1
|
López RR, Ben El Khyat CZ, Chen Y, Tsering T, Dickinson K, Bustamante P, Erzingatzian A, Bartolomucci A, Ferrier ST, Douanne N, Mounier C, Stiharu I, Nerguizian V, Burnier JV. A synthetic model of bioinspired liposomes to study cancer-cell derived extracellular vesicles and their uptake by recipient cells. Sci Rep 2025; 15:8430. [PMID: 40069225 PMCID: PMC11897354 DOI: 10.1038/s41598-025-91873-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2024] [Accepted: 02/24/2025] [Indexed: 03/15/2025] Open
Abstract
Extracellular vesicles (EVs) are secreted by most cell types and play a central role in cell-cell communication. These naturally occurring nanoparticles have been particularly implicated in cancer, but EV heterogeneity and lengthy isolation methods with low yield make them difficult to study. To circumvent the challenges in EV research, we aimed to develop a unique synthetic model by engineering bioinspired liposomes to study EV properties and their impact on cellular uptake. We produced EV-like liposomes mimicking the physicochemical properties as cancer EVs. First, using a panel of cancer and non-cancer cell lines, small EVs were isolated by ultracentrifugation and characterized by dynamic light scattering (DLS) and nanoparticle tracking analysis (NTA). Cancer EVs ranged in mean size from 107.9 to 161 nm by NTA, hydrodynamic diameter from 152 to 355 nm by DLS, with a zeta potential ranging from - 25 to -6 mV. EV markers TSG101 and CD81 were positive on all EVs. Using a microfluidics bottom-up approach, liposomes were produced using the nanoprecipitation method adapted to micromixers developed by our group. A library of liposome formulations was created that mimicked the ranges of size (90-222 nm) and zeta potential (anionic [-47 mV] to neutral [-1 mV]) at a production throughput of up to 41 mL/h and yielding a concentration of 1 × 1012 particles per mL. EV size and zeta potential were reproduced by controlling the flow conditions and lipid composition set by a statistical model based on the response surface methodology. The model was fairly accurate with an R-squared > 70% for both parameters between the targeted EV and the obtained liposomes. Finally, the internalization of fluorescently labeled EV-like liposomes was assessed by confocal microscopy and flow cytometry, and correlated with decreasing liposome size and less negative zeta potential, providing insights into the effects of key EV physicochemical properties. Our data demonstrated that liposomes can be used as a powerful synthetic model of EVs. By mimicking cancer cell-derived EV properties, the effects on cellular internalization can be assessed individually and in combination. Taken together, we present a novel system that can accelerate research on the effects of EVs in cancer models.
Collapse
Affiliation(s)
- Rubén R López
- Cancer Research Program, Research Institute of the McGill University Health Centre, 1001 Decarie Blvd, Montreal, QC, H4A 3J1, Canada
- Department of Pathology, McGill University, Quebec, Canada
- Department of Electrical Engineering, École de Technologie supérieure, 1100 Notre Dame West, Montreal, QC, H3C 1K3, Canada
| | - Chaymaa Zouggari Ben El Khyat
- Department of Electrical Engineering, École de Technologie supérieure, 1100 Notre Dame West, Montreal, QC, H3C 1K3, Canada
| | - Yunxi Chen
- Cancer Research Program, Research Institute of the McGill University Health Centre, 1001 Decarie Blvd, Montreal, QC, H4A 3J1, Canada
- Department of Pathology, McGill University, Quebec, Canada
| | - Thupten Tsering
- Cancer Research Program, Research Institute of the McGill University Health Centre, 1001 Decarie Blvd, Montreal, QC, H4A 3J1, Canada
- Department of Pathology, McGill University, Quebec, Canada
| | - Kyle Dickinson
- Cancer Research Program, Research Institute of the McGill University Health Centre, 1001 Decarie Blvd, Montreal, QC, H4A 3J1, Canada
| | - Prisca Bustamante
- Cancer Research Program, Research Institute of the McGill University Health Centre, 1001 Decarie Blvd, Montreal, QC, H4A 3J1, Canada
- Department of Pathology, McGill University, Quebec, Canada
| | - Armen Erzingatzian
- Cancer Research Program, Research Institute of the McGill University Health Centre, 1001 Decarie Blvd, Montreal, QC, H4A 3J1, Canada
| | - Alexandra Bartolomucci
- Cancer Research Program, Research Institute of the McGill University Health Centre, 1001 Decarie Blvd, Montreal, QC, H4A 3J1, Canada
- Department of Pathology, McGill University, Quebec, Canada
| | - Sarah Tadhg Ferrier
- Cancer Research Program, Research Institute of the McGill University Health Centre, 1001 Decarie Blvd, Montreal, QC, H4A 3J1, Canada
- Department of Pathology, McGill University, Quebec, Canada
| | - Noélie Douanne
- Cancer Research Program, Research Institute of the McGill University Health Centre, 1001 Decarie Blvd, Montreal, QC, H4A 3J1, Canada
- Department of Pathology, McGill University, Quebec, Canada
| | - Catherine Mounier
- Department of biological sciences, Université du Québec à Montréal, 141 avenue du président Kennedy, Montreal, QC, H2X 1Y4, Canada
- Department of Mechanical, Industrial and Aerospace Engineering, Concordia University, 1455 de Maisonneuve Blvd. West, Montreal, QC, H3G 1M8, Canada
| | - Ion Stiharu
- Gerald Bronfman Department of Oncology, McGill University, 5100 de Maisonneuve Blvd. West, Montreal, QC, H4A 3T2, Canada
| | - Vahé Nerguizian
- Department of Electrical Engineering, École de Technologie supérieure, 1100 Notre Dame West, Montreal, QC, H3C 1K3, Canada
| | - Julia V Burnier
- Cancer Research Program, Research Institute of the McGill University Health Centre, 1001 Decarie Blvd, Montreal, QC, H4A 3J1, Canada.
