1
|
Gao Y, Zhang L, Wang Z, Bai H, Wu C, Shuai Q, Yan Y. Enhanced Pulmonary and Splenic mRNA Delivery Using DOTAP-Incorporated Poly(β-Amino Ester)-Lipid Nanoparticles. Biomacromolecules 2025. [PMID: 39746921 DOI: 10.1021/acs.biomac.4c01445] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
Abstract
mRNA-based therapies hold tremendous promise for treating various diseases, yet their clinical success is hindered by delivery challenges. This study developed a library of 140 lipocationic Poly(β-amino ester)s (PBAEs) and formulated lipid-polymer hybrid nanoparticles (LPHs) with four helper lipids, including 1,2-dioleoyl-3-trimethylammonium-propane (DOTAP), to enhance mRNA delivery. Initial in vitro screening of four representative PBAEs identified the D/P4-1 formulation (DOTAP/PBAE molar ratio of 4:1) as the most effective. Further screening of the library using this formulation identified eight top-performing LPHs. In vivo experiments confirmed high luciferase expression in the spleen and lungs of mice following intravenous administration of Luc mRNA-loaded LPHs. Detailed analysis revealed that DOTAP incorporation influenced LPH properties, including apparent pKa, surface charge, and internal hydrophobicity, enabling enhanced mRNA release and cellular uptake. This study demonstrates potent approaches to modulate PBAE-lipid nanoparticle properties by altering PBAE structures and nanoparticle composition, offering insights for designing effective hybrid carriers for mRNA therapeutics.
Collapse
Affiliation(s)
- Yuduo Gao
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou, Zhejiang 310014, China
| | - Luwei Zhang
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou, Zhejiang 310014, China
| | - Ziyue Wang
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou, Zhejiang 310014, China
| | - Hao Bai
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou, Zhejiang 310014, China
| | - Chengfan Wu
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou, Zhejiang 310014, China
| | - Qi Shuai
- Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, Zhejiang University of Technology, Hangzhou, Zhejiang 310014, China
| | - Yunfeng Yan
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou, Zhejiang 310014, China
| |
Collapse
|
2
|
Li H, Zhu Y, Wang X, Feng Y, Qian Y, Ma Q, Li X, Chen Y, Chen K. Joining Forces: The Combined Application of Therapeutic Viruses and Nanomaterials in Cancer Therapy. Molecules 2023; 28:7679. [PMID: 38005401 PMCID: PMC10674375 DOI: 10.3390/molecules28227679] [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: 09/29/2023] [Revised: 11/10/2023] [Accepted: 11/17/2023] [Indexed: 11/26/2023] Open
Abstract
Cancer, on a global scale, presents a monumental challenge to our healthcare systems, posing a significant threat to human health. Despite the considerable progress we have made in the diagnosis and treatment of cancer, realizing precision cancer therapy, reducing side effects, and enhancing efficacy remain daunting tasks. Fortunately, the emergence of therapeutic viruses and nanomaterials provides new possibilities for tackling these issues. Therapeutic viruses possess the ability to accurately locate and attack tumor cells, while nanomaterials serve as efficient drug carriers, delivering medication precisely to tumor tissues. The synergy of these two elements has led to a novel approach to cancer treatment-the combination of therapeutic viruses and nanomaterials. This advantageous combination has overcome the limitations associated with the side effects of oncolytic viruses and the insufficient tumoricidal capacity of nanomedicines, enabling the oncolytic viruses to more effectively breach the tumor's immune barrier. It focuses on the lesion site and even allows for real-time monitoring of the distribution of therapeutic viruses and drug release, achieving a synergistic effect. This article comprehensively explores the application of therapeutic viruses and nanomaterials in tumor treatment, dissecting their working mechanisms, and integrating the latest scientific advancements to predict future development trends. This approach, which combines viral therapy with the application of nanomaterials, represents an innovative and more effective treatment strategy, offering new perspectives in the field of tumor therapy.
Collapse
Affiliation(s)
- Hongyu Li
- Shulan International Medical College, Zhejiang Shuren University, Hangzhou 310015, China; (Y.Z.); (Y.F.); (Y.Q.); (Q.M.); (X.L.); (Y.C.)
