU Joint Department of Biomedical Engineering; and §Department of Microbiology & Immunology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States
| | - Samuel K. Lai
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy; ‡UNC/NCSU Joint Department of Biomedical Engineering; and §Department of Microbiology & Immunology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States
| |
Collapse
|
43
|
Endo-Takahashi Y, Ooaku K, Ishida K, Suzuki R, Maruyama K, Negishi Y. Preparation of Angiopep-2 Peptide-Modified Bubble Liposomes for Delivery to the Brain. Biol Pharm Bull 2017; 39:977-83. [PMID: 27251499 DOI: 10.1248/bpb.b15-00994] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
In the development of therapeutic approaches for central nervous system diseases, a significant obstacle is efficient drug delivery across the blood-brain barrier owing to its low permeability. Various nanocarriers have been developed for brain-targeted drug delivery by modification with specific ligands. We have previously developed polyethylene glycol-modified liposomes (Bubble liposomes [BLs]) that entrap ultrasound (US) contrast gas and can serve as both plasmid DNA or small interfering RNA carriers and US contrast agents. In this study, we attempted to prepare brain-targeting BLs modified with Angiopep-2 (Ang2) peptide (Ang2-BLs). Ang2 is expected to be a useful ligand for the efficient delivery of nanocarriers to the brain. We showed that Ang2-BLs interacted specifically with brain endothelial cells via low-density lipoprotein receptor-related protein-1. We also confirmed that Ang2-BLs could entrap US contrast gas and had US imaging ability as well as unmodified BLs. Furthermore, we demonstrated that Ang2-BLs accumulated in brain tissue after intravascular injection. These results suggested that Ang2-BLs may be a useful tool for brain-targeted delivery and US imaging via systemic administration.
Collapse
Affiliation(s)
- Yoko Endo-Takahashi
- Department of Drug Delivery and Molecular Biopharmaceutics, School of Pharmacy, Tokyo University of Pharmacy and Life Sciences
| | | | | | | | | | | |
Collapse
|
44
|
d'Arcy R, Burke J, Tirelli N. Branched polyesters: Preparative strategies and applications. Adv Drug Deliv Rev 2016; 107:60-81. [PMID: 27189232 DOI: 10.1016/j.addr.2016.05.005] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2016] [Revised: 04/19/2016] [Accepted: 05/06/2016] [Indexed: 10/21/2022]
Abstract
In the last 20years, the availability of precision chemical tools (e.g. controlled/living polymerizations, 'click' reactions) has determined a step change in the complexity of both the macromolecular architecture and the chemical functionality of biodegradable polyesters. A major part in this evolution has been played by the possibilities that controlled macromolecular branching offers in terms of tailored physical/biological performance. This review paper aims to provide an updated overview of preparative techniques that derive hyperbranched, dendritic, comb, grafted polyesters through polycondensation or ring-opening polymerization mechanisms.
Collapse
|
45
|
Yang Q, Jacobs TM, McCallen JD, Moore DT, Huckaby JT, Edelstein JN, Lai SK. Analysis of Pre-existing IgG and IgM Antibodies against Polyethylene Glycol (PEG) in the General Population. Anal Chem 2016; 88:11804-11812. [PMID: 27804292 DOI: 10.1021/acs.analchem.6b03437] [Citation(s) in RCA: 236] [Impact Index Per Article: 26.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Circulating antibodies (Ab) that specifically bind polyethylene glycol (PEG), a biocompatible polymer routinely used in protein and nanoparticle therapeutics, have been associated with reduced efficacy of and/or adverse reactions to therapeutics modified with or containing PEG. Unlike most antidrug antibodies that are induced following initial drug dosing, anti-PEG Ab can be found in treatment-naïve individuals (i.e., individuals who have never undergone treatment with PEGylated drugs but most likely have been exposed to PEG through other means). Unfortunately, the true prevalence, quantitative levels, and Ab isotype of pre-existing anti-PEG Ab remain poorly understood. Here, using rigorously validated competitive ELISAs with engineered chimeric anti-PEG monoclonal Ab standards, we quantified the levels of anti-PEG IgM and different subclasses of anti-PEG IgG (IgG1-4) in both contemporary and historical human samples. We unexpectedly found, with 90% confidence, detectable levels of anti-PEG Ab in ∼72% of the contemporary specimens (18% IgG, 25% IgM, 30% both IgG and IgM). The vast majority of these samples contained low levels of anti-PEG Ab, with only ∼7% and ∼1% of all specimens possessing anti-PEG IgG and IgM in excess of 500 ng/mL, respectively. IgG2 was the predominant anti-PEG IgG subclass. Anti-PEG Ab's were also observed in ∼56% of serum samples collected during 1970-1999 (20% IgG, 19% IgM, and 16% both IgG and IgM), suggesting that the presence of PEG-specific antibodies may be a longstanding phenomenon. Anti-PEG IgG levels demonstrated correlation with patient age, but not with gender or race. The widespread prevalence of pre-existing anti-PEG Ab, coupled with high Ab levels in a subset of the population, underscores the potential importance of screening patients for anti-PEG Ab levels prior to administration of therapeutics containing PEG.
