1
|
Esmaeilpour D, Ghomi M, Zare EN, Sillanpää M. Nanotechnology-Enhanced siRNA Delivery: Revolutionizing Cancer Therapy. ACS APPLIED BIO MATERIALS 2025. [PMID: 40354673 DOI: 10.1021/acsabm.5c00489] [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: 05/14/2025]
Abstract
RNA interference (RNAi) has emerged as a transformative approach for cancer therapy, enabling precise gene silencing through small interfering RNA (siRNA). However, the clinical application of siRNA-based treatments faces challenges such as rapid degradation, inefficient cellular uptake, and immune system clearance. Nanotechnology-enhanced siRNA delivery has revolutionized cancer therapy by addressing these limitations, improving siRNA stability, tumor-specific targeting, and therapeutic efficacy. Recent advancements in nanocarrier engineering have introduced innovative strategies to enhance the safety and precision of siRNA-based therapies, offering new opportunities for personalized medicine. This review highlights three key innovations in nanotechnology-enhanced siRNA delivery: artificial intelligence (AI)-driven nanocarrier design, multifunctional nanoparticles for combined therapeutic strategies, and biomimetic nanocarriers for enhanced biocompatibility. AI-driven nanocarriers utilize machine learning algorithms to optimize nanoparticle properties, improving drug release profiles and minimizing off-target effects. Multifunctional nanoparticles integrate siRNA with chemotherapy, immunotherapy, or photothermal therapy, enabling synergistic treatment approaches that enhance therapeutic outcomes and reduce drug resistance. Biomimetic nanocarriers, including exosome-mimicking systems and cell-membrane-coated nanoparticles, improve circulation time, immune evasion, and targeted tumor delivery. These innovations collectively enhance the precision, efficiency, and safety of siRNA-based cancer therapies. The scope and novelty of these advancements lie in their ability to overcome the primary barriers of siRNA delivery while paving the way for clinically viable solutions. This review provides a comprehensive analysis of the latest developments in nanocarrier fabrication, preclinical and clinical studies, and safety assessments. By integrating AI-driven design, multifunctionality, and biomimicry, nanotechnology-enhanced siRNA delivery holds immense potential for the future of precision cancer therapy.
Collapse
Affiliation(s)
- Donya Esmaeilpour
- Center for Nanotechnology in Drug Delivery, School of Pharmacy, Shiraz University of Medical Science, Shiraz 71345-1583, Iran
| | - Matineh Ghomi
- Chemistry Department, Faculty of Science, Shahid Chamran University of Ahvaz, Ahvaz 6153753843 Iran
| | - Ehsan Nazarzadeh Zare
- School of Chemistry, Damghan University, Damghan 36716-45667, Iran
- Centre for Research Impact & Outcome, Chitkara University Institute of Engineering and Technology, Chitkara University, Rajpura 140401, Punjab, India
| | - Mika Sillanpää
- Institute of Research and Development, Duy Tan University, Da Nang 550000, Vietnam
- School of Engineering & Technology, Duy Tan University, Da Nang 550000, Vietnam
| |
Collapse
|
2
|
Xu Y, Fourniols T, Labrak Y, Préat V, Beloqui A, des Rieux A. Surface Modification of Lipid-Based Nanoparticles. ACS NANO 2022; 16:7168-7196. [PMID: 35446546 DOI: 10.1021/acsnano.2c02347] [Citation(s) in RCA: 81] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
There is a growing interest in the development of lipid-based nanocarriers for multiple purposes, including the recent increase of these nanocarriers as vaccine components during the COVID-19 pandemic. The number of studies that involve the surface modification of nanocarriers to improve their performance (increase the delivery of a therapeutic to its target site with less off-site accumulation) is enormous. The present review aims to provide an overview of various methods associated with lipid nanoparticle grafting, including techniques used to separate grafted nanoparticles from unbound ligands or to characterize grafted nanoparticles. We also provide a critical perspective on the usefulness and true impact of these modifications on overcoming different biological barriers, with our prediction on what to expect in the near future in this field.