- Department of Pathology, McGill University, Quebec, Canada.
| |
Collapse
|
2
|
Askarizadeh A, Vahdat-Lasemi F, Karav S, Kesharwani P, Sahebkar A. Lipid nanoparticle-based delivery of small interfering RNAs: New possibilities in the treatment of diverse diseases. Eur Polym J 2025; 223:113624. [DOI: 10.1016/j.eurpolymj.2024.113624] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
|
3
|
S PR, Banerjee R, Drummond CJ, Conn CE. Permanently Charged Cationic Lipids-Evolution from Excipients to Therapeutic Lipids. SMALL SCIENCE 2024; 4:2300270. [PMID: 40212121 PMCID: PMC11935225 DOI: 10.1002/smsc.202300270] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2023] [Revised: 04/28/2024] [Indexed: 04/13/2025] Open
Abstract
Cationic lipids are crucial in medical and biotechnological applications including cellular transfection and gene delivery. Ionizable cationic lipids are critical components of the mRNA-based COVID vaccines while permanently charged cationic lipids have shown promise in cancer treatment. Despite significant research progress over the past few decades in designing improved, biocompatible cationic lipids, their transfection efficiency remains lower than that of viral vectors. Cationic lipids with additional functionalities like fusogenicity, stimuli-responsiveness, targeting capabilities, and therapeutic activity have been engineered to improve their performance. This review highlights the importance of molecular hybridization toward the design of biocompatible cationic lipids having fusogenic, stimuli-responsive, targeting, or therapeutic properties. This review mainly focuses on cationic lipids, having a permanent positive charge in the headgroup region, as these are typically employed to both increase cellular interactions and for improved loading, particularly for anionic nucleic acid-based therapeutics and vaccines. Structure-activity relationships between the lipid chemical structure (headgroup, spacer, hydrocarbon chain) and, to a lesser extent, the self-assembled nanostructure and the intrinsic biological activity of the multi-functional cationic lipids are described. Finally, the challenges involved in developing smart lipids without affecting their inherent capacity to self-assemble into structured nano-carriers are discussed.
Collapse
Affiliation(s)
- Pushpa Ragini S
- Academy of Scientific and Innovation Research (AcSIR)Ghaziabad201002India
- Department of Oils, Lipid Science and TechnologyCSIR‐Indian Institute of Chemical TechnologyHyderabad500 007India
- School of ScienceSTEM CollegeRMIT University124 La Trobe StreetMelbourneVIC3000Australia
| | - Rajkumar Banerjee
- Academy of Scientific and Innovation Research (AcSIR)Ghaziabad201002India
- Department of Oils, Lipid Science and TechnologyCSIR‐Indian Institute of Chemical TechnologyHyderabad500 007India
| | - Calum J. Drummond
- School of ScienceSTEM CollegeRMIT University124 La Trobe StreetMelbourneVIC3000Australia
| | - Charlotte E. Conn
- School of ScienceSTEM CollegeRMIT University124 La Trobe StreetMelbourneVIC3000Australia
| |
Collapse
|
4
|
Sun W, Han C, Ge R, Jiang X, Wang Y, Han Y, Wang N, Song Y, Yang M, Chen G, Deng Y. Sialic Acid Conjugate-Modified Cationic Liposomal Paclitaxel for Targeted Therapy of Lung Metastasis in Breast Cancer: What a Difference the Cation Content Makes. Mol Pharm 2024; 21:1625-1638. [PMID: 38403951 DOI: 10.1021/acs.molpharmaceut.3c00767] [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: 02/27/2024]
Abstract
Cationic lipids play a pivotal role in developing novel drug delivery systems for diverse biomedical applications, owing to the success of mRNA vaccines against COVID-19 and the Phase III antitumor agent EndoTAG-1. However, the therapeutic potential of these positively charged liposomes is limited by dose-dependent toxicity. While an increased content of cationic lipids in the formulation can enhance the uptake and cytotoxicity toward tumor-associated cells, it is crucial to balance these advantages with the associated toxic side effects. In this work, we synthesized the cationic lipid HC-Y-2 and incorporated it into sialic acid (SA)-modified cationic liposomes loaded with paclitaxel to target tumor-associated immune cells efficiently. The SA-modified cationic liposomes exhibited enhanced binding affinity toward both RAW264.7 cells and 4T1 tumor cells in vitro due to the increased ratios of cationic HC-Y-2 content while effectively inhibiting 4T1 cell lung metastasis in vivo. By leveraging electrostatic forces and ligand-receptor interactions, the SA-modified cationic liposomes specifically target malignant tumor-associated immune cells such as tumor-associated macrophages (TAMs), reduce the proportion of cationic lipids in the formulation, and achieve dual objectives: high cellular uptake and potent antitumor efficacy. These findings highlight the potential advantages of this innovative approach utilizing cationic liposomes.
