1
|
Mahjoubin-Tehran M, Rezaei S, Kesharwani P, Karav S, Sahebkar A. Decoy oligodeoxynucleotides targeting STATs in non-cancer gene therapy. Gene 2025; 957:149482. [PMID: 40216342 DOI: 10.1016/j.gene.2025.149482] [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: 12/03/2024] [Revised: 04/06/2025] [Accepted: 04/07/2025] [Indexed: 04/14/2025]
Abstract
The Signal Transducer and Activator of Transcription (STAT) protein family is crucial for organizing the epigenetic configuration of immune cells and controlling various fundamental cell physiological functions including apoptosis, development, inflammation, immunological responses, and cell proliferation and differentiation. The human genome has seven known STAT genes, named 1, 2, 3, 4, 5a, 5b, and 6. Aberrant activation of STAT signaling pathways is associated with many human disorders, particularly cardiovascular diseases (CVDs), making these proteins promising therapeutic targets. Improved understanding of altered and pathological gene expression and its role in the pathophysiology of various hereditary and acquired disorders has enabled the development of novel treatment approaches based on gene expression modulation. One such promising development is the oligodeoxynucleotide decoy method, which may allow researchers to specifically influence gene activation or repression. Various oligodeoxynucleotide decoys target STATs and affect the expression of its downstream genes. We summarized cell culture and preclinical research, which evaluated the effects of oligodeoxynucleotide decoys target STATs in different types of non-cancer illnesses.
Collapse
Affiliation(s)
- Maryam Mahjoubin-Tehran
- Department of Biotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Samaneh Rezaei
- Department of Medical Biotechnology and Nanotechnology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Prashant Kesharwani
- Department of Pharmaceutical Sciences, Dr. Harisingh Gour Vishwavidyalaya, Sagar, Madhya Pradesh 470003, India.
| | - Sercan Karav
- Department of Molecular Biology and Genetics, Canakkale Onsekiz Mart University, Canakkale 17100, Turkey
| | - Amirhossein Sahebkar
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Centre for Research Impact and Outcome, Chitkara University, Rajpura 140417, Punjab, India; Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
2
|
de Avila Goncalves S, Ceccato BT, Moraes-Lacerda T, de Jesus MB, de la Torre LG, Vieira RP. Synthesis of poly[2-(dimethylamino)ethyl methacrylate] grafting from cellulose nanocrystals for DNA complexation employing a 3D-twisted cross-sectional microchannel microfluidic device. Int J Biol Macromol 2025; 305:140992. [PMID: 39952531 DOI: 10.1016/j.ijbiomac.2025.140992] [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: 06/30/2024] [Revised: 12/29/2024] [Accepted: 02/11/2025] [Indexed: 02/17/2025]
Abstract
Developing effective and safe non-viral gene vectors poses a challenge in gene therapy. A promising strategy emerged addressing this challenge, involving a synergistic approach combining biopolymers and cationic synthetic polymers to enhance gene delivery systems. In this study, for the first time, poly[2-(dimethylamino)ethyl methacrylate] (PDMAEMA) was grafted from cellulose nanocrystals (CNC) using metal-free organocatalyzed atom-transfer radical polymerization (O-ATRP). The synthesis was confirmed through morphological, spectroscopic, and thermal analysis. The reaction achieved a 34 % monomer conversion and 15 % grafting, resulting in a CNC-g-PDMAEMA copolymer with impressive responsiveness to pH and temperature. Furthermore, CNC-g-PDMAEMA was utilized to obtain copolymer/pDNA polyplexes using a microfluidic device, providing a practical and efficient method for producing uniform, stable, and reproducible gene delivery systems. These polyplexes had sizes around 160 nm and a low PDI (<0.250). As a proof of concept, preliminary cell viability and transfection assays were conducted to demonstrate the biomaterial's applicability. These findings suggest that polyplexes (N/P = 15) at a 10 μg/mL concentration may serve as an upper limit threshold and a starting point for further in vivo studies. In summary, this research advances the development of gene delivery platforms through innovative and straightforward synthesis methods, opening up potential applications in gene therapy.
Collapse
Affiliation(s)
- Sayeny de Avila Goncalves
- Department of Bioprocess and Materials Engineering, School of Chemical Engineering, University of Campinas, Campinas, São Paulo, Brazil.
| | - Bruno Telli Ceccato
- Department of Bioprocess and Materials Engineering, School of Chemical Engineering, University of Campinas, Campinas, São Paulo, Brazil
| | - Thaís Moraes-Lacerda
- Department of Biochemistry and Tissue Biology, Institute of Biology, University of Campinas, Campinas, São Paulo, Brazil
| | - Marcelo Bispo de Jesus
- Department of Biochemistry and Tissue Biology, Institute of Biology, University of Campinas, Campinas, São Paulo, Brazil
| | - Lucimara Gaziola de la Torre
- Department of Bioprocess and Materials Engineering, School of Chemical Engineering, University of Campinas, Campinas, São Paulo, Brazil
| | - Roniérik Pioli Vieira
- Department of Bioprocess and Materials Engineering, School of Chemical Engineering, University of Campinas, Campinas, São Paulo, Brazil.
| |
Collapse
|
3
|
Pan X, Zhao X, Lu Y, Xie P, Liu L, Chu X. Harnessing Nanomaterials for Enhanced DNA-Based Biosensing and Therapeutic Performance. Chembiochem 2025; 26:e202400936. [PMID: 39655520 DOI: 10.1002/cbic.202400936] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2024] [Revised: 12/09/2024] [Accepted: 12/09/2024] [Indexed: 12/19/2024]
Abstract
The integration of nanomaterials with DNA-based systems has emerged as a transformative approach in biosensing and therapeutic applications. Unique features of DNA, like its programmability and specificity, complement the diverse functions of nanomaterials, leading to the creation of advanced systems for detecting biomarkers and delivering treatments. Here, we review the developments in DNA-nanomaterial conjugates, emphasizing their enhanced functionalities and potential across various biomedical applications. We first discuss the methodologies for synthesizing these conjugates, distinguishing between covalent and non-covalent interactions. We then categorize DNA-nanomaterials conjugates based on the properties of the DNA and nanomaterials involved, respectively. DNA probes are classified by their application into biosensing or therapeutic uses, and, several nanomaterials are highlighted by their recent progress in living biological. Finally, we discuss the current challenges and future prospects in this field, anticipating that significant progress in DNA-nanomaterial conjugates will greatly enhance precision medicine.
Collapse
Affiliation(s)
- Xumin Pan
- State Key Laboratory of Chemo/Bio-Sensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, 410082, Changsha, P. R. China
| | - Xiaoman Zhao
- State Key Laboratory of Chemo/Bio-Sensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, 410082, Changsha, P. R. China
| | - Yanhong Lu
- State Key Laboratory of Chemo/Bio-Sensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, 410082, Changsha, P. R. China
| | - Ping Xie
- State Key Laboratory of Chemo/Bio-Sensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, 410082, Changsha, P. R. China
| | - Lan Liu
- State Key Laboratory of Chemo/Bio-Sensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, 410082, Changsha, P. R. China
| | - Xia Chu
- State Key Laboratory of Chemo/Bio-Sensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, 410082, Changsha, P. R. China
| |
Collapse
|
4
|
Jeleriu RM, Hajaj RK, Trăilă IA, Zaharie M, Puiu M. Theoretical Models and Simulations of Gene Delivery with Polyurethane: The Importance of Polyurethane as a Vector in Personalized Therapy. Biomedicines 2025; 13:692. [PMID: 40149668 PMCID: PMC11939958 DOI: 10.3390/biomedicines13030692] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2025] [Revised: 03/08/2025] [Accepted: 03/10/2025] [Indexed: 03/29/2025] Open
Abstract
Background/Objectives: Advancements in personalized medicine have revolutionized drug delivery, enabling tailored treatments based on genetic and molecular profiles. Non-viral vectors, such as polyurethane (PU)-based systems, offer promising alternatives for gene therapy. This study develops mathematical models to analyze PU degradation, DNA/RNA release kinetics, and cellular interactions, optimizing their application in personalized therapy. Methods: This theoretical study utilized mathematical modeling and numerical simulations to analyze PU-based gene delivery, focusing on diffusion, degradation, and cellular uptake. Implemented in Python 3.9, it employed differential equation solvers and adsorption/internalization models to predict vector behavior and optimize delivery efficiency. Results: This study demonstrated that PU degrades in biological environments following first-order kinetics, ensuring a controlled and predictable release of genetic material. The Higuchi diffusion model confirmed a gradual, sustained DNA/RNA release, essential for efficient gene delivery. Simulations of PU adsorption onto cellular membranes using the Langmuir model showed saturation-dependent binding, while the endocytosis model revealed a balance between uptake and degradation. These findings highlight PU's potential as a versatile gene delivery vector, offering controlled biodegradability, optimized release profiles, and effective cellular interaction. Conclusions: Our results confirm that PU-based vectors enable controlled biodegradability, sustained DNA/RNA release, and efficient cellular uptake. Mathematical modeling provides a framework for improving PU's properties, enhancing transport efficiency and therapeutic potential in personalized medicine and gene therapy applications.