- Ocean College, Beibu Gulf University, Qinzhou 535011, China
| | - Yunhuan Zhu
- Shulan International Medical College, Zhejiang Shuren University, Hangzhou 310015, China; (Y.Z.); (Y.F.); (Y.Q.); (Q.M.); (X.L.); (Y.C.)
| | - Xin Wang
- Center of Infectious Disease Research, School of Life Science, Westlake University, Hangzhou 310024, China;
| | - Yilu Feng
- Shulan International Medical College, Zhejiang Shuren University, Hangzhou 310015, China; (Y.Z.); (Y.F.); (Y.Q.); (Q.M.); (X.L.); (Y.C.)
| | - Yuncheng Qian
- Shulan International Medical College, Zhejiang Shuren University, Hangzhou 310015, China; (Y.Z.); (Y.F.); (Y.Q.); (Q.M.); (X.L.); (Y.C.)
| | - Qiman Ma
- Shulan International Medical College, Zhejiang Shuren University, Hangzhou 310015, China; (Y.Z.); (Y.F.); (Y.Q.); (Q.M.); (X.L.); (Y.C.)
| | - Xinyuan Li
- Shulan International Medical College, Zhejiang Shuren University, Hangzhou 310015, China; (Y.Z.); (Y.F.); (Y.Q.); (Q.M.); (X.L.); (Y.C.)
| | - Yihan Chen
- Shulan International Medical College, Zhejiang Shuren University, Hangzhou 310015, China; (Y.Z.); (Y.F.); (Y.Q.); (Q.M.); (X.L.); (Y.C.)
| | - Keda Chen
- Shulan International Medical College, Zhejiang Shuren University, Hangzhou 310015, China; (Y.Z.); (Y.F.); (Y.Q.); (Q.M.); (X.L.); (Y.C.)
| |
Collapse
|
3
|
Huang CH, Dong T, Phung AT, Shah JR, Larson C, Sanchez AB, Blair SL, Oronsky B, Trogler WC, Reid T, Kummel AC. Full Remission of CAR-Deficient Tumors by DOTAP-Folate Liposome Encapsulation of Adenovirus. ACS Biomater Sci Eng 2022; 8:5199-5209. [PMID: 36395425 DOI: 10.1021/acsbiomaterials.2c00966] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Adenovirus (Ad)-based vectors have shown considerable promise for gene therapy. However, Ad requires the coxsackievirus and adenovirus receptor (CAR) to enter cells efficiently and low CAR expression is found in many human cancers, which hinder adenoviral gene therapies. Here, cationic 1,2-dioleoyl-3-trimethylammonium-propane (DOTAP)-folate liposomes (Df) encapsulating replication-deficient Ad were synthesized, which showed improved transfection efficiency in various CAR-deficient cell lines, including epithelial and hematopoietic cell types. When encapsulating replication-competent oncolytic Ad (TAV255) in DOTAP-folate liposome (TAV255-Df), the adenoviral structural protein, hexon, was readily produced in CAR-deficient cells, and the tumor cell killing ability was 5× higher than that of the non-encapsulated Ad. In CAR-deficient CT26 colon carcinoma murine models, replication-competent TAV255-Df treatment of subcutaneous tumors by intratumoral injection resulted in 67% full tumor remission, prolonged survival, and anti-cancer immunity when mice were rechallenged with cancer cells with no further treatment. The preclinical data shows that DOTAP-folate liposomes could significantly enhance the transfection efficiency of Ad in CAR-deficient cells and, therefore, could be a feasible strategy for applications in cancer treatment.