Collapse
Affiliation(s)
- Yining Xu
- Advanced Drug Delivery and Biomaterials, UCLouvain, Université Catholique de Louvain, Louvain Drug Research Institute, Avenue Mounier, 73 B1.73.12, 1200 Brussels, Belgium
| | - Thibaut Fourniols
- Advanced Drug Delivery and Biomaterials, UCLouvain, Université Catholique de Louvain, Louvain Drug Research Institute, Avenue Mounier, 73 B1.73.12, 1200 Brussels, Belgium
| | - Yasmine Labrak
- Advanced Drug Delivery and Biomaterials, UCLouvain, Université Catholique de Louvain, Louvain Drug Research Institute, Avenue Mounier, 73 B1.73.12, 1200 Brussels, Belgium
- Bioanalysis and Pharmacology of Bioactive Lipids, UCLouvain, Université Catholique de Louvain, Louvain Drug Research Institute, Avenue Mounier, 72 B1.72.01, 1200 Brussels, Belgium
| | - Véronique Préat
- Advanced Drug Delivery and Biomaterials, UCLouvain, Université Catholique de Louvain, Louvain Drug Research Institute, Avenue Mounier, 73 B1.73.12, 1200 Brussels, Belgium
| | - Ana Beloqui
- Advanced Drug Delivery and Biomaterials, UCLouvain, Université Catholique de Louvain, Louvain Drug Research Institute, Avenue Mounier, 73 B1.73.12, 1200 Brussels, Belgium
| | - Anne des Rieux
- Advanced Drug Delivery and Biomaterials, UCLouvain, Université Catholique de Louvain, Louvain Drug Research Institute, Avenue Mounier, 73 B1.73.12, 1200 Brussels, Belgium
| |
Collapse
|
3
|
Ren J, Meng X, Xiang X, Ji F, Han L, Gao X, Jia L. Tyrosine-Based Dual-Functional Interface for Trapping and On-Site Photo-Induced Covalent Immobilization of Proteins. Bioconjug Chem 2022; 33:829-838. [PMID: 35413182 DOI: 10.1021/acs.bioconjchem.2c00091] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Tyrosine, a simple and well-available natural amino acid, is featured by the small size of the compound that contains multiple reactive groups. This study developed an efficient bioconjugation strategy using tyrosine-based dual-functional interfaces. When tyrosine molecules are immobilized on the surface of a supporting material through amino groups, their carboxyl groups can function as an attracting trap due to their anionic nature at neutral pH and ability to chelate nickel(II) ions (Ni2+), allowing the capture and enrichment of cationic proteins and histidine (His)-tagged proteins on the surface. The trapped proteins can be further covalently immobilized on site through ruthenium-mediated photochemical cross-linking, which has been found to be highly efficient and can be completed within minutes. This strategy was successfully applied to two different material systems. We found that tyrosine-modified agarose beads had a binding capacity of the His-tagged enhanced green fluorescent protein comparable to that of commonly used nitrilotriacetic acid-based resins, and further covalent coupling via dityrosine cross-linking achieved a yield of 85% within 5 min, without compromising much on its fluorescence activity. On the surface of tyrosine-modified 316L stainless steel, lysozyme was captured through electrostatic interaction and further immobilized. The resultant surface exhibited remarkable antibacterial activity against both Staphylococcus aureus and Escherichia coli. Such a tyrosine-based capture-then-coupling method is featured by its simplicity, high coupling efficiency, and high utilization rate of target molecules, making it particularly suitable for the proteins that are highly priced or vulnerable to general immobilization chemistry.