Collapse
Affiliation(s)
- Roxana Maria Jeleriu
- Ph.D. School, Faculty of Medicine, Department of Microscopic Morphology, Genetics Discipline, Center of Genomic Medicine, “Victor Babes” University of Medicine and Pharmacy Timisoara, 2 E. Murgu, Sq., 300041 Timisoara, Romania; (R.M.J.); (R.-K.H.); (M.P.)
| | - Roxana-Karin Hajaj
- Ph.D. School, Faculty of Medicine, Department of Microscopic Morphology, Genetics Discipline, Center of Genomic Medicine, “Victor Babes” University of Medicine and Pharmacy Timisoara, 2 E. Murgu, Sq., 300041 Timisoara, Romania; (R.M.J.); (R.-K.H.); (M.P.)
- Department of Pediatric Surgery and Orthopedics, “Louis Țurcanu” Emergency Clinical Hospital for Children, 300011 Timisoara, Romania
| | - Iuliana-Anamaria Trăilă
- Department of Pathology, ‘Pius Brinzeu’ Emergency County Clinical Hospital, 300723 Timisoara, Romania
| | - Mihaela Zaharie
- Department XII Obstetrics-Gynecology, ‘Victor Babeş’ University of Medicine and Pharmacy of Timișoara, 300041 Timisoara, Romania;
- Neonatology-Premature Unit, “Louis Țurcanu” Emergency Clinical Hospital for Children, 300011 Timisoara, Romania
| | - Maria Puiu
- Ph.D. School, Faculty of Medicine, Department of Microscopic Morphology, Genetics Discipline, Center of Genomic Medicine, “Victor Babes” University of Medicine and Pharmacy Timisoara, 2 E. Murgu, Sq., 300041 Timisoara, Romania; (R.M.J.); (R.-K.H.); (M.P.)
- Institute for Research and Development in Genomics, 020021 București, Romania
| |
Collapse
|
5
|
Zhou ZR, Wu MS, Yang Z, Wu Y, Guo W, Li DW, Qian RC, Lu Y. Synthetic transmembrane DNA receptors enable engineered sensing and actuation. Nat Commun 2025; 16:1464. [PMID: 39920144 PMCID: PMC11806108 DOI: 10.1038/s41467-025-56758-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Accepted: 01/30/2025] [Indexed: 02/09/2025] Open
Abstract
In living organisms, cells synergistically couple cascade reaction pathways to achieve inter- and intracellular signal transduction by transmembrane protein receptors. The construction and assembly of synthetic receptor analogs that can mimic such biological processes is a central goal of synthetic biochemistry and bionanotechnology to endow receptors with user-defined signal transduction effects. However, designing artificial transmembrane receptors with the desired input, output, and performance parameters are challenging. Here we show that the dimerization of synthetic transmembrane DNA receptors executes a systematically engineered sensing and actuation cascade in response to external molecular signals. The synthetic DNA receptors are composed of three parts, including an extracellular signal reception part, a lipophilic transmembrane anchoring part, and an intracellular signal output part. Upon the input of external signals, the DNA receptors can form dimers on the cell surface triggered by configuration changes, leading to a series of downstream cascade events including communication between donor and recipient cells, gene transcription regulation, protein level control, and cell apoptosis. We believe this work establishes a flexible cell surface engineering strategy that is broadly applicable to implement sophisticated biological functions.
Collapse
Affiliation(s)
- Ze-Rui Zhou
- Key Laboratory for Advanced Materials. East China University of Science and Technology, Shanghai, 200237, P. R. China
- Feringa Nobel Prize Scientist Joint Research Center, Joint International Laboratory for Precision Chemistry. East China University of Science and Technology, Shanghai, 200237, P. R. China
- Frontiers Science Center for Materiobiology & Dynamic Chemistry. East China University of Science and Technology, Shanghai, 200237, P. R. China
- School of Chemistry and Molecular Engineering, East China University of Science and Technology, Shanghai, 200237, P. R. China
- Department of Chemistry, University of Texas at Austin, Austin, TX, 78712, USA
| | - Man-Sha Wu
- Key Laboratory for Advanced Materials. East China University of Science and Technology, Shanghai, 200237, P. R. China
- Feringa Nobel Prize Scientist Joint Research Center, Joint International Laboratory for Precision Chemistry. East China University of Science and Technology, Shanghai, 200237, P. R. China
- Frontiers Science Center for Materiobiology & Dynamic Chemistry. East China University of Science and Technology, Shanghai, 200237, P. R. China
- School of Chemistry and Molecular Engineering, East China University of Science and Technology, Shanghai, 200237, P. R. China
| | - Zhenglin Yang
- Department of Chemistry, University of Texas at Austin, Austin, TX, 78712, USA
| | - Yuting Wu
- Department of Chemistry, University of Texas at Austin, Austin, TX, 78712, USA
| | - Weijie Guo
- Department of Molecular Biosciences, University of Texas at Austin, Austin, TX, 78712, USA
| | - Da-Wei Li
- Key Laboratory for Advanced Materials. East China University of Science and Technology, Shanghai, 200237, P. R. China
- Feringa Nobel Prize Scientist Joint Research Center, Joint International Laboratory for Precision Chemistry. East China University of Science and Technology, Shanghai, 200237, P. R. China
- Frontiers Science Center for Materiobiology & Dynamic Chemistry. East China University of Science and Technology, Shanghai, 200237, P. R. China
- School of Chemistry and Molecular Engineering, East China University of Science and Technology, Shanghai, 200237, P. R. China
| | - Ruo-Can Qian
- Key Laboratory for Advanced Materials. East China University of Science and Technology, Shanghai, 200237, P. R. China.
- Feringa Nobel Prize Scientist Joint Research Center, Joint International Laboratory for Precision Chemistry. East China University of Science and Technology, Shanghai, 200237, P. R. China.
- Frontiers Science Center for Materiobiology & Dynamic Chemistry. East China University of Science and Technology, Shanghai, 200237, P. R. China.
- School of Chemistry and Molecular Engineering, East China University of Science and Technology, Shanghai, 200237, P. R. China.
| | - Yi Lu
- Department of Chemistry, University of Texas at Austin, Austin, TX, 78712, USA.
- Department of Molecular Biosciences, University of Texas at Austin, Austin, TX, 78712, USA.
| |
Collapse
|
6
|
Sabet MJ, Hasanzadeh A, Vahabi A, Hosseini ES, Saeedi S, Chegeni BK, Kiani J, Azar BKY, Molabashi ZA, Shamsara M, Hamblin MR, Karimi M, Roustazadeh A. Metal-Coordinated Histidine-Functionalized Redox-Responsive Polyethyleneimine as a Smart Gene Delivery Vector. Mol Biotechnol 2025:10.1007/s12033-024-01360-x. [PMID: 39806116 DOI: 10.1007/s12033-024-01360-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2024] [Accepted: 12/12/2024] [Indexed: 01/16/2025]
Abstract
Despite significant advancements in gene delivery and CRISPR technology, several challenges remain. Chief among these are overcoming serum inhibition and achieving high transfection efficiency with minimal cytotoxicity. To address these issues, there is a need for novel vectors that exhibit lower toxicity, maintain stability in serum-rich environments, and effectively deliver plasmids of various sizes across diverse cell types. In this study, to convert common polyethylenimine (PEI1.8k) into high-performance DNA delivery vectors, an innovative multifunctional vector was constructed based on histidine linked to PEI1.8k by redox-responsive disulfide bonds. Apart from highly efficient transfection of both small and large plasmids into HEK 293T (Human Embryonic Kidney 293T cells) with negligible cytotoxicity, PEI1.8k-S-S-His showed great transfection potential even at low plasmid doses (0.5 µg), as well as at serum concentrations ranging from 5 to 30% into HEK 293T cells, and achieved excellent plasmid transfection into NIH/3T3 (Mouse Embryonic Fibroblast cells), and MCF7 (Human Breast Cancer cells). Additionally, several metals were tested (Co, Cu, Cd, Ni, Zn, and Mn) to promote the plasmid packaging functionality and improve transfection efficiency. We observed that, in comparison to PEI1.8k-S-S-His, the manganese-functionalized nanocarrier (PEI1.8k-S-S-His-Mn) could transfect a large plasmid with equal efficiency (~ 30%) into MSCs (Mesenchymal Stem Cells). Interestingly, PEI1.8k-S-S-His-Mn showed higher transfection efficiency with the small plasmid (~ 90%) and the large one (~ 80%) into HEK 293T cells, even better than its backbone. We propose that the presence of metal-coordinated His ligand, redox-responsive S-S bonds, and the cationic polymer can synergistically provide robust DNA binding, efficient endosomal disruption, tolerance of serum protein adsorption, and low cytotoxicity. These new vectors could be promising for gene delivery and may be therapeutically relevant.