Collapse
Affiliation(s)
- Wenliang Sun
- College of Pharmacy, Shenyang Pharmaceutical University, Shenyang 110016, China
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang l10016, China
| | - Chao Han
- Key Laboratory of Structure-Based Drug Design & Discovery of Ministry of Education, School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Ruirui Ge
- College of Pharmacy, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Xiaotong Jiang
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang l10016, China
| | - Yu Wang
- College of Pharmacy, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Yingchao Han
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang l10016, China
| | - Ning Wang
- College of Pharmacy, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Yanzhi Song
- College of Pharmacy, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Mingshi Yang
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang l10016, China
- Department of Pharmacy, Faculty of Health and Medical Sciences, University of Copenhagen, Universitetsparken 2, DK-2100 Copenhagen, Denmark
| | - Guoliang Chen
- Key Laboratory of Structure-Based Drug Design & Discovery of Ministry of Education, School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Yihui Deng
- College of Pharmacy, Shenyang Pharmaceutical University, Shenyang 110016, China
| |
Collapse
|
5
|
Moreira L, Guimarães NM, Santos RS, Loureiro JA, Pereira MC, Azevedo NF. Promising strategies employing nucleic acids as antimicrobial drugs. MOLECULAR THERAPY. NUCLEIC ACIDS 2024; 35:102122. [PMID: 38333674 PMCID: PMC10850860 DOI: 10.1016/j.omtn.2024.102122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/10/2024]
Abstract
Antimicrobial resistance (AMR) is a growing concern because it causes microorganisms to develop resistance to drugs commonly used to treat infections. This results in increased difficulty in treating infections, leading to higher mortality rates and significant economic effects. Investing in new antimicrobial agents is, therefore, necessary to prevent and control AMR. Antimicrobial nucleic acids have arisen as potential key players in novel therapies for AMR infections. They have been designed to serve as antimicrobials and to act as adjuvants to conventional antibiotics or to inhibit virulent mechanisms. This new category of antimicrobial drugs consists of antisense oligonucleotides and oligomers, DNAzymes, and transcription factor decoys, differing in terms of structure, target molecules, and mechanisms of action. They are synthesized using nucleic acid analogs to enhance their resistance to nucleases. Because bacterial envelopes are generally impermeable to oligonucleotides, delivery into the cytoplasm typically requires the assistance of nanocarriers, which can affect their therapeutic potency. Given that numerous factors contribute to the success of these antimicrobial drugs, this review aims to provide a summary of the key advancements in the use of oligonucleotides for treating bacterial infections. Their mechanisms of action and the impact of factors such as nucleic acid design, target sequence, and nanocarriers on the antimicrobial potency are discussed.
Collapse
Affiliation(s)
- Luís Moreira
- LEPABE–Laboratory for Process Engineering, Environment, Biotechnology, and Energy, Faculty of Engineering, University of Porto, Rua Dr. Roberto Frias, 4200-465 Porto, Portugal
- ALiCE–Associate Laboratory in Chemical Engineering, Faculty of Engineering, University of Porto, Rua Dr. Roberto Frias, 4200-465 Porto, Portugal
| | - Nuno M. Guimarães
- LEPABE–Laboratory for Process Engineering, Environment, Biotechnology, and Energy, Faculty of Engineering, University of Porto, Rua Dr. Roberto Frias, 4200-465 Porto, Portugal
- ALiCE–Associate Laboratory in Chemical Engineering, Faculty of Engineering, University of Porto, Rua Dr. Roberto Frias, 4200-465 Porto, Portugal
| | - Rita S. Santos
- LEPABE–Laboratory for Process Engineering, Environment, Biotechnology, and Energy, Faculty of Engineering, University of Porto, Rua Dr. Roberto Frias, 4200-465 Porto, Portugal
- ALiCE–Associate Laboratory in Chemical Engineering, Faculty of Engineering, University of Porto, Rua Dr. Roberto Frias, 4200-465 Porto, Portugal
| | - Joana A. Loureiro
- LEPABE–Laboratory for Process Engineering, Environment, Biotechnology, and Energy, Faculty of Engineering, University of Porto, Rua Dr. Roberto Frias, 4200-465 Porto, Portugal