Collapse
Affiliation(s)
- Qi Yang
- Division of Molecular Pharmaceutics, Eshelman School of Pharmacy; ‡Lineberger Cancer Center, Biostatistics Shared Resource; §UNC/NCSU Joint Department of Biomedical Engineering; ∥Department of Microbiology & Immunology, University of North Carolina at Chapel Hill , Chapel Hill, North Carolina 27599, United States
| | - Timothy M Jacobs
- Division of Molecular Pharmaceutics, Eshelman School of Pharmacy; ‡Lineberger Cancer Center, Biostatistics Shared Resource; §UNC/NCSU Joint Department of Biomedical Engineering; ∥Department of Microbiology & Immunology, University of North Carolina at Chapel Hill , Chapel Hill, North Carolina 27599, United States
| | - Justin D McCallen
- Division of Molecular Pharmaceutics, Eshelman School of Pharmacy; ‡Lineberger Cancer Center, Biostatistics Shared Resource; §UNC/NCSU Joint Department of Biomedical Engineering; ∥Department of Microbiology & Immunology, University of North Carolina at Chapel Hill , Chapel Hill, North Carolina 27599, United States
| | - Dominic T Moore
- Division of Molecular Pharmaceutics, Eshelman School of Pharmacy; ‡Lineberger Cancer Center, Biostatistics Shared Resource; §UNC/NCSU Joint Department of Biomedical Engineering; ∥Department of Microbiology & Immunology, University of North Carolina at Chapel Hill , Chapel Hill, North Carolina 27599, United States
| | - Justin T Huckaby
- Division of Molecular Pharmaceutics, Eshelman School of Pharmacy; ‡Lineberger Cancer Center, Biostatistics Shared Resource; §UNC/NCSU Joint Department of Biomedical Engineering; ∥Department of Microbiology & Immunology, University of North Carolina at Chapel Hill , Chapel Hill, North Carolina 27599, United States
| | - Jasmine N Edelstein
- Division of Molecular Pharmaceutics, Eshelman School of Pharmacy; ‡Lineberger Cancer Center, Biostatistics Shared Resource; §UNC/NCSU Joint Department of Biomedical Engineering; ∥Department of Microbiology & Immunology, University of North Carolina at Chapel Hill , Chapel Hill, North Carolina 27599, United States
| | - Samuel K Lai
- Division of Molecular Pharmaceutics, Eshelman School of Pharmacy; ‡Lineberger Cancer Center, Biostatistics Shared Resource; §UNC/NCSU Joint Department of Biomedical Engineering; ∥Department of Microbiology & Immunology, University of North Carolina at Chapel Hill , Chapel Hill, North Carolina 27599, United States
| |
Collapse
|
46
|
Zhang Y, Mintzer E, Uhrich KE. Synthesis and characterization of PEGylated bolaamphiphiles with enhanced retention in liposomes. J Colloid Interface Sci 2016; 482:19-26. [DOI: 10.1016/j.jcis.2016.07.013] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2016] [Revised: 07/05/2016] [Accepted: 07/07/2016] [Indexed: 10/21/2022]
|
47
|
Bunker A, Magarkar A, Viitala T. Rational design of liposomal drug delivery systems, a review: Combined experimental and computational studies of lipid membranes, liposomes and their PEGylation. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2016; 1858:2334-2352. [DOI: 10.1016/j.bbamem.2016.02.025] [Citation(s) in RCA: 103] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/21/2015] [Revised: 02/09/2016] [Accepted: 02/10/2016] [Indexed: 01/22/2023]
|
48
|
Ban DK, Paul S. Protein corona over silver nanoparticles triggers conformational change of proteins and drop in bactericidal potential of nanoparticles: Polyethylene glycol capping as preventive strategy. Colloids Surf B Biointerfaces 2016; 146:577-84. [DOI: 10.1016/j.colsurfb.2016.06.050] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2016] [Revised: 06/25/2016] [Accepted: 06/27/2016] [Indexed: 01/27/2023]
|
49
|
Ran R, Middelberg APJ, Zhao CX. Microfluidic synthesis of multifunctional liposomes for tumour targeting. Colloids Surf B Biointerfaces 2016; 148:402-410. [PMID: 27639490 DOI: 10.1016/j.colsurfb.2016.09.016] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2016] [Revised: 09/09/2016] [Accepted: 09/11/2016] [Indexed: 01/09/2023]
Abstract