Collapse
Affiliation(s)
- Makkieh Jahanpeimay Sabet
- Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran, Iran
- Department of Medical Nanotechnology, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran
- Advanced Nanobiotechnology and Nanomedicine Research Group (ANNRG), Iran University of Medical Sciences, Tehran, Iran
| | - Akbar Hasanzadeh
- Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran, Iran
- Department of Medical Nanotechnology, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran
- Advanced Nanobiotechnology and Nanomedicine Research Group (ANNRG), Iran University of Medical Sciences, Tehran, Iran
| | - Amirhossein Vahabi
- Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran, Iran
- Department of Medical Nanotechnology, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran
- Advanced Nanobiotechnology and Nanomedicine Research Group (ANNRG), Iran University of Medical Sciences, Tehran, Iran
| | - Elaheh Sadat Hosseini
- Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran, Iran
- Department of Medical Nanotechnology, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran
- Advanced Nanobiotechnology and Nanomedicine Research Group (ANNRG), Iran University of Medical Sciences, Tehran, Iran
| | - Sara Saeedi
- Department of Medical Nanotechnology, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran
- Neuroscience Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Beheshteh Khodadadi Chegeni
- Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran, Iran
- Department of Medical Nanotechnology, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran
- Advanced Nanobiotechnology and Nanomedicine Research Group (ANNRG), Iran University of Medical Sciences, Tehran, Iran
| | - Jafar Kiani
- Oncopathology Research Center, Iran University of Medical Sciences, Tehran, Iran
- Department of Molecular Medicine, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Behjat Kheiri Yeghaneh Azar
- Department of Molecular Medicine, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran
- Student Research Committee, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Zahra Asghari Molabashi
- Department of Plant Molecular Biotechnology, National Institute of Genetic Engineering and Biotechnology, Tehran, Iran
| | - Mehdi Shamsara
- Department of Animal Biotechnology, National Institute of Genetic Engineering and Biotechnology, Tehran, Iran
| | - Michael R Hamblin
- Laser Research Centre, Faculty of Health Science, University of Johannesburg, Doornfontein, 2028, South Africa
| | - Mahdi Karimi
- Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran, Iran.
- Department of Medical Nanotechnology, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran.
- Advanced Nanobiotechnology and Nanomedicine Research Group (ANNRG), Iran University of Medical Sciences, Tehran, Iran.
- Oncopathology Research Center, Iran University of Medical Sciences, Tehran, Iran.
- Research Center for Science and Technology in Medicine, Tehran University of Medical Sciences, Tehran, Iran.
- Applied Biotechnology Research Centre, Tehran Medical Science, Islamic Azad University, Tehran, Iran.
| | - Abazar Roustazadeh
- Noncommunicable Disease Research Center, Jahrom University of Medical Sciences, Jahrom, Iran.
- Dapartment of Biochemistry, Jahrom University of Medical Sciences, Jahrom, Iran.
- Department of Advanced Medical Sciences and Technologies, Jahrom University of Medical Sciences, Jahrom, Iran.
| |
Collapse
|
7
|
Beigi A, Naghib SM, Matini A, Tajabadi M, Mozafari MR. Lipid-Based Nanocarriers for Targeted Gene Delivery in Lung Cancer Therapy: Exploring a Novel Therapeutic Paradigm. Curr Gene Ther 2025; 25:92-112. [PMID: 38778601 DOI: 10.2174/0115665232292768240503050508] [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: 11/14/2023] [Revised: 03/28/2024] [Accepted: 04/03/2024] [Indexed: 05/25/2024]
Abstract
Lung cancer is a significant cause of cancer-related death worldwide. It can be broadly categorised into small-cell lung cancer (SCLC) and Non-small cell lung cancer (NSCLC). Surgical intervention, radiation therapy, and the administration of chemotherapeutic medications are among the current treatment modalities. However, the application of chemotherapy may be limited in more advanced stages of metastasis due to the potential for adverse effects and a lack of cell selectivity. Although small-molecule anticancer treatments have demonstrated effectiveness, they still face several challenges. The challenges at hand in this context comprise insufficient solubility in water, limited bioavailability at specific sites, adverse effects, and the requirement for epidermal growth factor receptor inhibitors that are genetically tailored. Bio-macromolecular drugs, including small interfering RNA (siRNA) and messenger RNA (mRNA), are susceptible to degradation when exposed to the bodily fluids of humans, which can reduce stability and concentration. In this context, nanoscale delivery technologies are utilised. These agents offer encouraging prospects for the preservation and regulation of pharmaceutical substances, in addition to improving the solubility and stability of medications. Nanocarrier-based systems possess the notable advantage of facilitating accurate and sustained drug release, as opposed to traditional systemic methodologies. The primary focus of scientific investigation has been to augment the therapeutic efficacy of nanoparticles composed of lipids. Numerous nanoscale drug delivery techniques have been implemented to treat various respiratory ailments, such as lung cancer. These technologies have exhibited the potential to mitigate the limitations associated with conventional therapy. As an illustration, applying nanocarriers may enhance the solubility of small-molecule anticancer drugs and prevent the degradation of bio-macromolecular drugs. Furthermore, these devices can administer medications in a controlled and extended fashion, thereby augmenting the therapeutic intervention's effectiveness and reducing adverse reactions. However, despite these promising results, challenges remain that must be addressed. Multiple factors necessitate consideration when contemplating the application of nanoparticles in medical interventions. To begin with, the advancement of more efficient delivery methods is imperative. In addition, a comprehensive investigation into the potential toxicity of nanoparticles is required. Finally, additional research is needed to comprehend these treatments' enduring ramifications. Despite these challenges, the field of nanomedicine demonstrates considerable promise in enhancing the therapy of lung cancer and other respiratory diseases.
Collapse
Affiliation(s)
- Anahita Beigi
- Nanotechnology Department, School of Advanced Technologies, Iran University of Science and Technology, Tehran, Iran
| | - Seyed Morteza Naghib
- Nanotechnology Department, School of Advanced Technologies, Iran University of Science and Technology, Tehran, Iran
| | - Amir Matini
- Nanotechnology Department, School of Advanced Technologies, Iran University of Science and Technology, Tehran, Iran
| | - Maryam Tajabadi
- School of Metallurgy and Materials Engineering, Iran University of Science and Technology (IUST), Narmak, Tehran, 16844, Iran
| | - Mohammad Reza Mozafari
- Australasian Nanoscience and Nanotechnology Initiative (ANNI), Monash University LPO, Clayton, VIC 3168, Australia
| |
Collapse
|
8
|
Sun M, Song R, Fang Y, Xu J, Yang Z, Zhang H. DNA-Based Complexes and Composites: A Review of Fabrication Methods, Properties, and Applications. ACS APPLIED MATERIALS & INTERFACES 2024; 16:51899-51915. [PMID: 39314016 DOI: 10.1021/acsami.4c13357] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/25/2024]
Abstract
Deoxyribonucleic acid (DNA), a macromolecule that stores genetic information in organisms, has recently been gradually developed into a building block for new materials due to its stable chemical structure and excellent biocompatibility. The efficient preparation and functional integration of various molecular complexes and composite materials based on nucleic acid skeletons have been successfully achieved. These versatile materials possess excellent physical and chemical properties inherent to certain inorganic or organic molecules but are endowed with specific physiological functions by nucleic acids, demonstrating unique advantages and potential applications in materials science, nanotechnology, and biomedical engineering in recent years. However, issues such as the production cost, biological stability, and potential immunogenicity of DNA have presented some unprecedented challenges to the application of these materials in the field. This review summarizes the cutting-edge manufacturing techniques and unique properties of DNA-based complexes and composites and discusses the trends, challenges, and opportunities for the future development of nucleic acid-based materials.
Collapse
Affiliation(s)
- Mengqiu Sun
- School of Physical Sciences, Great Bay University, Dongguan 523000, China
- Department of Materials Science and Engineering, Southern University of Science and Technology, Shenzhen 518055, China
| | - Rui Song
- School of Physical Sciences, Great Bay University, Dongguan 523000, China
- Research & Development Institute of Northwestern Polytechnical University in Shenzhen, Shenzhen 518063, China
| | - Yangwu Fang
- School of Physical Sciences, Great Bay University, Dongguan 523000, China
| | - Jiuzhou Xu
- School of Life Sciences, Southern University of Science and Technology, Shenzhen 518055, China
| | - Zhaoqi Yang
- School of Life Sciences and Health Engineering, Jiangnan University, Wuxi 214122, China
| | - Hao Zhang
- School of Physical Sciences, Great Bay University, Dongguan 523000, China
- Research & Development Institute of Northwestern Polytechnical University in Shenzhen, Shenzhen 518063, China
| |
Collapse
|
9
|
Khan M. Polymers as Efficient Non-Viral Gene Delivery Vectors: The Role of the Chemical and Physical Architecture of Macromolecules. Polymers (Basel) 2024; 16:2629. [PMID: 39339093 PMCID: PMC11435517 DOI: 10.3390/polym16182629] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2024] [Revised: 09/13/2024] [Accepted: 09/15/2024] [Indexed: 09/30/2024] Open
Abstract
Gene therapy is the technique of inserting foreign genetic elements into host cells to achieve a therapeutic effect. Although gene therapy was initially formulated as a potential remedy for specific genetic problems, it currently offers solutions for many diseases with varying inheritance patterns and acquired diseases. There are two major groups of vectors for gene therapy: viral vector gene therapy and non-viral vector gene therapy. This review examines the role of a macromolecule's chemical and physical architecture in non-viral gene delivery, including their design and synthesis. Polymers can boost circulation, improve delivery, and control cargo release through various methods. The prominent examples discussed include poly-L-lysine, polyethyleneimine, comb polymers, brush polymers, and star polymers, as well as hydrogels and natural polymers and their modifications. While significant progress has been made, challenges still exist in gene stabilization, targeting specificity, and cellular uptake. Overcoming cytotoxicity, improving delivery efficiency, and utilizing natural polymers and hybrid systems are vital factors for prospects. This comprehensive review provides an illuminating overview of the field, guiding the way toward innovative non-viral-based gene delivery solutions.