- ALiCE–Associate Laboratory in Chemical Engineering, Faculty of Engineering, University of Porto, Rua Dr. Roberto Frias, 4200-465 Porto, Portugal
| | - Maria C. Pereira
- LEPABE–Laboratory for Process Engineering, Environment, Biotechnology, and Energy, Faculty of Engineering, University of Porto, Rua Dr. Roberto Frias, 4200-465 Porto, Portugal
- ALiCE–Associate Laboratory in Chemical Engineering, Faculty of Engineering, University of Porto, Rua Dr. Roberto Frias, 4200-465 Porto, Portugal
| | - Nuno F. Azevedo
- LEPABE–Laboratory for Process Engineering, Environment, Biotechnology, and Energy, Faculty of Engineering, University of Porto, Rua Dr. Roberto Frias, 4200-465 Porto, Portugal
- ALiCE–Associate Laboratory in Chemical Engineering, Faculty of Engineering, University of Porto, Rua Dr. Roberto Frias, 4200-465 Porto, Portugal
| |
Collapse
|
6
|
Chen JW, Liew FF, Tan HW, Misran M, Chung I. Cholesterol-linoleic acid liposomes induced extracellular vesicles secretion from immortalized adipose-derived mesenchymal stem cells for in vitro cell migration. ARTIFICIAL CELLS, NANOMEDICINE, AND BIOTECHNOLOGY 2023; 51:346-360. [PMID: 37524112 DOI: 10.1080/21691401.2023.2237534] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/06/2023] [Revised: 06/28/2023] [Accepted: 07/12/2023] [Indexed: 08/02/2023]
Abstract
Extracellular vesicles (EVs) are small vesicles that are naturally released by cells and play a crucial role in cell-to-cell communication, tissue repair and regeneration. As naturally secreted EVs are limited, liposomes with different physicochemical properties, such as 1,2-dioleoyl-3-trimethylammonium propane (DOTAP) and linoleic acid (LA) with modifications have been formulated to improve EVs secretion for in vitro wound healing. Various analyses, including dynamic light scattering (DLS) and transmission electron microscopy (TEM) were performed to monitor the successful preparation of different types of liposomes. The results showed that cholesterol-LA liposomes significantly improved the secretion of EVs from immortalized adipose-derived mesenchymal stem cells (AD-MSCs) by 1.5-fold. Based on the cell migration effects obtained from scratch assay, both LA liposomal-induced EVs and cholesterol-LA liposomal-induced EVs significantly enhanced the migration of human keratinocytes (HaCaT) cell line. These findings suggested that LA and cholesterol-LA liposomes that enhance EVs secretion are potentially useful and can be extended for various tissue regeneration applications.
Collapse
Affiliation(s)
- Jzit Weii Chen
- Department of Pharmacology, Faculty of Medicine, Universiti Malaya, Kuala Lumpur, Malaysia
| | - Fong Fong Liew
- Department of Oral Biology and Biomedical Science, Faculty of Dentistry, MAHSA University, Selangor, Malaysia
| | - Hsiao Wei Tan
- Institute of Research Management and Services, Research and Innovation Management Complex, Universiti Malaya, Kuala Lumpur, Malaysia
| | - Misni Misran
- Department of Chemistry, Faculty of Science, Universiti Malaya, Kuala Lumpur, Malaysia
| | - Ivy Chung
- Department of Pharmacology, Faculty of Medicine, Universiti Malaya, Kuala Lumpur, Malaysia
| |
Collapse
|
7
|
Sanap SN, Bisen AC, Agrawal S, Kedar A, Bhatta RS. Ophthalmic nano-bioconjugates: critical challenges and technological advances. Ther Deliv 2023; 14:419-441. [PMID: 37535389 DOI: 10.4155/tde-2023-0031] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/04/2023] Open
Abstract
Ophthalmic disease can cause permanent loss of vision and blindness. Easy-to-administer topical and systemic treatments are preferred for treating sight-threatening disorders. Typical ocular anatomy makes topical and systemic ophthalmic drug delivery challenging. Various novel nano-drug delivery approaches are developed to attain the desired bioavailability in the eye by increasing residence time and improved permeability across the cornea. The review focuses on novel methods that are biocompatible, safe and highly therapeutic. Novelty in nanocarrier design and modification can overcome their drawbacks and make them potential drug carriers for eye disorders in both the anterior and posterior eye segments. This review briefly discussed technologies, patented developments, and clinical trial data to support nanocarriers' use in ocular drug delivery.