Collapse
Affiliation(s)
- Ching-Hsin Huang
- Moores Cancer Center, University of California San Diego, 3855 Health Sciences Drive, La Jolla, California 92037, United States
| | - Tao Dong
- Moores Cancer Center, University of California San Diego, 3855 Health Sciences Drive, La Jolla, California 92037, United States
| | - Abraham T Phung
- Moores Cancer Center, University of California San Diego, 3855 Health Sciences Drive, La Jolla, California 92037, United States
| | - Jaimin R Shah
- Moores Cancer Center, University of California San Diego, 3855 Health Sciences Drive, La Jolla, California 92037, United States
| | - Christopher Larson
- EpicentRx, Inc., 11099 North Torrey Pines Road, Suite 160, La Jolla, California 92037, United States
| | - Ana B Sanchez
- EpicentRx, Inc., 11099 North Torrey Pines Road, Suite 160, La Jolla, California 92037, United States
| | - Sarah L Blair
- Moores Cancer Center, University of California San Diego, 3855 Health Sciences Drive, La Jolla, California 92037, United States
| | - Bryan Oronsky
- EpicentRx, Inc., 11099 North Torrey Pines Road, Suite 160, La Jolla, California 92037, United States
| | - William C Trogler
- Department of Chemistry & Biochemistry, University of California San Diego, 9500 Gilman Drive, La Jolla, California 92093, United States
| | - Tony Reid
- EpicentRx, Inc., 11099 North Torrey Pines Road, Suite 160, La Jolla, California 92037, United States
| | - Andrew C Kummel
- Department of Chemistry & Biochemistry, University of California San Diego, 9500 Gilman Drive, La Jolla, California 92093, United States
| |
Collapse
|
4
|
Sun M, Dang UJ, Yuan Y, Psaras AM, Osipitan O, Brooks TA, Lu F, Di Pasqua AJ. Optimization of DOTAP/chol Cationic Lipid Nanoparticles for mRNA, pDNA, and Oligonucleotide Delivery. AAPS PharmSciTech 2022; 23:135. [PMID: 35534697 PMCID: PMC9084260 DOI: 10.1208/s12249-022-02294-w] [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: 02/23/2022] [Accepted: 04/27/2022] [Indexed: 12/27/2022] Open
Abstract
Lipid nanoparticles (LNPs) can be used as delivery vehicles for nucleic acid biotherapeutics. In fact, LNPs are currently being used in the Pfizer/BioNTech and Moderna COVID-19 vaccines. Cationic LNPs composed of 1,2-dioleoyl-3-trimethylammonium-propane (DOTAP)/cholesterol (chol) LNPs have been classified as one of the most efficient gene delivery systems and are being tested in numerous clinical trials. The objective of this study was to examine the effect of the molar ratio of DOTAP/chol, PEGylation, and lipid to mRNA ratio on mRNA transfection, and explore the applications of DOTAP/chol LNPs in pDNA and oligonucleotide transfection. Here we showed that PEGylation significantly decreased mRNA transfection efficiency of DOTAP/chol LNPs. Among non-PEGylated LNP formulations, 1:3 molar ratio of DOTAP/chol in DOTAP/chol LNPs showed the highest mRNA transfection efficiency. Furthermore, the optimal ratio of DOTAP/chol LNPs to mRNA was tested to be 62.5 µM lipid to 1 μg mRNA. More importantly, these mRNA-loaded nanoparticles were stable for 60 days at 4 °C storage without showing reduction in transfection efficacy. We further found that DOTAP/chol LNPs were able to transfect pDNA and oligonucleotides, demonstrating the ability of these LNPs to transport the cargo into the cell nucleus. The influence of various factors in the formulation of DOTAP/chol cationic LNPs is thus described and will help improve drug delivery of nucleic acid-based vaccines and therapies.
Collapse
|
5
|
Lam JH, Khan AK, Cornell TA, Chia TW, Dress RJ, Yeow WWW, Mohd-Ismail NK, Venkataraman S, Ng KT, Tan YJ, Anderson DE, Ginhoux F, Nallani M. Polymersomes as Stable Nanocarriers for a Highly Immunogenic and Durable SARS-CoV-2 Spike Protein Subunit Vaccine. ACS NANO 2021; 15:15754-15770. [PMID: 34618423 PMCID: PMC8525042 DOI: 10.1021/acsnano.1c01243] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Accepted: 09/30/2021] [Indexed: 05/05/2023]
Abstract
Multiple successful vaccines against severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) are urgently needed to address the ongoing coronavirus disease 2019 (Covid-19) pandemic. In the present work, we describe a subunit vaccine based on the SARS-CoV-2 spike protein coadministered with CpG adjuvant. To enhance the immunogenicity of our formulation, both antigen and adjuvant were encapsulated with our proprietary artificial cell membrane (ACM) polymersome technology. Structurally, ACM polymersomes are self-assembling nanoscale vesicles made up of an amphiphilic block copolymer comprising poly(butadiene)-b-poly(ethylene glycol) and a cationic lipid, 1,2-dioleoyl-3-trimethylammonium-propane. Functionally, ACM polymersomes serve as delivery vehicles that are efficiently taken up by dendritic cells (DC1 and DC2), which are key initiators of the adaptive immune response. Two doses of our formulation elicit robust neutralizing antibody titers in C57BL/6 mice that persist at least 40 days. Furthermore, we confirm the presence of functional memory CD4+ and CD8+ T cells that produce T helper type 1 cytokines. This study is an important step toward the development of an efficacious vaccine in humans.