Collapse
Affiliation(s)
- Majad Khan
- Department of Chemistry, King Fahd University of Petroleum & Minerals KFUPM, Dahran 31261, Saudi Arabia
- Interdisciplinary Research Center for Hydrogen Technologies and Carbon Management (IRC-HTCM), King Fahd University of Petroleum & Minerals KFUPM, Dahran 31261, Saudi Arabia
- Interdisciplinary Research Center for Refining and Advanced Chemicals (IRC-CRAC), King Fahd University of Petroleum & Minerals (KFUPM), Dhahran 31261, Saudi Arabia
| |
Collapse
|
10
|
Lu K, Jia D, Zhang H, Cheng J, Zhang Y, Zhang Y, Yu Q, Chen H. A Photothermal Polymeric Platform for Efficient and Safe Gene Transfection: When Polyethylenimine Collaborates with Indocyanine Green. ACS APPLIED MATERIALS & INTERFACES 2024; 16:44376-44385. [PMID: 39145762 DOI: 10.1021/acsami.4c10144] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/16/2024]
Abstract
Gene transfection, defined by the delivery of nucleic acids into cellular compartments, stands as a crucial procedure in gene therapy. While branched polyethylenimine (PEI) is widely regarded as the "gold standard" for nonviral vectors, its cationic nature presents several issues, including nonspecific protein adsorption and notable cytotoxicity. Additionally, it often fails to achieve high transfection efficiency, particularly with hard-to-transfect cell types. To overcome these challenges associated with PEI as a vector for plasmid DNA (pDNA), the photothermal agent indocyanine green (ICG) is integrated with PEI and pDNA to form the PEI/ICG/pDNA (PI/pDNA) complex for more efficient and safer gene transfection. The negatively charged ICG serves a dual purpose: neutralizing PEI's excessive positive charges to reduce cytotoxicity and, under near-infrared irradiation, inducing local heating that enhances cell membrane permeability, thus facilitating the uptake of PI/pDNA complexes to boost transfection efficiency. Using pDNA encoding vascular endothelial growth factor as a model, our system shows enhanced transfection efficiency in vitro for hard-to-transfect endothelial cells, leading to improved cell proliferation and migration. Furthermore, in vivo studies reveal the therapeutic potential of this system in accelerating the healing of infected wounds by promoting angiogenesis and reducing inflammation. This approach offers a straightforward and effective method for gene transfection, showing potentials for tissue engineering and cell-based therapies.
Collapse
Affiliation(s)
- Kunyan Lu
- State and Local Joint Engineering Laboratory for Novel Functional Polymeric Materials, College of Chemistry, Chemical Engineering and Materials Science, Soochow University, Suzhou, 215123, P. R. China
| | - Dongxu Jia
- State and Local Joint Engineering Laboratory for Novel Functional Polymeric Materials, College of Chemistry, Chemical Engineering and Materials Science, Soochow University, Suzhou, 215123, P. R. China
| | - Haixin Zhang
- State and Local Joint Engineering Laboratory for Novel Functional Polymeric Materials, College of Chemistry, Chemical Engineering and Materials Science, Soochow University, Suzhou, 215123, P. R. China
| | - Jingjing Cheng
- State and Local Joint Engineering Laboratory for Novel Functional Polymeric Materials, College of Chemistry, Chemical Engineering and Materials Science, Soochow University, Suzhou, 215123, P. R. China
| | - Yuheng Zhang
- State and Local Joint Engineering Laboratory for Novel Functional Polymeric Materials, College of Chemistry, Chemical Engineering and Materials Science, Soochow University, Suzhou, 215123, P. R. China
| | - Yanxia Zhang
- Department of Cardiovascular Surgery of the First Affiliated Hospital and Institute for Cardiovascular Science, Suzhou Medical College of Soochow University, Soochow University, Suzhou 215007, P. R. China
| | - Qian Yu
- State and Local Joint Engineering Laboratory for Novel Functional Polymeric Materials, College of Chemistry, Chemical Engineering and Materials Science, Soochow University, Suzhou, 215123, P. R. China
| | - Hong Chen
- State and Local Joint Engineering Laboratory for Novel Functional Polymeric Materials, College of Chemistry, Chemical Engineering and Materials Science, Soochow University, Suzhou, 215123, P. R. China
| |
Collapse
|
11
|
He W, Kirmizialtin S. Mechanism of Cationic Lipid Induced DNA Condensation: Lipid-DNA Coordination and Divalent Cation Charge Fluctuations. Biomacromolecules 2024; 25:4819-4830. [PMID: 39011747 PMCID: PMC11323003 DOI: 10.1021/acs.biomac.4c00192] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2024] [Revised: 06/30/2024] [Accepted: 07/01/2024] [Indexed: 07/17/2024]
Abstract
The condensation of nucleic acids by lipids is a widespread phenomenon in biology with crucial implications for drug delivery. However, the mechanisms of DNA assembly in lipid bilayers remain insufficiently understood due to challenges in measuring and assessing each component's contribution in the lipid-DNA-cation system. This study uses all-atom molecular dynamics simulations to investigate DNA condensation in cationic lipid bilayers. Our exhaustive exploration of the thermodynamic factors reveals unique roles for phospholipid head groups and cations. We observed that bridging cations between lipid and DNA drastically reduce charges, while mobile magnesium cations "ping-ponging" between double strands create charge fluctuations. While the first factor stabilizes the DNA-lipid complex, the latter creates attractive forces to induce the spontaneous condensation of DNAs. This novel mechanism not only sheds light on the current data regarding cationic lipid-induced DNA condensation but also provides potential design strategies for creating efficient gene delivery vectors for drug delivery.
Collapse
Affiliation(s)
- Weiwei He
- Chemistry
Program, Science Division, New York University
Abu Dhabi, Abu Dhabi, United Arab Emirates
- Department
of Chemistry, New York University, New York, New York 10003, United States
| | - Serdal Kirmizialtin
- Chemistry
Program, Science Division, New York University
Abu Dhabi, Abu Dhabi, United Arab Emirates
- Department
of Chemistry, New York University, New York, New York 10003, United States
| |
Collapse
|
12
|
Mapfumo P, Reichel LS, André T, Hoeppener S, Rudolph LK, Traeger A. Optimizing Biocompatibility and Gene Delivery with DMAEA and DMAEAm: A Niacin-Derived Copolymer Approach. Biomacromolecules 2024; 25:4749-4761. [PMID: 38963401 PMCID: PMC11323007 DOI: 10.1021/acs.biomac.4c00007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2024] [Revised: 06/14/2024] [Accepted: 06/14/2024] [Indexed: 07/05/2024]
Abstract
Gene therapy is pivotal in nanomedicine, offering a versatile approach to disease treatment. This study aims to achieve an optimal balance between biocompatibility and efficacy, which is a common challenge in the field. A copolymer library is synthesized, incorporating niacin-derived monomers 2-acrylamidoethyl nicotinate (AAEN) or 2-(acryloyloxy)ethyl nicotinate (AEN) with N,N-(dimethylamino)ethyl acrylamide (DMAEAm) or hydrolysis-labile N,N-(dimethylamino)ethyl acrylate (DMAEA). Evaluation of the polymers' cytotoxicity profiles reveals that an increase in AAEN or DMAEA molar ratios correlates with improved biocompatibility. Remarkably, an increase in AAEN in both DMAEA and DMAEAm copolymers demonstrated enhanced transfection efficiencies of plasmid DNA in HEK293T cells. Additionally, the top-performing polymers demonstrate promising gene expression in challenging-to-transfect cells (THP-1 and Jurkat cells) and show no significant effect on modulating immune response induction in ex vivo treated murine monocytes. Overall, the best performing candidates exhibit an optimal balance between biocompatibility and efficacy, showcasing potential advancements in gene therapy.
Collapse
Affiliation(s)
- Prosper
P. Mapfumo
- Institute
of Organic and Macromolecular Chemistry (IOMC), Friedrich Schiller University Jena, Humboldtstrasse 10, Jena 07743, Germany
| | - Liên S. Reichel
- Institute
of Organic and Macromolecular Chemistry (IOMC), Friedrich Schiller University Jena, Humboldtstrasse 10, Jena 07743, Germany
| | - Thomas André
- Leibniz
Institute on Aging-Fritz Lipmann Institute, Jena 07745, Germany
| | - Stephanie Hoeppener
- Institute
of Organic and Macromolecular Chemistry (IOMC), Friedrich Schiller University Jena, Humboldtstrasse 10, Jena 07743, Germany
- Jena
Center for Soft Matter (JCSM), Friedrich
Schiller University Jena, Philosophenweg 7, Jena 07743, Germany
| | | | - Anja Traeger
- Institute
of Organic and Macromolecular Chemistry (IOMC), Friedrich Schiller University Jena, Humboldtstrasse 10, Jena 07743, Germany
- Jena
Center for Soft Matter (JCSM), Friedrich
Schiller University Jena, Philosophenweg 7, Jena 07743, Germany
| |
Collapse
|
13
|
Pereira LFT, Tredus JGR, Corá LO, Novacki LL, Oliveira GED, Vodiani M, Dias IP, Filho RXV, Picheth GF. Advanced biopolymeric materials and nanosystems for RNA/DNA vaccines: a review. Nanomedicine (Lond) 2024; 19:2027-2043. [PMID: 39110059 PMCID: PMC11485706 DOI: 10.1080/17435889.2024.2382077] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2024] [Accepted: 07/16/2024] [Indexed: 10/09/2024] Open
Abstract
The post COVID-19 pandemic era has emerged with more efficient vaccines, all based on genetic materials. However, to expand the use of nucleic components as vaccines, a new generation of nanosystems particularly constructed to increase RNA/DNA stability, half-life and facilitate administration are still required. This review highlights novel developments in mRNA and pDNA vaccines formulated into nanostructures exclusively composed by biopolymeric materials. Recent advances suggest that a new generation of vaccines may arise by adapting the structural features of biopolymers with the effectiveness of nucleic acids. The advantages offered by biopolymers, such as increased stability and targeting ability may cause a revolution in the immunization field for offering promptly adaptable and effective formulations for worldwide distribution.