Collapse
Affiliation(s)
- Jun Ren
- Liaoning Key Laboratory of Molecular Recognition and Imaging, School of Bioengineering, Dalian University of Technology, Dalian 116023, P. R. China
| | - Xiao Meng
- Liaoning Key Laboratory of Molecular Recognition and Imaging, School of Bioengineering, Dalian University of Technology, Dalian 116023, P. R. China
| | - Xu Xiang
- Liaoning Key Laboratory of Molecular Recognition and Imaging, School of Bioengineering, Dalian University of Technology, Dalian 116023, P. R. China
| | - Fangling Ji
- Liaoning Key Laboratory of Molecular Recognition and Imaging, School of Bioengineering, Dalian University of Technology, Dalian 116023, P. R. China
| | - Lulu Han
- Liaoning Key Laboratory of Molecular Recognition and Imaging, School of Bioengineering, Dalian University of Technology, Dalian 116023, P. R. China
| | - Xiaorong Gao
- Liaoning Key Laboratory of Molecular Recognition and Imaging, School of Bioengineering, Dalian University of Technology, Dalian 116023, P. R. China
| | - Lingyun Jia
- Liaoning Key Laboratory of Molecular Recognition and Imaging, School of Bioengineering, Dalian University of Technology, Dalian 116023, P. R. China
| |
Collapse
|
4
|
Glassman PM, Hood ED, Ferguson LT, Zhao Z, Siegel DL, Mitragotri S, Brenner JS, Muzykantov VR. Red blood cells: The metamorphosis of a neglected carrier into the natural mothership for artificial nanocarriers. Adv Drug Deliv Rev 2021; 178:113992. [PMID: 34597748 PMCID: PMC8556370 DOI: 10.1016/j.addr.2021.113992] [Citation(s) in RCA: 52] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Revised: 08/26/2021] [Accepted: 09/24/2021] [Indexed: 12/18/2022]
Abstract
Drug delivery research pursues many types of carriers including proteins and other macromolecules, natural and synthetic polymeric structures, nanocarriers of diverse compositions and cells. In particular, liposomes and lipid nanoparticles represent arguably the most advanced and popular human-made nanocarriers, already in multiple clinical applications. On the other hand, red blood cells (RBCs) represent attractive natural carriers for the vascular route, featuring at least two distinct compartments for loading pharmacological cargoes, namely inner space enclosed by the plasma membrane and the outer surface of this membrane. Historically, studies of liposomal drug delivery systems (DDS) astronomically outnumbered and surpassed the RBC-based DDS. Nevertheless, these two types of carriers have different profile of advantages and disadvantages. Recent studies showed that RBC-based drug carriers indeed may feature unique pharmacokinetic and biodistribution characteristics favorably changing benefit/risk ratio of some cargo agents. Furthermore, RBC carriage cardinally alters behavior and effect of nanocarriers in the bloodstream, so called RBC hitchhiking (RBC-HH). This article represents an attempt for the comparative analysis of liposomal vs RBC drug delivery, culminating with design of hybrid DDSs enabling mutual collaborative advantages such as RBC-HH and camouflaging nanoparticles by RBC membrane. Finally, we discuss the key current challenges faced by these and other RBC-based DDSs including the issue of potential unintended and adverse effect and contingency measures to ameliorate this and other concerns.
Collapse
Affiliation(s)
- Patrick M Glassman
- Department of Systems Pharmacology and Translational Therapeutics, Center for Targeted Therapeutics and Translational Nanomedicine of the Institute for Translational Medicine and Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, United States
| | - Elizabeth D Hood
- Department of Systems Pharmacology and Translational Therapeutics, Center for Targeted Therapeutics and Translational Nanomedicine of the Institute for Translational Medicine and Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, United States
| | - Laura T Ferguson
- Department of Systems Pharmacology and Translational Therapeutics, Center for Targeted Therapeutics and Translational Nanomedicine of the Institute for Translational Medicine and Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, United States; Department of Medicine, Division of Pulmonary, Allergy, and Critical Care Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, United States
| | - Zongmin Zhao
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Illinois at Chicago, Chicago, IL 60612, United States
| | - Don L Siegel
- Department of Pathology & Laboratory Medicine, Division of Transfusion Medicine & Therapeutic Pathology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, United States
| | - Samir Mitragotri
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02138, United States
| | - Jacob S Brenner
- Department of Systems Pharmacology and Translational Therapeutics, Center for Targeted Therapeutics and Translational Nanomedicine of the Institute for Translational Medicine and Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, United States; Department of Medicine, Division of Pulmonary, Allergy, and Critical Care Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, United States
| | - Vladimir R Muzykantov
- Department of Systems Pharmacology and Translational Therapeutics, Center for Targeted Therapeutics and Translational Nanomedicine of the Institute for Translational Medicine and Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, United States.