Collapse
Affiliation(s)
| | - Amit K. Khan
- ACM
Biolabs Pte Ltd, Singapore 638075, Singapore
| | | | | | - Regine J. Dress
- Singapore Immunology
Network, Agency for Science, Technology and Research, Singapore 138648, Singapore
| | | | - Nur Khairiah Mohd-Ismail
- Infectious
Diseases Translational Research Program, Department of Microbiology
and Immunology, Yong Loo Lin School of Medicine, National University
Health System, National University of Singapore, Singapore 117545, Singapore
| | | | - Kim Tien Ng
- Infectious
Diseases Translational Research Program, Department of Microbiology
and Immunology, Yong Loo Lin School of Medicine, National University
Health System, National University of Singapore, Singapore 117545, Singapore
| | - Yee-Joo Tan
- Infectious
Diseases Translational Research Program, Department of Microbiology
and Immunology, Yong Loo Lin School of Medicine, National University
Health System, National University of Singapore, Singapore 117545, Singapore
- Institute
of Molecular and Cell Biology, Agency for Science, Technology
and Research, Singapore 138673, Singapore
| | - Danielle E. Anderson
- Program
in Emerging Infectious Diseases, Duke-NUS
Medical School, Singapore 169857, Singapore
| | - Florent Ginhoux
- Singapore Immunology
Network, Agency for Science, Technology and Research, Singapore 138648, Singapore
- SingHealth
Translational Immunology and Inflammation Centre, Singapore 169856, Singapore
| | | |
Collapse
|
6
|
Hall R, Alasmari A, Mozaffari S, Mahdipoor P, Parang K, Montazeri Aliabadi H. Peptide/Lipid-Associated Nucleic Acids (PLANAs) as a Multicomponent siRNA Delivery System. Mol Pharm 2021; 18:986-1002. [PMID: 33496597 DOI: 10.1021/acs.molpharmaceut.0c00969] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
RNAi is a biological process that utilizes small interfering RNA (siRNA) to prevent the translation of mRNA to protein. This mechanism could be beneficial in preventing the overexpression of proteins in cancer. However, the cellular delivery of siRNA has proven to be challenging due to its inherent negative charge and relative instability. Here, we designed a multicomponent delivery system composed of a specifically designed peptide (linear or cyclic fatty acyl peptide conjugates and hybrid cyclic/linear peptides) and several lipids (DOTAP, DOPE, cholesterol, and phosphatidylcholine) to form a nanoparticle, which we have termed as peptide lipid-associated nucleic acids (PLANAs). Five formulations were prepared (a formulation with no peptide, which was named lipid-associated nucleic acid or LANA, and PLANA formulations A-D) using a mini extruder to form uniform nanoparticles around 100 nm in size with a slightly positive charge (less than +10 mv). Formulations were evaluated for peptide incorporation, siRNA encapsulation efficiency, release profile, toxicity, cellular uptake, and protein silencing. Our experiments showed effective encapsulation of siRNA (>95%), a controlled release profile, and negligible toxicity in formulations that did not contain a positively charged lipid. The results also revealed that PLANAs C and D exhibited optimum cellular uptake (with 80-90% siRNA-positive cells for most of the formulations). PLANA D formulation was selected to silence two model proteins (Src and RPS6KA5) in the triple-negative human breast cancer cell line MDA-MB-231, with promising silencing efficiency, which diminished the expression of RPS6KA5 and Src to approximately 29 and 38% compared to naïve cells, respectively. Many approaches have been investigated for safe and efficient delivery of nucleic acids in the last 20 years; however, many have failed due to the multifaceted challenges to overcome. Our results show a promising potential for a multicomponent design that incorporates different components for a variety of delivery tasks, which warrants further investigation of PLANAs in vivo.