Collapse
Affiliation(s)
- Luis F T Pereira
- School of Medicine, Pontifical Catholic University of Paraná, Curitiba, PR, Brazil
| | - João G R Tredus
- School of Medicine, Pontifical Catholic University of Paraná, Curitiba, PR, Brazil
| | - Larissa O Corá
- School of Medicine, Pontifical Catholic University of Paraná, Curitiba, PR, Brazil
| | - Luisa L Novacki
- School of Medicine, Federal University of Paraná, Curitiba, PR, Brazil
| | | | - Mariana Vodiani
- Department of Biochemistry, Federal University of Paraná, Curitiba, PR, Brazil
| | - Isabela P Dias
- Department of Biochemistry, Federal University of Paraná, Curitiba, PR, Brazil
| | - Rafael X V Filho
- Department of Biochemistry, Federal University of Paraná, Curitiba, PR, Brazil
| | - Guilherme F Picheth
- School of Medicine, Pontifical Catholic University of Paraná, Curitiba, PR, Brazil
- Department of Biochemistry, Federal University of Paraná, Curitiba, PR, Brazil
| |
Collapse
|
14
|
Shan L, Li Y, Ma Y, Yang Y, Wang J, Peng L, Wang W, Zhao F, Li W, Chen X. Hairpin DNA-Based Nanomaterials for Tumor Targeting and Synergistic Therapy. Int J Nanomedicine 2024; 19:5781-5792. [PMID: 38882546 PMCID: PMC11180469 DOI: 10.2147/ijn.s461774] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2024] [Accepted: 05/29/2024] [Indexed: 06/18/2024] Open
Abstract
Background While nanoplatform-based cancer theranostics have been researched and investigated for many years, enhancing antitumor efficacy and reducing toxic side effects is still an essential problem. Methods We exploited nanoparticle coordination between ferric (Fe2+) ions and telomerase-targeting hairpin DNA structures to encapsulate doxorubicin (DOX) and fabricated Fe2+-DNA@DOX nanoparticles (BDDF NPs). This work studied the NIR fluorescence imaging and pharmacokinetic studies targeting the ability and biodistribution of BDDF NPs. In vitro and vivo studies investigated the nano formula's toxicity, imaging, and synergistic therapeutic effects. Results The enhanced permeability and retention (EPR) effect and tumor targeting resulted in prolonged blood circulation times and high tumor accumulation. Significantly, BDDF NPs could reduce DOX-mediated cardiac toxicity by improving the antioxidation ability of cardiomyocytes based on the different telomerase activities and iron dependency in normal and tumor cells. The synergistic treatment efficacy is enhanced through Fe2+-mediated ferroptosis and the β-catenin/p53 pathway and improved the tumor inhibition rate. Conclusion Harpin DNA-based nanoplatforms demonstrated prolonged blood circulation, tumor drug accumulation via telomerase-targeting, and synergistic therapy to improve antitumor drug efficacy. Our work sheds new light on nanomaterials for future synergistic chemotherapy.
Collapse
Affiliation(s)
- Lingling Shan
- Institute of Pharmaceutical Biotechnology, School of Biology and Food Engineering, Suzhou University, Suzhou, People's Republic of China
| | - Yudie Li
- Institute of Pharmaceutical Biotechnology, School of Biology and Food Engineering, Suzhou University, Suzhou, People's Republic of China
| | - Yifan Ma
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, People's Republic of China
| | - Yang Yang
- Institute of Pharmaceutical Biotechnology, School of Biology and Food Engineering, Suzhou University, Suzhou, People's Republic of China
| | - Jing Wang
- Institute of Pharmaceutical Biotechnology, School of Biology and Food Engineering, Suzhou University, Suzhou, People's Republic of China
| | - Lei Peng
- Institute of Pharmaceutical Biotechnology, School of Biology and Food Engineering, Suzhou University, Suzhou, People's Republic of China
| | - Weiwei Wang
- Institute of Pharmaceutical Biotechnology, School of Biology and Food Engineering, Suzhou University, Suzhou, People's Republic of China
| | - Fang Zhao
- Institute of Pharmaceutical Biotechnology, School of Biology and Food Engineering, Suzhou University, Suzhou, People's Republic of China
| | - Wanrong Li
- Institute of Pharmaceutical Biotechnology, School of Biology and Food Engineering, Suzhou University, Suzhou, People's Republic of China
| | - Xiaoyuan Chen
- Departments of Diagnostic Radiology, Chemical and Biomolecular Engineering, and Biomedical Engineering, Yong Loo Lin School of Medicine and College of Design and Engineering, National University of Singapore, Singapore, Singapore
- Clinical Imaging Research Centre, Centre for Translational Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Nanomedicine Translational Research Program, NUS Center for Nanomedicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Institute of Molecular and Cell Biology, Agency for Science, Technology, and Research (A*STAR), Singapore, Singapore
| |
Collapse
|
15
|
Kashani GK, Naghib SM, Soleymani S, Mozafari MR. A review of DNA nanoparticles-encapsulated drug/gene/protein for advanced controlled drug release: Current status and future perspective over emerging therapy approaches. Int J Biol Macromol 2024; 268:131694. [PMID: 38642693 DOI: 10.1016/j.ijbiomac.2024.131694] [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: 01/14/2024] [Revised: 04/16/2024] [Accepted: 04/17/2024] [Indexed: 04/22/2024]
Abstract
In the last ten years, the field of nanomedicine has experienced significant progress in creating novel drug delivery systems (DDSs). An effective strategy involves employing DNA nanoparticles (NPs) as carriers to encapsulate drugs, genes, or proteins, facilitating regulated drug release. This abstract examines the utilization of DNA NPs and their potential applications in strategies for controlled drug release. Researchers have utilized the distinctive characteristics of DNA molecules, including their ability to self-assemble and their compatibility with living organisms, to create NPs specifically for the purpose of delivering drugs. The DNA NPs possess numerous benefits compared to conventional drug carriers, such as exceptional stability, adjustable dimensions and structure, and convenient customization. Researchers have successfully achieved a highly efficient encapsulation of different therapeutic agents by carefully designing their structure and composition. This advancement enables precise and targeted delivery of drugs. The incorporation of drugs, genes, or proteins into DNA NPs provides notable advantages in terms of augmenting therapeutic effectiveness while reducing adverse effects. DNA NPs serve as a protective barrier for the enclosed payloads, preventing their degradation and extending their duration in the body. The protective effect is especially vital for delicate biologics, such as proteins or gene-based therapies that could otherwise be vulnerable to enzymatic degradation or quick elimination. Moreover, the surface of DNA NPs can be altered to facilitate specific targeting towards particular tissues or cells, thereby augmenting the accuracy of delivery. A significant benefit of DNA NPs is their capacity to regulate the kinetics of drug release. Through the manipulation of the DNA NPs structure, scientists can regulate the rate at which the enclosed cargo is released, enabling a prolonged and regulated dispensation of medication. This control is crucial for medications with limited therapeutic ranges or those necessitating uninterrupted administration to attain optimal therapeutic results. In addition, DNA NPs have the ability to react to external factors, including alterations in temperature, pH, or light, which can initiate the release of the payload at precise locations or moments. This feature enhances the precision of drug release control. The potential uses of DNA NPs in the controlled release of medicines are extensive. The NPs have the ability to transport various therapeutic substances, for example, drugs, peptides, NAs (NAs), and proteins. They exhibit potential for the therapeutic management of diverse ailments, including cancer, genetic disorders, and infectious diseases. In addition, DNA NPs can be employed for targeted drug delivery, traversing biological barriers, and surpassing the constraints of conventional drug administration methods.