Collapse
Affiliation(s)
- Sachin Nashik Sanap
- Pharmaceutics & Pharmacokinetics Division, CSIR-Central Drug Research Institute, Lucknow, 226031, India
- Academy of Scientific & Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Amol Chhatrapati Bisen
- Pharmaceutics & Pharmacokinetics Division, CSIR-Central Drug Research Institute, Lucknow, 226031, India
- Academy of Scientific & Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Sristi Agrawal
- Pharmaceutics & Pharmacokinetics Division, CSIR-Central Drug Research Institute, Lucknow, 226031, India
- Academy of Scientific & Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Ashwini Kedar
- Pharmaceutics & Pharmacokinetics Division, CSIR-Central Drug Research Institute, Lucknow, 226031, India
| | - Rabi Sankar Bhatta
- Pharmaceutics & Pharmacokinetics Division, CSIR-Central Drug Research Institute, Lucknow, 226031, India
- Academy of Scientific & Innovative Research (AcSIR), Ghaziabad, 201002, India
| |
Collapse
|
8
|
Hoffmann M, Gerlach S, Takamiya M, Tarazi S, Hersch N, Csiszár A, Springer R, Dreissen G, Scharr H, Rastegar S, Beil T, Strähle U, Merkel R, Hoffmann B. Smuggling on the Nanoscale-Fusogenic Liposomes Enable Efficient RNA-Transfer with Negligible Immune Response In Vitro and In Vivo. Pharmaceutics 2023; 15:pharmaceutics15041210. [PMID: 37111695 PMCID: PMC10146161 DOI: 10.3390/pharmaceutics15041210] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 04/04/2023] [Accepted: 04/05/2023] [Indexed: 04/29/2023] Open
Abstract
The efficient and biocompatible transfer of nucleic acids into mammalian cells for research applications or medical purposes is a long-standing, challenging task. Viral transduction is the most efficient transfer system, but often entails high safety levels for research and potential health impairments for patients in medical applications. Lipo- or polyplexes are commonly used transfer systems but result in comparably low transfer efficiencies. Moreover, inflammatory responses caused by cytotoxic side effects were reported for these transfer methods. Often accountable for these effects are various recognition mechanisms for transferred nucleic acids. Using commercially available fusogenic liposomes (Fuse-It-mRNA), we established highly efficient and fully biocompatible transfer of RNA molecules for in vitro as well as in vivo applications. We demonstrated bypassing of endosomal uptake routes and, therefore, of pattern recognition receptors that recognize nucleic acids with high efficiency. This may underlie the observed almost complete abolishment of inflammatory cytokine responses. RNA transfer experiments into zebrafish embryos and adult animals fully confirmed the functional mechanism and the wide range of applications from single cells to organisms.
Collapse
Affiliation(s)
- Marco Hoffmann
- IBI-2: Mechanobiology, Institute of Biological Information Processing, Forschungszentrum Jülich, 52428 Jülich, Germany
| | - Sven Gerlach
- IBI-2: Mechanobiology, Institute of Biological Information Processing, Forschungszentrum Jülich, 52428 Jülich, Germany
| | - Masanari Takamiya
- Institute of Biological and Chemical Systems-Biological Information Processing (IBCS-BIP), Karlsruhe Institute of Technology (KIT), Postfach 3640, 76021 Karlsruhe, Germany
| | - Samar Tarazi
- IBI-2: Mechanobiology, Institute of Biological Information Processing, Forschungszentrum Jülich, 52428 Jülich, Germany
| | - Nils Hersch
- IBI-2: Mechanobiology, Institute of Biological Information Processing, Forschungszentrum Jülich, 52428 Jülich, Germany
| | - Agnes Csiszár
- IBI-2: Mechanobiology, Institute of Biological Information Processing, Forschungszentrum Jülich, 52428 Jülich, Germany
| | - Ronald Springer
- IBI-2: Mechanobiology, Institute of Biological Information Processing, Forschungszentrum Jülich, 52428 Jülich, Germany
| | - Georg Dreissen
- IBI-2: Mechanobiology, Institute of Biological Information Processing, Forschungszentrum Jülich, 52428 Jülich, Germany
| | - Hanno Scharr
- IAS-8: Data Analytics and Machine Learning, Institute for Advanced Simulation, Forschungszentrum Jülich, 52428 Jülich, Germany
| | - Sepand Rastegar
- Institute of Biological and Chemical Systems-Biological Information Processing (IBCS-BIP), Karlsruhe Institute of Technology (KIT), Postfach 3640, 76021 Karlsruhe, Germany
| | - Tanja Beil
- Institute of Biological and Chemical Systems-Biological Information Processing (IBCS-BIP), Karlsruhe Institute of Technology (KIT), Postfach 3640, 76021 Karlsruhe, Germany
| | - Uwe Strähle
- Institute of Biological and Chemical Systems-Biological Information Processing (IBCS-BIP), Karlsruhe Institute of Technology (KIT), Postfach 3640, 76021 Karlsruhe, Germany
| | - Rudolf Merkel
- IBI-2: Mechanobiology, Institute of Biological Information Processing, Forschungszentrum Jülich, 52428 Jülich, Germany
| | - Bernd Hoffmann
- IBI-2: Mechanobiology, Institute of Biological Information Processing, Forschungszentrum Jülich, 52428 Jülich, Germany
| |
Collapse
|
9
|
Andrade S, Loureiro JA, Ramirez S, Catumbela CSG, Soto C, Morales R, Pereira MC. Multi-Dose Intravenous Administration of Neutral and Cationic Liposomes in Mice: An Extensive Toxicity Study. Pharmaceuticals (Basel) 2022; 15:761. [PMID: 35745680 PMCID: PMC9229811 DOI: 10.3390/ph15060761] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Revised: 06/13/2022] [Accepted: 06/17/2022] [Indexed: 02/04/2023] Open
Abstract
Liposomes are widely used as delivery systems for therapeutic purposes. However, the toxicity associated with the multi-dose administration of these nanoparticles is not fully elucidated. Here, we evaluated the toxicity of the prolonged administration of liposomes composed of neutral or cationic phospholipids often used in drug and gene delivery. For that purpose, adult wild-type mice (C57Bl6) were randomly distributed into three groups receiving either vehicle (PBS), neutral, or cationic liposomes and subjected to repeated intravenous injections for a total of 10 doses administered over 3 weeks. Several parameters, including mortality, body weight, and glucose levels, were monitored throughout the trial. While these variables did not change in the group treated with neutral liposomes, the group treated with the positively charged liposomes displayed a mortality rate of 45% after 10 doses of administration. Additional urinalysis, blood tests, and behavioral assays to evaluate impairments of motor functions or lesions in major organs were also performed. The cationic group showed less forelimb peak force than the control group, alterations at the hematological level, and inflammatory components, unlike the neutral group. Overall, the results demonstrate that cationic liposomes are toxic for multi-dose administration, while the neutral liposomes did not induce changes associated with toxicity. Therefore, our results support the use of the well-known neutral liposomes as safe drug shuttles, even when repetitive administrations are needed.