Collapse
Affiliation(s)
- Ryley Hall
- Center for Targeted Drug Delivery, Department of Biomedical and Pharmaceutical Sciences, Chapman University School of Pharmacy, Harry and Diane Rinker Health Science Campus, Irvine, California 92618, United States
| | - Abdulaziz Alasmari
- Center for Targeted Drug Delivery, Department of Biomedical and Pharmaceutical Sciences, Chapman University School of Pharmacy, Harry and Diane Rinker Health Science Campus, Irvine, California 92618, United States
| | - Saghar Mozaffari
- Center for Targeted Drug Delivery, Department of Biomedical and Pharmaceutical Sciences, Chapman University School of Pharmacy, Harry and Diane Rinker Health Science Campus, Irvine, California 92618, United States
| | - Parvin Mahdipoor
- Center for Targeted Drug Delivery, Department of Biomedical and Pharmaceutical Sciences, Chapman University School of Pharmacy, Harry and Diane Rinker Health Science Campus, Irvine, California 92618, United States
| | - Keykavous Parang
- Center for Targeted Drug Delivery, Department of Biomedical and Pharmaceutical Sciences, Chapman University School of Pharmacy, Harry and Diane Rinker Health Science Campus, Irvine, California 92618, United States
| | - Hamidreza Montazeri Aliabadi
- Center for Targeted Drug Delivery, Department of Biomedical and Pharmaceutical Sciences, Chapman University School of Pharmacy, Harry and Diane Rinker Health Science Campus, Irvine, California 92618, United States
| |
Collapse
|
7
|
Écija-Arenas Á, Román-Pizarro V, Fernández-Romero JM. Separation and characterization of liposomes using asymmetric flow field-flow fractionation with online multi-angle light scattering detection. J Chromatogr A 2020; 1636:461798. [PMID: 33341435 DOI: 10.1016/j.chroma.2020.461798] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2020] [Revised: 12/02/2020] [Accepted: 12/07/2020] [Indexed: 12/24/2022]
Abstract
Liposomes, mainly formed by phospholipids and cholesterol that entrapped different compounds, were separated and characterized using asymmetric flow field-flow fractionation (AF4) coupled with a multi-angle light scattering detector (MALS). AF4 allows the separation of liposomes according to their hydrodynamic size, and the particle size can be estimated directly by their elution time. Besides, different synthesized liposome suspensions of liposomes with different species encapsulated in different places in liposomes were prepared with analytical purposes to be studied. These liposomes were: empty liposomes (e-Ls), magnetoliposomes (MLs) with Fe3O4@AuNPs-C12SH inside the lipid bilayer, and long-wavelength fluorophores encapsulated into the aqueous cavity of liposomes (Ls-LWF). The optimization process of the variables that affect the fractionation has been established. The separation effectiveness has been compared with the results achieved with a photon-correlation spectroscopy analyzer based on dynamic light scattering (DLS) and transmission electron microscopy (TEM), used in self-assembly structures characterization. In all cases, three different classes of liposomes have been obtained; two are commonly appaired in all studied samples, while only a third class is characteristic for each of the liposomes. This mean that the proposed methodology could be used for identifying liposomes according to the encapsulated material.
Collapse
Affiliation(s)
- Ángela Écija-Arenas
- Departamento de Química Analítica, Instituto Universitario de Investigación en Química Fina y Nanoquímica (IUNAN), Universidad de Córdoba, Campus de Rabanales, Edificio Anexo "Marie Curie", Córdoba E-14071, España
| | - Vanesa Román-Pizarro
- Departamento de Química Analítica, Instituto Universitario de Investigación en Química Fina y Nanoquímica (IUNAN), Universidad de Córdoba, Campus de Rabanales, Edificio Anexo "Marie Curie", Córdoba E-14071, España
| | - Juan Manuel Fernández-Romero
- Departamento de Química Analítica, Instituto Universitario de Investigación en Química Fina y Nanoquímica (IUNAN), Universidad de Córdoba, Campus de Rabanales, Edificio Anexo "Marie Curie", Córdoba E-14071, España.
| |
Collapse
|
8
|
Complex Size and Surface Charge Determine Nucleic Acid Transfer by Fusogenic Liposomes. Int J Mol Sci 2020; 21:ijms21062244. [PMID: 32213928 PMCID: PMC7139958 DOI: 10.3390/ijms21062244] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2020] [Revised: 03/16/2020] [Accepted: 03/21/2020] [Indexed: 12/12/2022] Open
Abstract
Highly efficient, biocompatible, and fast nucleic acid delivery methods are essential for biomedical applications and research. At present, two main strategies are used to this end. In non-viral transfection liposome- or polymer-based formulations are used to transfer cargo into cells via endocytosis, whereas viral carriers enable direct nucleic acid delivery into the cell cytoplasm. Here, we introduce a new generation of liposomes for nucleic acid delivery, which immediately fuse with the cellular plasma membrane upon contact to transfer the functional nucleic acid directly into the cell cytoplasm. For maximum fusion efficiency combined with high cargo transfer, nucleic acids had to be complexed and partially neutralized before incorporation into fusogenic liposomes. Among the various neutralization agents tested, small, linear, and positively charged polymers yielded the best complex properties. Systematic variation of liposomal composition and nucleic acid complexation identified surface charge as well as particle size as essential parameters for cargo-liposome interaction and subsequent fusion induction. Optimized protocols were tested for the efficient transfer of different kinds of nucleic acids like plasmid DNA, messenger RNA, and short-interfering RNA into various mammalian cells in culture and into primary tissues.