Collapse
Affiliation(s)
- Ghazal Kadkhodaie Kashani
- Nanotechnology Department, School of Advanced Technologies, Iran University of Science and Technology (IUST), Tehran 1684613114, Iran
| | - Seyed Morteza Naghib
- Nanotechnology Department, School of Advanced Technologies, Iran University of Science and Technology (IUST), Tehran 1684613114, Iran.
| | - Sina Soleymani
- Nanotechnology Department, School of Advanced Technologies, Iran University of Science and Technology (IUST), Tehran 1684613114, Iran; Australasian Nanoscience and Nanotechnology Initiative (ANNI), Monash University LPO, Clayton, VIC 3168, Australia; Biomaterials and Tissue Engineering Research Group, Interdisciplinary Technologies Department, Breast Cancer Research Center, Motamed Cancer Institute, ACECR, Iran University of Science and Technology (IUST), Tehran, Iran
| | - M R Mozafari
- Australasian Nanoscience and Nanotechnology Initiative (ANNI), Monash University LPO, Clayton, VIC 3168, Australia
| |
Collapse
|
16
|
Oikeh E, Ziebarth J, Dinar MAM, Kirchhoff D, Aronova A, Dziubla TD, Wang Y, DeRouchey JE. DNA Packaging and Polycation Length Determine DNA Susceptibility to Free Radical Damage in Condensed DNA. J Phys Chem B 2024; 128:3329-3339. [PMID: 38557033 DOI: 10.1021/acs.jpcb.3c06116] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/04/2024]
Abstract
In nature, DNA exists primarily in a highly compacted form. The compaction of DNA in vivo is mediated by cationic proteins: histones in somatic nuclei and protamines in sperm chromatin. The extreme, nearly crystalline packaging of DNA by protamines in spermatozoa is thought to be essential for both efficient genetic delivery as well as DNA protection against damage by mutagens and oxidative species. The protective role of protamines is required in sperm, as they are sensitive to ROS damage due to the progressive loss of DNA repair mechanisms during maturation. The degree to which DNA packaging directly relates to DNA protection in the condensed state, however, is poorly understood. Here, we utilized different polycation condensing agents to achieve varying DNA packaging densities and quantify DNA damage by free radical oxidation within the condensates. Although we see that tighter DNA packaging generally leads to better protection, the length of the polycation also plays a significant role. Molecular dynamics simulations suggest that longer polyarginine chains offer increased protection by occupying more space on the DNA surface and forming more stable interactions. Taken together, our results suggest a complex interplay among polycation properties, DNA packaging density, and DNA protection against free radical damage within condensed states.
Collapse
Affiliation(s)
- Ehigbai Oikeh
- Department of Chemistry, University of Kentucky, Lexington, Kentucky 40506, United States
| | - Jesse Ziebarth
- Department of Chemistry, University of Memphis, Memphis, Tennessee 38152, United States
| | - Md Abu Monsur Dinar
- Department of Chemistry, University of Kentucky, Lexington, Kentucky 40506, United States
| | - Daniel Kirchhoff
- Department of Chemistry, University of Kentucky, Lexington, Kentucky 40506, United States
| | - Anastasiia Aronova
- Chemical and Materials Engineering Department, University of Kentucky, Lexington, Kentucky 40506, United States
| | - Thomas D Dziubla
- Chemical and Materials Engineering Department, University of Kentucky, Lexington, Kentucky 40506, United States
| | - Yongmei Wang
- Department of Chemistry, University of Memphis, Memphis, Tennessee 38152, United States
| | - Jason E DeRouchey
- Department of Chemistry, University of Kentucky, Lexington, Kentucky 40506, United States
| |
Collapse
|
17
|
Eusébio D, Paul M, Biswas S, Cui Z, Costa D, Sousa Â. Mannosylated polyethylenimine-cholesterol-based nanoparticles for targeted delivery of minicircle DNA vaccine against COVID-19 to antigen-presenting cells. Int J Pharm 2024; 654:123959. [PMID: 38430949 DOI: 10.1016/j.ijpharm.2024.123959] [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: 06/19/2023] [Revised: 01/25/2024] [Accepted: 02/28/2024] [Indexed: 03/05/2024]
Abstract
DNA vaccines can be a potential solution to protect global health, triggering both humoral and cellular immune responses. DNA vaccines are valuable in preventing intracellular pathogen infections, and therefore can be explored against coronavirus disease (COVID-19) caused by severe acute respiratory syndrome coronavirus (SARS-CoV-2). This work explored different systems based on polyethylenimine (PEI), functionalized for the first time with both cholesterol (CHOL) and mannose (MAN) to deliver parental plasmid (PP) and minicircle DNA (mcDNA) vectors encoding the receptor-binding domain (RBD) of SARS-CoV-2 to antigen-presenting cells (APCs). For comparative purposes, three different systems were evaluated: PEI, PEI-CHOL and PEI-CHOL-MAN. The systems were prepared at various nitrogen-to-phosphate group (N/P) ratios and characterized in terms of encapsulation efficiency, surface charge, size, polydispersity index (PDI), morphology, and stability over time. Moreover, in vitro transfection studies of dendritic cells (JAWS II) and human fibroblast cells were performed. Viability studies assured the biocompatibility of all nanocarriers. Confocal microscopy studies confirmed intracellular localization of systems, resulting in enhanced cellular uptake using PEI-CHOL and PEI-CHOL-MAN systems when compared with the PEI system. Regarding the RBD expression, PEI-CHOL-MAN was the system that led to the highest levels of transcripts and protein expression in JAWS II cells. Furthermore, the nanosystems significantly stimulated pro-inflammatory cytokines production and dendritic cell maturation in vitro. Overall, mannosylated systems can be considered a valuable tool in the delivery of plasmid DNA or mcDNA vaccines to APCs.
Collapse
Affiliation(s)
- Dalinda Eusébio
- CICS-UBI - Health Sciences Research Centre, University of Beira Interior, Covilhã, Portugal
| | - Milan Paul
- Nanomedicine Research Laboratory, Department of Pharmacy, Birla Institute of Technology and Science-Pilani, Hyderabad Campus. Jawahar Nagar, Medchal, Hyderabad 500078, India
| | - Swati Biswas
- Nanomedicine Research Laboratory, Department of Pharmacy, Birla Institute of Technology and Science-Pilani, Hyderabad Campus. Jawahar Nagar, Medchal, Hyderabad 500078, India
| | - Zhengrong Cui
- The University of Texas at Austin, College of Pharmacy, Division of Molecular Pharmaceutics and Drug Delivery, Austin, TX 78712, USA
| | - Diana Costa
- CICS-UBI - Health Sciences Research Centre, University of Beira Interior, Covilhã, Portugal
| | - Ângela Sousa
- CICS-UBI - Health Sciences Research Centre, University of Beira Interior, Covilhã, Portugal.
| |
Collapse
|
18
|
Albuquerque LJC, de Oliveira FA, Christoffolete MA, Nascimento-Sales M, Berger S, Wagner E, Lächelt U, Giacomelli FC. Nucleic acid delivery to retinal cells using lipopeptides as a potential tool towards ocular gene therapies. J Colloid Interface Sci 2024; 655:346-356. [PMID: 37948808 DOI: 10.1016/j.jcis.2023.11.003] [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: 08/16/2023] [Revised: 10/11/2023] [Accepted: 11/01/2023] [Indexed: 11/12/2023]
Abstract
We evaluated the use of lipopeptides capable to bind to nucleic acids towards plasmid DNA (pDNA) delivery. The investigations were particularly focused on arising retinal pigment epithelial cells (ARPE-19) as motivated by the considerable number of ocular disorders linked to gene aberrations. The lipopeptides comprised the artificial oligoamino acid succinyl-tetraethylene pentamine (Stp) as well as incorporated lysines, histidines, cysteines, fatty acids, and tyrosine trimers. Regardless of the structural differences, the lipopeptides demonstrated to efficiently condense pDNA at nitrogen-to-phosphate molar ratio (N/P) ≥ 6. Spheric nanoparticles were observed by cryo-TEM and dynamic light scattering determined hydrodynamic sizes ranging from 50 to 130 nm. The biological assays evidenced highly efficient pDNA delivery with a lower degree of cytotoxicity compared to the well-known transfecting agent linear polyethylenimine (LPEI). Although more efficient than LPEI, cysteine-containing carriers were demonstrated to be less efficient than the other counterparts possibly due to exceeding polyplex stabilization via disulfide cross links, which could hamper pDNA unpacking at the target site. Therefore, clearly a balance between complex stability and cargo release should be taken into account to optimize the transfection efficiency of the non-viral vectors. The gene transfer activity in ARPE-19 cells suggests the applicability of this kind of carrier for ocular treatments based on retinal gene delivery.
Collapse
Affiliation(s)
| | | | | | | | - Simone Berger
- Department of Pharmacy and Center for NanoScience (CeNs), Ludwig-Maximilians-Universität, Munich, Germany
| | - Ernst Wagner
- Department of Pharmacy and Center for NanoScience (CeNs), Ludwig-Maximilians-Universität, Munich, Germany
| | - Ulrich Lächelt
- Department of Pharmacy and Center for NanoScience (CeNs), Ludwig-Maximilians-Universität, Munich, Germany; Department of Pharmaceutical Sciences, University of Vienna, Vienna, Austria
| | - Fernando C Giacomelli
- Centro de Ciências Naturais e Humanas, Universidade Federal do ABC, Santo André, Brazil.
| |
Collapse
|
19
|
Porello I, Bono N, Candiani G, Cellesi F. Advancing nucleic acid delivery through cationic polymer design: non-cationic building blocks from the toolbox. Polym Chem 2024; 15:2800-2826. [DOI: 10.1039/d4py00234b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
Abstract
The rational integration of non-cationic building blocks into cationic polymers can be devised to enhance the performance of the resulting gene delivery vectors, improving cell targeting behavior, uptake, endosomal escape, toxicity, and transfection efficiency.