Collapse
Affiliation(s)
- Stéphanie Andrade
- LEPABE, Department of Chemical Engineering, Faculty of Engineering, University of Porto, 4200-465 Porto, Portugal; (S.A.); (J.A.L.)
- ALiCE—Associate Laboratory in Chemical Engineering, Faculty of Engineering, University of Porto, Rua Dr. Roberto Frias, 4200-465 Porto, Portugal
| | - Joana A. Loureiro
- LEPABE, Department of Chemical Engineering, Faculty of Engineering, University of Porto, 4200-465 Porto, Portugal; (S.A.); (J.A.L.)
- ALiCE—Associate Laboratory in Chemical Engineering, Faculty of Engineering, University of Porto, Rua Dr. Roberto Frias, 4200-465 Porto, Portugal
| | - Santiago Ramirez
- Department of Neurology, The University of Texas Health Science Center at Houston, 6431 Fannin St., Houston, TX 77030, USA; (S.R.); (C.S.G.C.); (C.S.)
| | - Celso S. G. Catumbela
- Department of Neurology, The University of Texas Health Science Center at Houston, 6431 Fannin St., Houston, TX 77030, USA; (S.R.); (C.S.G.C.); (C.S.)
| | - Claudio Soto
- Department of Neurology, The University of Texas Health Science Center at Houston, 6431 Fannin St., Houston, TX 77030, USA; (S.R.); (C.S.G.C.); (C.S.)
| | - Rodrigo Morales
- Department of Neurology, The University of Texas Health Science Center at Houston, 6431 Fannin St., Houston, TX 77030, USA; (S.R.); (C.S.G.C.); (C.S.)
- Centro Integrativo de Biologia y Quimica Aplicada (CIBQA), Universidad Bernardo O’Higgins, Santiago 1497, Chile
| | - Maria Carmo Pereira
- LEPABE, Department of Chemical Engineering, Faculty of Engineering, University of Porto, 4200-465 Porto, Portugal; (S.A.); (J.A.L.)
- ALiCE—Associate Laboratory in Chemical Engineering, Faculty of Engineering, University of Porto, Rua Dr. Roberto Frias, 4200-465 Porto, Portugal
| |
Collapse
|
10
|
Dravid AA, M. Dhanabalan K, Agarwal S, Agarwal R. Resolvin D1-loaded nanoliposomes promote M2 macrophage polarization and are effective in the treatment of osteoarthritis. Bioeng Transl Med 2022; 7:e10281. [PMID: 35600665 PMCID: PMC9115708 DOI: 10.1002/btm2.10281] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2021] [Revised: 09/19/2021] [Accepted: 09/22/2021] [Indexed: 11/24/2022] Open
Abstract
Current treatments for osteoarthritis (OA) offer symptomatic relief but do not prevent or halt the disease progression. Chronic low-grade inflammation is considered a significant driver of OA. Specialized proresolution mediators are powerful agents of resolution but have a short in vivo half-life. In this study, we have engineered a Resolvin D1 (RvD1)-loaded nanoliposomal formulation (Lipo-RvD1) that targets and resolves the OA-associated inflammation. This formulation creates a depot of the RvD1 molecules that allows the controlled release of the molecule for up to 11 days in vitro. In surgically induced mice model of OA, only controlled-release formulation of Lipo-RvD1 was able to treat the progressing cartilage damage when administered a month after the surgery, while the free drug was unable to prevent cartilage damage. We found that Lipo-RvD1 functions by damping the proinflammatory activity of synovial macrophages and recruiting a higher number of M2 macrophages at the site of inflammation. Our Lipo-RvD1 formulation was able to target and suppress the formation of the osteophytes and showed analgesic effect, thus emphasizing its ability to treat clinical symptoms of OA. Such controlled-release formulation of RvD1 could represent a patient-compliant treatment for OA.