| |
Collapse
|
5
|
Parada N, Romero-Trujillo A, Georges N, Alcayaga-Miranda F. Camouflage strategies for therapeutic exosomes evasion from phagocytosis. J Adv Res 2021; 31:61-74. [PMID: 34194832 PMCID: PMC8240105 DOI: 10.1016/j.jare.2021.01.001] [Citation(s) in RCA: 133] [Impact Index Per Article: 33.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Revised: 12/29/2020] [Accepted: 01/01/2021] [Indexed: 12/14/2022] Open
Abstract
Background Even though exosome-based therapy has been shown to be able to control the progression of different pathologies, the data revealed by pharmacokinetic studies warn of the low residence time of exogenous exosomes in circulation that can hinder the clinical translation of therapeutic exosomes. The macrophages related to the organs of the mononuclear phagocytic system are responsible primarily for the rapid clearance and retention of exosomes, which strongly limits the amount of exosomal particles available to reach the target tissue, accumulate in it and release with high efficiency its therapeutic cargo in acceptor target cells to exert the desired biological effect. Aim of review Endowing exosomes with surface modifications to evade the immune system is a plausible strategy to contribute to the suppression of exosomal clearance and increase the efficiency of their targeted content delivery. Here, we summarize the current evidence about the mechanisms underlying the recognition and sequestration of therapeutic exosomes by phagocytic cells. Also, we propose different strategies to generate 'invisible' exosomes for the immune system, through the incorporation of different anti-phagocytic molecules on the exosomes’ surface that allow increasing the circulating half-life of therapeutic exosomes with the purpose to increase their bioavailability to reach the target tissue, transfer their therapeutic molecular cargo and improve their efficacy profile. Key scientific concepts of review Macrophage-mediated phagocytosis are the main responsible behind the short half-life in circulation of systemically injected exosomes, hindering their therapeutic effect. Exosomes ‘Camouflage Cloak’ strategy using antiphagocytic molecules can contribute to the inhibition of exosomal clearance, hence, increasing the on-target effect. Some candidate molecules that could exert an antiphagocytic role are CD47, CD24, CD44, CD31, β2M, PD-L1, App1, and DHMEQ. Pre- and post-isolation methods for exosome engineering are compatible with the loading of therapeutic cargo and the expression of antiphagocytic surface molecules.
Collapse
Affiliation(s)
- Nicol Parada
- School of Medicine, Faculty of Medicine, Universidad de los Andes, Santiago, Chile.,Laboratory of Nano-Regenerative Medicine, Centro de Investigación e Innovación Biomédica (CIIB), Faculty of Medicine, Universidad de los Andes, Santiago, Chile
| | - Alfonso Romero-Trujillo
- School of Medicine, Faculty of Medicine, Universidad de los Andes, Santiago, Chile.,Laboratory of Nano-Regenerative Medicine, Centro de Investigación e Innovación Biomédica (CIIB), Faculty of Medicine, Universidad de los Andes, Santiago, Chile
| | - Nicolás Georges
- School of Medicine, Faculty of Medicine, Universidad de los Andes, Santiago, Chile.,Laboratory of Nano-Regenerative Medicine, Centro de Investigación e Innovación Biomédica (CIIB), Faculty of Medicine, Universidad de los Andes, Santiago, Chile
| | - Francisca Alcayaga-Miranda
- School of Medicine, Faculty of Medicine, Universidad de los Andes, Santiago, Chile.,Laboratory of Nano-Regenerative Medicine, Centro de Investigación e Innovación Biomédica (CIIB), Faculty of Medicine, Universidad de los Andes, Santiago, Chile.,Cells for Cells, Santiago, Chile.,Consorcio Regenero, Chilean Consortium for Regenerative Medicine, Santiago, Chile
| |
Collapse
|
6
|
Tang SY, Wei H, Yu CY. Peptide-functionalized delivery vehicles for enhanced cancer therapy. Int J Pharm 2021; 593:120141. [DOI: 10.1016/j.ijpharm.2020.120141] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2020] [Revised: 11/17/2020] [Accepted: 11/28/2020] [Indexed: 02/08/2023]
|
7
|
Abstract
Molecular conjugation refers to methods used in biomedicine, advanced materials and nanotechnology to link two partners - from small molecules to large and sometimes functionally complex biopolymers. The methods ideally have a broad structural scope, proceed under very mild conditions (including in H2O), occur at a rapid rate and in quantitative yield with no by-products, enable bioorthogonal reactivity and have zero toxicity. Over the past two decades, the field of click chemistry has emerged to afford us new and efficient methods of molecular conjugation. These methods are based on chemical reactions that produce permanently linked conjugates, and we refer to this field here as covalent click chemistry. Alternatively, if molecular conjugation is undertaken using a pair of complementary molecular recognition partners that associate strongly and selectively to form a thermodynamically stable non-covalent complex, then we refer to this strategy as non-covalent click chemistry. This Perspective is concerned with this latter approach and highlights two distinct applications of non-covalent click chemistry in molecular conjugation: the pre-assembly of molecular conjugates or surface-coated nanoparticles and the in situ capture of tagged biomolecular targets for imaging or analysis.