Collapse
Affiliation(s)
- Ilaria Porello
- Department of Chemistry, Materials and Chemical Engineering “G. Natta”, Politecnico di Milano, Via Luigi Mancinelli 7, 20131, Milan, Italy
| | - Nina Bono
- Department of Chemistry, Materials and Chemical Engineering “G. Natta”, Politecnico di Milano, Via Luigi Mancinelli 7, 20131, Milan, Italy
| | - Gabriele Candiani
- Department of Chemistry, Materials and Chemical Engineering “G. Natta”, Politecnico di Milano, Via Luigi Mancinelli 7, 20131, Milan, Italy
| | - Francesco Cellesi
- Department of Chemistry, Materials and Chemical Engineering “G. Natta”, Politecnico di Milano, Via Luigi Mancinelli 7, 20131, Milan, Italy
| |
Collapse
|
20
|
Franck CO, Bistrovic Popov A, Ahmed I, Hewitt RE, Franslau L, Tyagi P, Fruk L. A catch-and-release nano-based gene delivery system. NANOSCALE HORIZONS 2023; 8:1588-1594. [PMID: 37691551 DOI: 10.1039/d3nh00269a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/12/2023]
Abstract
The design of nanomaterial-based nucleic acid formulations is one of the biggest endeavours in the search for clinically applicable gene delivery systems. Biopolymers represent a promising subclass of gene carriers due to their physicochemical properties, biodegradability and biocompatibility. By modifying melanin-like polydopamine nanoparticles with poly-L-arginine and poly-L-histidine blends, we obtained a novel catch-and-release gene delivery system for efficient trafficking of pDNA to human cells. A synergistic interplay of nanoparticle-bound poly-L-arginine and poly-L-histidine was observed and evaluated for pDNA binding affinity, cell viability, gene release and transfection. Although the functionalisation with poly-L-arginine was crucial for pDNA binding, the resulting nanocarriers failed to release pDNA intracellularly, resulting in limited protein expression. However, optimal pDNA release was achieved through the co-formulation with poly-L-histidine, essential for pDNA release. This effect enabled the design of gene delivery systems, which were comparable to Lipofectamine in terms of transfection efficacy and the catch-and-release surface modification strategy can be translated to other nanocarriers and surfaces.
Collapse
Affiliation(s)
- Christoph O Franck
- BioNano Engineering Lab, Department of Chemical Engineering and Biotechnology, University of Cambridge, Philippa Fawcett Drive, Cambridge CB3 0AS, UK.
| | - Andrea Bistrovic Popov
- BioNano Engineering Lab, Department of Chemical Engineering and Biotechnology, University of Cambridge, Philippa Fawcett Drive, Cambridge CB3 0AS, UK.
| | - Ishtiaq Ahmed
- BioNano Engineering Lab, Department of Chemical Engineering and Biotechnology, University of Cambridge, Philippa Fawcett Drive, Cambridge CB3 0AS, UK.
| | - Rachel E Hewitt
- Department of Veterinary Medicine, University of Cambridge, Madingley Rd, Cambridge CB3 0ES, UK
| | - Luise Franslau
- Institut für Physikalische Chemie, Georg-August-Universität Göttingen, Tammanstraße 6, Göttingen 37077, Germany
| | - Puneet Tyagi
- AstraZeneca, One MedImmune Way, Gaithersburg, MD 20878, USA
| | - Ljiljana Fruk
- BioNano Engineering Lab, Department of Chemical Engineering and Biotechnology, University of Cambridge, Philippa Fawcett Drive, Cambridge CB3 0AS, UK.
| |
Collapse
|
21
|
Casper J, Schenk SH, Parhizkar E, Detampel P, Dehshahri A, Huwyler J. Polyethylenimine (PEI) in gene therapy: Current status and clinical applications. J Control Release 2023; 362:667-691. [PMID: 37666302 DOI: 10.1016/j.jconrel.2023.09.001] [Citation(s) in RCA: 72] [Impact Index Per Article: 36.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2023] [Revised: 08/24/2023] [Accepted: 09/01/2023] [Indexed: 09/06/2023]
Abstract
Polyethlyenimine (PEI) was introduced 1995 as a cationic polymer for nucleic acid delivery. PEI and its derivatives are extensively used in basic research and as reference formulations in the field of polymer-based gene delivery. Despite its widespread use, the number of clinical applications to date is limited. Thus, this review aims to consolidate the past applications of PEI in DNA delivery, elucidate the obstacles that hinder its transition to clinical use, and highlight potential prospects for novel iterations of PEI derivatives. The present review article is divided into three sections. The first section examines the mechanism of action employed by PEI, examining fundamental aspects of cellular delivery including uptake mechanisms, release from endosomes, and transport into the cell nucleus, along with potential strategies for enhancing these delivery phases. Moreover, an in-depth analysis is conducted concerning the mechanism underlying cellular toxicity, accompanied with approaches to overcome this major challenge. The second part is devoted to the in vivo performance of PEI and its application in various therapeutic indications. While systemic administration has proven to be challenging, alternative localized delivery routes hold promise, such as treatment of solid tumors, application as a vaccine, or serving as a therapeutic agent for pulmonary delivery. In the last section, the outcome of completed and ongoing clinical trials is summarized. Finally, an expert opinion is provided on the potential of PEI and its future applications. PEI-based formulations for nucleic acid delivery have a promising potential, it will be an important task for the years to come to introduce innovations that address PEI-associated shortcomings by introducing well-designed PEI formulations in combination with an appropriate route of administration.
Collapse
Affiliation(s)
- Jens Casper
- Division of Pharmaceutical Technology, Department of Pharmaceutical Sciences, University of Basel, Klingelbergstrasse 50, 4056 Basel, Switzerland
| | - Susanne H Schenk
- Division of Pharmaceutical Technology, Department of Pharmaceutical Sciences, University of Basel, Klingelbergstrasse 50, 4056 Basel, Switzerland
| | - Elahehnaz Parhizkar
- Department of Pharmaceutics, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Pascal Detampel
- Division of Pharmaceutical Technology, Department of Pharmaceutical Sciences, University of Basel, Klingelbergstrasse 50, 4056 Basel, Switzerland
| | - Ali Dehshahri
- Department of Pharmaceutical Biotechnology, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran.
| | - Jörg Huwyler
- Division of Pharmaceutical Technology, Department of Pharmaceutical Sciences, University of Basel, Klingelbergstrasse 50, 4056 Basel, Switzerland.
| |
Collapse
|
22
|
Spitzbarth B, Eelkema R. Chemical reaction networks based on conjugate additions on β'-substituted Michael acceptors. Chem Commun (Camb) 2023; 59:11174-11187. [PMID: 37529876 PMCID: PMC10508045 DOI: 10.1039/d3cc02126b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2023] [Accepted: 07/25/2023] [Indexed: 08/03/2023]
Abstract
Over the last few decades, the study of more complex, chemical systems closer to those found in nature, and the interactions within those systems, has grown immensely. Despite great efforts, the need for new, versatile, and robust chemistry to apply in CRNs remains. In this Feature Article, we give a brief overview over previous developments in the field of systems chemistry and how β'-substituted Michael acceptors (MAs) can be a great addition to the systems chemist's toolbox. We illustrate their versatility by showcasing a range of examples of applying β'-substituted MAs in CRNs, both as chemical signals and as substrates, to open up the path to many applications ranging from responsive materials, to pathway control in CRNs, drug delivery, analyte detection, and beyond.
Collapse
Affiliation(s)
- Benjamin Spitzbarth
- Department of Chemical Engineering, Delft University of Technology, Van der Maasweg 9, 2629 HZ Delft, The Netherlands.
| | - Rienk Eelkema
- Department of Chemical Engineering, Delft University of Technology, Van der Maasweg 9, 2629 HZ Delft, The Netherlands.
| |
Collapse
|
23
|
de Oliveira FA, Albuquerque LJC, Nascimento-Sales M, Christoffolete MA, Bellettini IC, Giacomelli FC. Balancing gene transfection and cytotoxicity of nucleic acid carriers with focus on ocular and hepatic disorders: evaluation of hydrophobic and hydrophilic polyethyleneimine derivatives. J Mater Chem B 2023; 11:4556-4571. [PMID: 37161773 DOI: 10.1039/d3tb00477e] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/11/2023]
Abstract
Polyethyleneimine (PEI) derivatives substituted by lactose, succinic acid or alkyl domains were evaluated as nonviral gene delivery vectors towards balancing gene transfection and cytotoxicity. The investigations were focused on pDNA transfection into arising retinal pigment epithelia (ARPE-19) and human hepatocellular carcinoma (HepG2) cell lines. The first mentioned cell line was chosen as motivated by the non-negligible number of ocular disorders linked to gene aberrations, whereas the second one is a cell line overexpressing the asialoglycoprotein receptor (ASGP-R), which can bind to galactose residues. The presence of short alkyl domains (C4 and C6), and particularly the succinylation of the PEI chains, improved the biological outputs of the gene vectors. The presence of hydrophobic units possibly enhances lytic activity, whereas the incorporation of succinic acid slightly reduces polymer-DNA interaction strength, thereby enabling more efficient intracellular unpacking and cargo release. Succinylation is also supposed to decrease cytotoxicity and avoid protein adsorption to the polyplexes. The presence of long carbon chains (for instance, C12) nevertheless, results in higher levels of cytotoxicity and respective lower transfection rates. The sugar-decorated polyplexes are overall less cytotoxic, but the presence of lactose moieties also leads to larger polyplexes and notably weak polymer-DNA binding, which compromise the transfection efficiency. Yet, along with the presence of short lytic alkyl domains, the double-substitution of PEI synergistically boosts gene transfection probably due to the uptake of higher DNA and polymer amounts without cell damage. Overall, the experimental data suggest that ocular and hepatic gene therapies may be potentialized by fine-tuning the hydrophobic-to-hydrophilic balance, and succinic acid is a favorable motif for the modification of PEI.