Collapse
Affiliation(s)
- Ameya A. Dravid
- BioSystems Science and EngineeringIndian Institute of ScienceBangaloreKarnatakaIndia
| | - Kaamini M. Dhanabalan
- BioSystems Science and EngineeringIndian Institute of ScienceBangaloreKarnatakaIndia
| | - Smriti Agarwal
- BioSystems Science and EngineeringIndian Institute of ScienceBangaloreKarnatakaIndia
| | - Rachit Agarwal
- BioSystems Science and EngineeringIndian Institute of ScienceBangaloreKarnatakaIndia
| |
Collapse
|
11
|
Development of Theranostic Cationic Liposomes Designed for Image-Guided Delivery of Nucleic Acid. Pharmaceutics 2020; 12:pharmaceutics12090854. [PMID: 32911863 PMCID: PMC7559777 DOI: 10.3390/pharmaceutics12090854] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Revised: 08/21/2020] [Accepted: 09/01/2020] [Indexed: 02/01/2023] Open
Abstract
Cationic liposomes have been considered as potential vectors for gene delivery thanks to their ability to transfect cells with high efficiency. Recently, the combination of diagnostic agent and therapeutic agents in the same particle to form a theranostic system has been reported. Magnetic liposomes are one of these examples. Due to the magnetic nanoparticles encapsulated in the liposomes, they can act as a drug delivery system and, at the same time, a magnetic resonance imaging contrast enhancement agent or hyperthermia. In this work, nucleic acid delivery systems based on magnetic cationic liposomes (MCLs) were developed. Two different techniques, reverse phase evaporation and cosolvent sonication, were employed for liposome preparation. Both strategies produced MCLs of less than 200 nm with highly positive charge. Enhancement of their transverse and longitudinal relaxivities r2 and r1 was obtained with both kinds of magnetic liposomes compared to free magnetic nanoparticles. Moreover, these MCLs showed high capacity to form complexes and transfect CT-26 cells using the antibiotic-free pFAR4-luc plasmid. The transfection enhancement with magnetofection was also carried out in CT26 cells. These results suggested that our MCLs could be a promising candidate for image-guided gene therapy.
Collapse
|
12
|
Karabasz A, Szuwarzyński M, Nowakowska M, Bzowska M, Lewandowska-Łańcucka J. Stabilization of liposomes with silicone layer improves their elastomechanical properties while not compromising biological features. Colloids Surf B Biointerfaces 2020; 195:111272. [PMID: 32791473 DOI: 10.1016/j.colsurfb.2020.111272] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2020] [Revised: 07/20/2020] [Accepted: 07/21/2020] [Indexed: 11/28/2022]
Abstract
The liposomes are among the most promising types of drug delivery systems but low stability significantly limits their application. Some approaches proposed to overcome this drawback may affect the liposomes toxicity profile. It is assumed that developed by us and presented here stabilization method involving formation of silicone network within the liposomal bilayer will improve elastomechanical properties of vesicles while not deteriorating their biocompatibility. The silicone-stabilized liposomes were prepared by base-catalyzed polycondensation process of the 1,3,5,7-tetramethylcyclotetrasiloxane (D4H) within the liposomal bilayer. The systematic biological in vitro studies of vesicles obtained were carried out. Moreover, the elastomechanical features investigation employing atomic force microscopy (AFM) measurements was performed. These properties of the liposome membrane are of great importance since they define the nanocarriers' stability as well as play a significant role in their cellular uptake via endocytosis. Applying the Derjaguin-Muller-Toporov (DMT) model, the elastic modulus of the silicone-stabilized liposomes was determined and compared to that characteristic for the pristine liposomes. The in vitro biological evaluation of silicone-stabilized liposomes demonstrated that these vesicles are not toxic for blood cells isolated from healthy donors and they do not induce oxidative stress in HepG2 cells. AFM results confirmed the stabilizing effect of silicone and revealed that the silicone network improves the elastomechanical properties of the resulted liposomes. This is the first report demonstrating that the silicone-stabilized liposomes retain biocompatibility of pristine liposomes' while acquire significantly better elastomechanical features.