Collapse
Affiliation(s)
- Cynthia L Schreiber
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, IN, USA
| | - Bradley D Smith
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, IN, USA
| |
Collapse
|
8
|
Rigo S, Gunkel-Grabole G, Meier W, Palivan CG. Surfaces with Dual Functionality through Specific Coimmobilization of Self-Assembled Polymeric Nanostructures. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2019; 35:4557-4565. [PMID: 30296105 DOI: 10.1021/acs.langmuir.8b02812] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
Coimmobilization of functional, nanosized assemblies broadens the possibility to engineer dually functionalized active surfaces with a nanostructured texture. Surfaces decorated with different nanoassemblies, such as micelles, polymersomes, or nanoparticles are in high demand for various applications ranging from catalysis, biosensing up to antimicrobial surfaces. Here, we present a combination of bio-orthogonal and catalyst-free strain-promoted azide-alkyne click (SPAAC) and thiol-ene reactions to simultaneously coimmobilize various nanoassemblies; we selected polymersome-polymersome and polymersome-micelle assemblies. For the first time, the immobilization method using SPAAC reaction was studied in detail to attach soft, polymeric assemblies on a solid support. Together, the SPAAC and thiol-ene reactions successfully coimmobilized two unique self-assembled structures on the surfaces. Additionally, poly(dimethylsiloxane) (PDMS)-based polymersomes were used as "ink" for direct immobilization from a PDMS-based microstamp onto a surface creating locally defined patterns. Combining immobilization reactions has the advantage to attach any kind of nanoassembly pairs, resulting in surfaces with "desired" interfacial properties. Different nanoassemblies that encapsulate multiple active compounds coimmobilized on a surface will pave the way for the development of multifunctional surfaces with controlled properties and efficiency.
Collapse
Affiliation(s)
- Serena Rigo
- Department of Chemistry , University of Basel , Mattenstrasse 24a, BPR 1096 , CH-4002 Basel , Switzerland
| | - Gesine Gunkel-Grabole
- Department of Chemistry , University of Basel , Mattenstrasse 24a, BPR 1096 , CH-4002 Basel , Switzerland
| | - Wolfgang Meier
- Department of Chemistry , University of Basel , Mattenstrasse 24a, BPR 1096 , CH-4002 Basel , Switzerland
| | - Cornelia G Palivan
- Department of Chemistry , University of Basel , Mattenstrasse 24a, BPR 1096 , CH-4002 Basel , Switzerland
| |
Collapse
|
9
|
Hood ED, Greineder CF, Shuvaeva T, Walsh L, Villa CH, Muzykantov VR. Vascular Targeting of Radiolabeled Liposomes with Bio-Orthogonally Conjugated Ligands: Single Chain Fragments Provide Higher Specificity than Antibodies. Bioconjug Chem 2018; 29:3626-3637. [PMID: 30240185 DOI: 10.1021/acs.bioconjchem.8b00564] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Liposomes are a proven, versatile, and clinically viable technology platform for vascular delivery of drugs and imaging probes. Although targeted liposomes have the potential to advance these applications, complex formulations and the need for optimal affinity ligands and conjugation strategies challenge their translation. Herein, we employed copper-free click chemistry functionalized liposomes to target platelet-endothelial cell adhesion molecule (PECAM-1) and intracellular adhesion molecule (ICAM-1) by conjugating clickable monoclonal antibodies (Ab) or their single chain variable fragments (scFv). For direct, quantitative tracing, liposomes were surface chelated with 111In to a >90% radiochemical yield and purity. Particle size and distribution, stability, ligand surface density, and specific binding to target cells were characterized in vitro. Biodistribution of liposomes after IV injection was characterized in mice using isotope detection in organs and by noninvasive imaging (single-photon emission computed tomography/computed tomography, SPECT/CT). As much as 20-25% of injected dose of liposomes carrying PECAM and ICAM ligands, but not control IgG accumulated in the pulmonary vasculature. The immunospecificity of pulmonary targeting of scFv/liposomes to PECAM-1 and ICAM-1, respectively, was 10-fold and 2.5-fold higher than of Ab/liposomes. Therefore, the combination of optimal ligands, benign conjugation, and labeling yields liposomal formulations that may be used for highly effective and specific vascular targeting.