Nanotechnology has started a new era in engineering multifunctional nanoparticles for diagnosis and therapeutics by incorporating therapeutic drugs, targeting ligands, stimuli-responsive release and imaging molecules. However, more functionality requires more complex synthesis processes, resulting in poor reproducibility, low yield and high production cost, hence difficulties in clinical translation. Herein we report a one-step microfluidic method for making multifunctional liposomes. Three formulations were prepared using this simple method, including plain liposomes, PEGylated liposomes and folic acid functionalised liposomes, all with a fluorescence dye encapsulated for imaging. The size and surface properties of these liposomes can be precisely controlled by simply tuning the flow rate ratio and the ratio of the lipids to PEGylated lipid (DSPE-PEG2000) and to the DSPE-PEG2000-Folate, respectively. The synthesised liposomes remained stable under mimic serum conditions. Compared to the plain liposomes and PEGylated liposomes, the targeted folic acid functionalised liposomes exhibited enhanced cellular uptake by the FA receptor positive SKOV3 cells, but not the negative MCF7 cells, and this enhanced uptake could be inhibited by adding excess free folic acid, indicating high specificity of FA ligand-receptor endocytosis. Further evaluation using the 3D tumour spheroid model also showed higher internalisation of the targeted liposome formulation in comparison with the PEGylated one. To the best of our knowledge, this work demonstrates for the first time the versatility of this microfluidic method for making different liposome formulations in a single step, their superior physicochemical properties as well as the enhanced cellular uptake and tumour spheroid uptake of the targeted liposomes.
Collapse
Affiliation(s)
- Rui Ran
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, St. Lucia, QLD, 4072, Australia
| | - Anton P J Middelberg
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, St. Lucia, QLD, 4072, Australia
| | - Chun-Xia Zhao
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, St. Lucia, QLD, 4072, Australia.
| |
Collapse
|
50
|
Corbo C, Parodi A, Evangelopoulos M, Engler DA, Matsunami RK, Engler AC, Molinaro R, Scaria S, Salvatore F, Tasciotti E. Proteomic Profiling of a Biomimetic Drug Delivery Platform. Curr Drug Targets 2016; 16:1540-7. [PMID: 25382209 DOI: 10.2174/1389450115666141109211413] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2014] [Accepted: 10/31/2014] [Indexed: 01/01/2023]
Abstract
Current delivery platforms are typically designed for prolonged circulation that favors superior accumulation of the payload in the targeted tissue. The design of efficient surface modifications determines both a longer circulation time and targeting abilities of particles. The optimization of synthesis protocols to efficiently combine targeting molecules and elements that allow for an increased circulation time can be challenging and almost impossible when several functional elements are needed. On the other hand, in the last decade, the development of bioinspired technologies was proposed as a new approach with which to increase particle safety, biocompatibility and targeting, while maintaining the synthesis protocols simple and reproducible. Recently, we developed a new drug delivery system inspired by the biology of immune cells called leukolike vector (LLV) and formed by a nanoporous silicon core and a shell derived from the leucocyte cell membrane. The goal of this study is to investigate the protein content of the LLV. Here we report the proteomic profiling of the LLV and demonstrate that our approach can be used to modify the surface of synthetic particles with more than 150 leukocyte membrane associated proteins that determine particle safety, circulation time and targeting abilities towards inflamed endothelium.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | - Ennio Tasciotti
- Department of Nanomedicine, The Houston Methodist Research Institute, Houston, 6670 Bertner Avenue, Houston Texas 77030, USA.
| |
Collapse
|