Collapse
Affiliation(s)
- Alicja Karabasz
- Department of Cell Biochemistry, Faculty of Biochemistry, Biophysics and Biotechnology Jagiellonian University, Gronostajowa 7, 30-387 Kraków, Poland
| | - Michał Szuwarzyński
- AGH University of Science and Technology, Academic Centre for Materials and Nanotechnology, Al. Mickiewicza 30, 30-059 Krakow, Poland
| | - Maria Nowakowska
- Department of Physical Chemistry, Faculty of Chemistry, Jagiellonian University, Gronostajowa 2, 30-387 Kraków, Poland
| | - Monika Bzowska
- Department of Cell Biochemistry, Faculty of Biochemistry, Biophysics and Biotechnology Jagiellonian University, Gronostajowa 7, 30-387 Kraków, Poland.
| | - Joanna Lewandowska-Łańcucka
- Department of Physical Chemistry, Faculty of Chemistry, Jagiellonian University, Gronostajowa 2, 30-387 Kraków, Poland.
| |
Collapse
|
13
|
Trigueros S, Domènech EB, Toulis V, Marfany G. In Vitro Gene Delivery in Retinal Pigment Epithelium Cells by Plasmid DNA-Wrapped Gold Nanoparticles. Genes (Basel) 2019; 10:genes10040289. [PMID: 30970664 PMCID: PMC6523520 DOI: 10.3390/genes10040289] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2019] [Revised: 04/05/2019] [Accepted: 04/08/2019] [Indexed: 01/20/2023] Open
Abstract
Many rare diseases course with affectation of neurosensory organs. Among them, the neuroepithelial retina is very vulnerable due to constant light/oxidative stress, but it is also the most accessible and amenable to gene manipulation. Currently, gene addition therapies targeting retinal tissue (either photoreceptors or the retinal pigment epithelium), as a therapy for inherited retinal dystrophies, use adeno-associated virus (AAV)-based approaches. However, efficiency and safety of therapeutic strategies are relevant issues that are not always resolved in virus-based gene delivery and alternative methodologies should be explored. Based on our experience, we are currently assessing the novel physical properties at the nanoscale of inorganic gold nanoparticles for delivering genes to the retinal pigment epithelium (RPE) as a safe and efficient alternative approach. In this work, we present our preliminary results using DNA-wrapped gold nanoparticles (DNA-gold NPs) for successful in vitro gene delivery on human retinal pigment epithelium cell cultures, as a proof-of-principle to assess its feasibility for retina in vivo gene delivery. Our results show faster expression of a reporter gene in cells transfected with DNA-gold NPs compared to DNA-liposome complexes. Furthermore, we show that the DNA-gold NPs follow different uptake, internalization and intracellular vesicle trafficking routes compared to pristine NPs.
Collapse
Affiliation(s)
- Sònia Trigueros
- Department of Zoology, University of Oxford, Oxford, OX1 3PS, UK.
| | - Elena B Domènech
- Departament de Genètica, Microbiologia i Estadística, Universitat de Barcelona, 08028 Barcelona, Spain.
- CIBERER, ISCIII, Universitat de Barcelona, 08028 Barcelona, Spain.
| | - Vasileios Toulis
- Departament de Genètica, Microbiologia i Estadística, Universitat de Barcelona, 08028 Barcelona, Spain.
- CIBERER, ISCIII, Universitat de Barcelona, 08028 Barcelona, Spain.
| | - Gemma Marfany
- Departament de Genètica, Microbiologia i Estadística, Universitat de Barcelona, 08028 Barcelona, Spain.
- CIBERER, ISCIII, Universitat de Barcelona, 08028 Barcelona, Spain.
- Institute of Biomedicine (IBUB-IRSJD), Universitat de Barcelona, Barcelona, Spain.
| |
Collapse
|
14
|
Yang B, Mao J, Jiang S, Wei J, Li Y, Gao B, Lu X. Cholesterol depletion induced by RNA interference targeting DHCR24 protects cells from liposome-induced cytotoxicity. Prep Biochem Biotechnol 2019; 49:453-458. [PMID: 30896287 DOI: 10.1080/10826068.2019.1591979] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Existing evidence has demonstrated liposomes as the gene transporter induce the cytotoxicity during the transfection process through several known pathways. In the present study, we investigated the possibility of siRNAs targeting 3-β-hydroxysterol △-24-reductase (DHCR24), which encodes an enzyme catalyzing the last step of cholesterol biosynthesis, to suppress the liposome cytotoxicity induced by lipid-based transfection reagent in the neuroblastoma cell line N2A. We found that the siRNAs targeting DHCR24 mRNA protect cells from the liposome-induced cell death, probably through the effect of siDHCR24s on the reduction of the cellular cholesterol and decrease in the generation of reactive oxygen species (ROS). This suggests that siRNAs targeting DHCR24 or other methods that reduce the intracellular cholesterol levels might be a good strategy for avoiding the cytotoxicity of liposomes, without impairing its efficiency of gene-delivering.
Collapse
Affiliation(s)
- Baoyu Yang
- a School of Life Science , Liaoning University , Shenyang , China
| | - Jing Mao
- a School of Life Science , Liaoning University , Shenyang , China
| | - Shan Jiang
- a School of Life Science , Liaoning University , Shenyang , China
| | - Jie Wei
- a School of Life Science , Liaoning University , Shenyang , China
| | - Yang Li
- b Institute of Basic Medical Sciences , Shenyang Medical College , Shenyang , China
| | - Bing Gao
- b Institute of Basic Medical Sciences , Shenyang Medical College , Shenyang , China
| | - Xiuli Lu
- a School of Life Science , Liaoning University , Shenyang , China
| |
Collapse
|