Collapse
Affiliation(s)
- Elizabeth D Hood
- Department of Systems Pharmacology and Translational Therapeutics , Perelman School of Medicine , 3400 Civic Center Boulevard, Bldg 421 , Philadelphia , Pennsylvania 19104-5158 , United States
| | - Colin F Greineder
- Department of Systems Pharmacology and Translational Therapeutics , Perelman School of Medicine , 3400 Civic Center Boulevard, Bldg 421 , Philadelphia , Pennsylvania 19104-5158 , United States
| | - Tea Shuvaeva
- Department of Systems Pharmacology and Translational Therapeutics , Perelman School of Medicine , 3400 Civic Center Boulevard, Bldg 421 , Philadelphia , Pennsylvania 19104-5158 , United States
| | - Landis Walsh
- Department of Systems Pharmacology and Translational Therapeutics , Perelman School of Medicine , 3400 Civic Center Boulevard, Bldg 421 , Philadelphia , Pennsylvania 19104-5158 , United States
| | - Carlos H Villa
- Department of Systems Pharmacology and Translational Therapeutics , Perelman School of Medicine , 3400 Civic Center Boulevard, Bldg 421 , Philadelphia , Pennsylvania 19104-5158 , United States
| | - Vladimir R Muzykantov
- Department of Systems Pharmacology and Translational Therapeutics , Perelman School of Medicine , 3400 Civic Center Boulevard, Bldg 421 , Philadelphia , Pennsylvania 19104-5158 , United States
| |
Collapse
|
10
|
Lee TH, Hirst DJ, Kulkarni K, Del Borgo MP, Aguilar MI. Exploring Molecular-Biomembrane Interactions with Surface Plasmon Resonance and Dual Polarization Interferometry Technology: Expanding the Spotlight onto Biomembrane Structure. Chem Rev 2018; 118:5392-5487. [PMID: 29793341 DOI: 10.1021/acs.chemrev.7b00729] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The molecular analysis of biomolecular-membrane interactions is central to understanding most cellular systems but has emerged as a complex technical challenge given the complexities of membrane structure and composition across all living cells. We present a review of the application of surface plasmon resonance and dual polarization interferometry-based biosensors to the study of biomembrane-based systems using both planar mono- or bilayers or liposomes. We first describe the optical principals and instrumentation of surface plasmon resonance, including both linear and extraordinary transmission modes and dual polarization interferometry. We then describe the wide range of model membrane systems that have been developed for deposition on the chips surfaces that include planar, polymer cushioned, tethered bilayers, and liposomes. This is followed by a description of the different chemical immobilization or physisorption techniques. The application of this broad range of engineered membrane surfaces to biomolecular-membrane interactions is then overviewed and how the information obtained using these techniques enhance our molecular understanding of membrane-mediated peptide and protein function. We first discuss experiments where SPR alone has been used to characterize membrane binding and describe how these studies yielded novel insight into the molecular events associated with membrane interactions and how they provided a significant impetus to more recent studies that focus on coincident membrane structure changes during binding of peptides and proteins. We then discuss the emerging limitations of not monitoring the effects on membrane structure and how SPR data can be combined with DPI to provide significant new information on how a membrane responds to the binding of peptides and proteins.