Collapse
|
9
|
Kim JH, Hong SS, Lee M, Lee EH, Rhee I, Chang SY, Lim SJ. Krill Oil-Incorporated Liposomes As An Effective Nanovehicle To Ameliorate The Inflammatory Responses Of DSS-Induced Colitis. Int J Nanomedicine 2019; 14:8305-8320. [PMID: 31806959 PMCID: PMC6844156 DOI: 10.2147/ijn.s220053] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2019] [Accepted: 08/23/2019] [Indexed: 01/05/2023] Open
Abstract
Background Phosphatidylcholine (PC) and Omega-3 fatty acid (Omega-3) are promising therapeutic molecules for treating inflammatory bowel disease (IBD). Purpose Based on the IBD therapeutic potential of nanoparticles, we herein sought to develop Omega-3-incorporated PC nanoparticles (liposomes) as an orally administrable vehicle for treating IBD. Methods Liposomes prepared with or without Omega-3 incorporation were compared in terms of colloidal stability and anitiinflammatory effects. Results The incorporation of free Omega-3 (alpha-linolenic acid, eicosapentaenoic acid or docosahexaenoic acid) into liposomes induced time-dependent membrane fusion, resulting in particle size increase from nm to μm during storage. In contrast, krill oil incorporation into liposomes (KO liposomes) did not induce the fusion and the particle size maintained <250 nm during storage. KO liposomes also maintained colloidal stability in simulated gastrointestinal conditions and exhibited a high capacity to entrap the IBD drug, budesonide (BDS). KO liposomes greatly suppressed the lipopolysaccharide-induced production of pro-inflammatory cytokines in cultured macrophages and completely restored inflammation-impaired membrane barrier function in an intestinal barrier model. In mice subjected to dextran sulfate sodium-induced colitis, oral administration of BDS-entrapped KO liposomes suppressed tumor necrosis factor-α production (by 84.1%), interleukin-6 production (by 35.3%), and the systemic level of endotoxin (by 96.8%), and slightly reduced the macroscopic signs of the disease. Conclusion Taken together, KO liposomes may have great potential as a nanovehicle for oral delivery of IBD drugs.
Collapse
Affiliation(s)
- Jin-Hee Kim
- Department of Integrated Bioscience and Biotechnology, Sejong University, Seoul 05006, Republic of Korea
| | - Soon-Seok Hong
- Department of Integrated Bioscience and Biotechnology, Sejong University, Seoul 05006, Republic of Korea
| | - Myoungsoo Lee
- Laboratory of Microbiology, College of Pharmacy, and Research Institute of Pharmaceutical Science and Technology (RIPST), Ajou University, Suwon 16499, Republic of Korea
| | - Eun-Hye Lee
- Department of Integrated Bioscience and Biotechnology, Sejong University, Seoul 05006, Republic of Korea
| | - Inmoo Rhee
- Department of Integrated Bioscience and Biotechnology, Sejong University, Seoul 05006, Republic of Korea
| | - Sun-Young Chang
- Laboratory of Microbiology, College of Pharmacy, and Research Institute of Pharmaceutical Science and Technology (RIPST), Ajou University, Suwon 16499, Republic of Korea
| | - Soo-Jeong Lim
- Department of Integrated Bioscience and Biotechnology, Sejong University, Seoul 05006, Republic of Korea
| |
Collapse
|
10
|
Multivesicular Liposomes for the Sustained Release of Angiotensin I-Converting Enzyme (ACE) Inhibitory Peptides from Peanuts: Design, Characterization, and In Vitro Evaluation. Molecules 2019; 24:molecules24091746. [PMID: 31060345 PMCID: PMC6539825 DOI: 10.3390/molecules24091746] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2019] [Revised: 04/18/2019] [Accepted: 04/29/2019] [Indexed: 11/17/2022] Open
Abstract
The multivesicular liposome (MVL) provides a potential delivery approach to avoid the destruction of the structure of drugs by digestive enzymes of the oral cavity and gastrointestinal system. It also serves as a sustained-release drug delivery system. In this study, we aimed to incorporate a water-soluble substance into MVLs to enhance sustained release, prevent the destruction of drugs, and to expound the function of different components and their mechanism. MVLs were prepared using the spherical packing model. The morphology, structure, size distribution, and zeta potential of MVLs were examined using an optical microscope (OM), confocal microscopy (CLSM), transmission electron cryomicroscope (cryo-EM) micrograph, a Master Sizer 2000, and a zeta sizer, respectively. The digestion experiment was conducted using a bionic mouse digestive system model in vitro. An in vitro release and releasing mechanism were investigated using a dialysis method. The average particle size, polydispersity index, zeta potential, and encapsulation efficiency are 47.6 nm, 1.880, −70.5 ± 2.88 mV, and 82.00 ± 0.25%, respectively. The studies on the controlled release in vitro shows that MVLs have excellent controlled release and outstanding thermal stability. The angiotensin I-converting enzyme (ACE) inhibitory activity of ACE-inhibitory peptide (AP)-MVLs decreased only 2.84% after oral administration, and ACE inhibitory activity decreased by 5.03% after passing through the stomach. Therefore, it could serve as a promising sustained-release drug delivery system.
Collapse
|
11
|
Hernandez Y, González-Pastor R, Belmar-Lopez C, Mendoza G, de la Fuente JM, Martin-Duque P. Gold nanoparticle coatings as efficient adenovirus carriers to non-infectable stem cells. RSC Adv 2019; 9:1327-1334. [PMID: 35517997 PMCID: PMC9059632 DOI: 10.1039/c8ra09088b] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2018] [Revised: 02/07/2019] [Accepted: 12/24/2018] [Indexed: 12/11/2022] Open
Abstract
Mesenchymal stem cells (MSCs) are adult pluripotent cells with the plasticity to be converted into different cell types. Their self-renewal capacity, relative ease of isolation, expansion and inherent migration to tumors, make them perfect candidates for cell therapy against cancer. However, MSCs are notoriously refractory to adenoviral infection, mainly because CAR (Coxsackie-Adenovirus Receptor) expression is absent or downregulated. Over the last years, nanoparticles have attracted a great deal of attention as potential vehicle candidates for gene delivery, but with limited effects on their own. Our data showed that the use of positively charged 14 nm gold nanoparticles either functionalized with arginine-glycine-aspartate (RGD) motif or not, increases the efficiency of adenovirus infection in comparison to commercial reagents without altering cell viability or cell phenotype. This system represents a simple, efficient and safe method for the transduction of MSCs, being attractive for cancer gene and cell therapies.
Collapse
Affiliation(s)
- Yulan Hernandez
- Instituto de Nanociencia de Aragón (INA), Universidad de Zaragoza 50018 Spain
| | - Rebeca González-Pastor
- Instituto Aragonés de Ciencias de la Salud 50009 Zaragoza Spain
- Instituto de Investigaciones Sanitarias de Aragón (IIS Aragón) 50009 Zaragoza Spain
| | - Carolina Belmar-Lopez
- Instituto Aragonés de Ciencias de la Salud 50009 Zaragoza Spain
- Instituto de Investigaciones Sanitarias de Aragón (IIS Aragón) 50009 Zaragoza Spain
| | - Gracia Mendoza
- Instituto de Nanociencia de Aragón (INA), Universidad de Zaragoza 50018 Spain
- Instituto de Investigaciones Sanitarias de Aragón (IIS Aragón) 50009 Zaragoza Spain
| | - Jesus M de la Fuente
- Instituto de Ciencias de Materiales (ICMA), CSIC 50009 Zaragoza Spain
- CIBER-BBN 28029 Madrid Spain
| | - Pilar Martin-Duque
- Instituto Aragonés de Ciencias de la Salud 50009 Zaragoza Spain
- Instituto de Investigaciones Sanitarias de Aragón (IIS Aragón) 50009 Zaragoza Spain
- Fundación Araid 50001 Zaragoza Spain
| |
Collapse
|