Collapse
Affiliation(s)
- Tzong-Hsien Lee
- Department of Biochemistry and Molecular Biology and Biomedicine Discovery Institute , Monash University , Clayton , VIC 3800 , Australia
| | - Daniel J Hirst
- Department of Biochemistry and Molecular Biology and Biomedicine Discovery Institute , Monash University , Clayton , VIC 3800 , Australia
| | - Ketav Kulkarni
- Department of Biochemistry and Molecular Biology and Biomedicine Discovery Institute , Monash University , Clayton , VIC 3800 , Australia
| | - Mark P Del Borgo
- Department of Biochemistry and Molecular Biology and Biomedicine Discovery Institute , Monash University , Clayton , VIC 3800 , Australia
| | - Marie-Isabel Aguilar
- Department of Biochemistry and Molecular Biology and Biomedicine Discovery Institute , Monash University , Clayton , VIC 3800 , Australia
| |
Collapse
|
11
|
Shaw SK, Liu W, Brennan SP, de Lourdes Betancourt-Mendiola M, Smith BD. Non-Covalent Assembly Method that Simultaneously Endows a Liposome Surface with Targeting Ligands, Protective PEG Chains, and Deep-Red Fluorescence Reporter Groups. Chemistry 2017; 23:12646-12654. [PMID: 28736857 DOI: 10.1002/chem.201702649] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2017] [Indexed: 12/28/2022]
Abstract
A new self-assembly method is used to rapidly functionalize the surface of liposomes without perturbing the membrane integrity or causing leakage of the aqueous contents. The key molecule is a cholesterol-squaraine-PEG conjugate with three important structural elements: a cholesterol membrane anchor, a fluorescent squaraine docking station that allows rapid and high-affinity macrocycle threading, and a long PEG-2000 chain to provide steric shielding of the decorated liposome. The two-step method involves spontaneous insertion of the conjugate into the outer leaflet of pre-formed liposomes followed by squaraine threading with a tetralactam macrocycle that has appended targeting ligands. A macrocycle with six carboxylates permitted immobilization of intact fluorescent liposomes on the surface of cationic polymer beads, whereas a macrocycle with six zinc(II)-dipicolylamine units enabled selective targeting of anionic membranes, including agglutination of bacteria in the presence of human cells.
Collapse
Affiliation(s)
- Scott K Shaw
- Department of Chemistry & Biochemistry, University of Notre Dame, 236 Nieuwland Science Hall, Notre Dame, IN., 46545, USA
| | - Wenqi Liu
- Department of Chemistry & Biochemistry, University of Notre Dame, 236 Nieuwland Science Hall, Notre Dame, IN., 46545, USA
| | - Seamus P Brennan
- Department of Chemistry & Biochemistry, University of Notre Dame, 236 Nieuwland Science Hall, Notre Dame, IN., 46545, USA
| | | | - Bradley D Smith
- Department of Chemistry & Biochemistry, University of Notre Dame, 236 Nieuwland Science Hall, Notre Dame, IN., 46545, USA
| |
Collapse
|
12
|
Moratz J, Klepel F, Ravoo BJ. Dynamic glycosylation of liposomes by thioester exchange. Org Biomol Chem 2017; 15:5089-5094. [PMID: 28585976 DOI: 10.1039/c7ob00805h] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
The interplay of dynamic functionalization and specific molecular recognition on biological membranes is key to numerous physiological processes. In this work we present a simple glycocalyx model based on the covalent yet reversible glycosylation of liposomes and subsequent recognition by a lectin. Reversible thioester exchange of membrane embedded amphiphilic thioesters with thiol-tagged d-mannose in solution is performed at physiologically relevant conditions. Recognition with the lectin concanavalin A is possible directly from this reaction mixture, leading to liposome agglutination. To the best of our knowledge, the dynamic covalent glycosylation of liposomes is so far unprecedented.
Collapse
Affiliation(s)
- Johanna Moratz
- Organic Chemistry Institute, Westfälische Wilhelms-Universität Münster, Corrensstrasse 40, 48149 Münster, Germany.
| | | | | |
Collapse
|