Collapse
Affiliation(s)
- Fernando A de Oliveira
- Centro de Ciências Naturais e Humanas, Universidade Federal do ABC, Santo André, Brazil.
| | | | | | | | - Ismael C Bellettini
- Departamento de Ciências Exatas e Educação, Universidade Federal de Santa Catarina, Blumenau, Brazil
| | - Fernando C Giacomelli
- Centro de Ciências Naturais e Humanas, Universidade Federal do ABC, Santo André, Brazil.
| |
Collapse
|
24
|
Zhang X, Liu Y, Xiao C, Guan Y, Gao Z, Huang W. Research Advances in Nucleic Acid Delivery System for Rheumatoid Arthritis Therapy. Pharmaceutics 2023; 15:1237. [PMID: 37111722 PMCID: PMC10145518 DOI: 10.3390/pharmaceutics15041237] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Revised: 03/31/2023] [Accepted: 04/05/2023] [Indexed: 04/29/2023] Open
Abstract
Rheumatoid arthritis (RA) is a chronic inflammatory autoimmune disease that affects the lives of nearly 1% of the total population worldwide. With the understanding of RA, more and more therapeutic drugs have been developed. However, lots of them possess severe side effects, and gene therapy may be a potential method for RA treatment. A nanoparticle delivery system is vital for gene therapy, as it can keep the nucleic acids stable and enhance the efficiency of transfection in vivo. With the development of materials science, pharmaceutics and pathology, more novel nanomaterials and intelligent strategies are applied to better and safer gene therapy for RA. In this review, we first summarized the existing nanomaterials and active targeting ligands used for RA gene therapy. Then, we introduced various gene delivery systems for RA treatment, which may enlighten the relevant research in the future.
Collapse
Affiliation(s)
- Xintong Zhang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China; (X.Z.); (Y.L.); (C.X.)
- Beijing Key Laboratory of Drug Delivery Technology and Novel Formulations, Department of Pharmaceutics, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Yanhong Liu
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China; (X.Z.); (Y.L.); (C.X.)
- Beijing Key Laboratory of Drug Delivery Technology and Novel Formulations, Department of Pharmaceutics, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Congcong Xiao
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China; (X.Z.); (Y.L.); (C.X.)
- Beijing Key Laboratory of Drug Delivery Technology and Novel Formulations, Department of Pharmaceutics, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Youyan Guan
- Department of Urology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China;
| | - Zhonggao Gao
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China; (X.Z.); (Y.L.); (C.X.)
- Beijing Key Laboratory of Drug Delivery Technology and Novel Formulations, Department of Pharmaceutics, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Wei Huang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China; (X.Z.); (Y.L.); (C.X.)
- Beijing Key Laboratory of Drug Delivery Technology and Novel Formulations, Department of Pharmaceutics, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| |
Collapse
|
25
|
Liu J, Zhang J, Gao Y, Jiang Y, Guan Z, Xie Y, Hu J, Chen J. Barrier permeation and improved nanomedicine delivery in tumor microenvironments. Cancer Lett 2023; 562:216166. [PMID: 37028698 DOI: 10.1016/j.canlet.2023.216166] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Revised: 03/10/2023] [Accepted: 04/01/2023] [Indexed: 04/09/2023]
Abstract
Nanomedicines can effectively penetrate tumor sites compared to traditionally used drugs. However, effective drugs that reach the interior of tumors remain limited. Based on studies of the complex tumor microenvironment, we summarized the barriers restricting tumor penetration of nanomedicines in this review. Penetration barriers are mainly caused by tumor blood vessels, stroma, and cell abnormalities. The repair of abnormal tumor blood vessels and tumor stroma and adjusting the physicochemical properties of nanoparticles are considered promising strategies to improve the tumor permeation of nanomedicines. The effects of nanoparticle properties, including size, shape, and surface charge, on tumor penetration were also reviewed. We expect to provide research ideas and a scientific basis for nanomedicines to increase intratumoral permeability and improve anti-tumor effects.
Collapse
Affiliation(s)
- Jinxiang Liu
- School of Pharmacy, The Key Laboratory of Prescription Effect and Clinical Evaluation of State Administration of Traditional Chinese Medicine of China, Binzhou Medical University, Yantai, 264003, PR China
| | - Jiaying Zhang
- School of Pharmacy, The Key Laboratory of Prescription Effect and Clinical Evaluation of State Administration of Traditional Chinese Medicine of China, Binzhou Medical University, Yantai, 264003, PR China
| | - Yang Gao
- School of Pharmacy, The Key Laboratory of Prescription Effect and Clinical Evaluation of State Administration of Traditional Chinese Medicine of China, Binzhou Medical University, Yantai, 264003, PR China; School of Food Science and Pharmaceutical Engineering, Nanjing Normal University, Nanjing, 210023, PR China
| | - Yuxuan Jiang
- School of Pharmacy, The Key Laboratory of Prescription Effect and Clinical Evaluation of State Administration of Traditional Chinese Medicine of China, Binzhou Medical University, Yantai, 264003, PR China
| | - Zhenxin Guan
- School of Pharmacy, The Key Laboratory of Prescription Effect and Clinical Evaluation of State Administration of Traditional Chinese Medicine of China, Binzhou Medical University, Yantai, 264003, PR China
| | - Yiying Xie
- School of Pharmacy, The Key Laboratory of Prescription Effect and Clinical Evaluation of State Administration of Traditional Chinese Medicine of China, Binzhou Medical University, Yantai, 264003, PR China
| | - Jinghui Hu
- School of Rehabilitation, Institute of Rehabilitation Engineering, Binzhou Medical University, Yantai, 264003, PR China.
| | - Jing Chen
- School of Pharmacy, The Key Laboratory of Prescription Effect and Clinical Evaluation of State Administration of Traditional Chinese Medicine of China, Binzhou Medical University, Yantai, 264003, PR China.
| |
Collapse
|
26
|
Zhang Y, Xian H, Strounina E, Gunther KS, Sweet MJ, Chen C, Yu C, Wang Y. Mesoporous Organosilica Nanoparticles with Tetrasulphide Bond to Enhance Plasmid DNA Delivery. Pharmaceutics 2023; 15:1013. [DOI: https:/doi.org/10.3390/pharmaceutics15031013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/14/2024] Open
Abstract
Cellular delivery of plasmid DNA (pDNA) specifically into dendritic cells (DCs) has provoked wide attention in various applications. However, delivery tools that achieve effective pDNA transfection in DCs are rare. Herein, we report that tetrasulphide bridged mesoporous organosilica nanoparticles (MONs) have enhanced pDNA transfection performance in DC cell lines compared to conventional mesoporous silica nanoparticles (MSNs). The mechanism of enhanced pDNA delivery efficacy is attributed to the glutathione (GSH) depletion capability of MONs. Reduction of initially high GSH levels in DCs further increases the mammalian target of rapamycin complex 1 (mTORc1) pathway activation, enhancing translation and protein expression. The mechanism was further validated by showing that the increased transfection efficiency was apparent in high GSH cell lines but not in low GSH ones. Our findings may provide a new design principle of nano delivery systems where the pDNA delivery to DCs is important.
Collapse
Affiliation(s)
- Yue Zhang
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, QLD 4072, Australia
| | - He Xian
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Ekaterina Strounina
- Centre for Advanced Imaging, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Kimberley S. Gunther
- Institute for Molecular Bioscience (IMB), IMB Centre for Inflammation and Disease Research, and Australian Infectious Diseases Research Centre, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Matthew J. Sweet
- Institute for Molecular Bioscience (IMB), IMB Centre for Inflammation and Disease Research, and Australian Infectious Diseases Research Centre, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Chen Chen
- School of Biomedical Sciences, Faculty of Medicine, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Chengzhong Yu
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Yue Wang
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, QLD 4072, Australia
| |
Collapse
|
27
|
Zhang Y, Xian H, Strounina E, Gunther KS, Sweet MJ, Chen C, Yu C, Wang Y. Mesoporous Organosilica Nanoparticles with Tetrasulphide Bond to Enhance Plasmid DNA Delivery. Pharmaceutics 2023; 15:pharmaceutics15031013. [PMID: 36986873 PMCID: PMC10053670 DOI: 10.3390/pharmaceutics15031013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Revised: 02/08/2023] [Accepted: 03/14/2023] [Indexed: 03/30/2023] Open
Abstract
Cellular delivery of plasmid DNA (pDNA) specifically into dendritic cells (DCs) has provoked wide attention in various applications. However, delivery tools that achieve effective pDNA transfection in DCs are rare. Herein, we report that tetrasulphide bridged mesoporous organosilica nanoparticles (MONs) have enhanced pDNA transfection performance in DC cell lines compared to conventional mesoporous silica nanoparticles (MSNs). The mechanism of enhanced pDNA delivery efficacy is attributed to the glutathione (GSH) depletion capability of MONs. Reduction of initially high GSH levels in DCs further increases the mammalian target of rapamycin complex 1 (mTORc1) pathway activation, enhancing translation and protein expression. The mechanism was further validated by showing that the increased transfection efficiency was apparent in high GSH cell lines but not in low GSH ones. Our findings may provide a new design principle of nano delivery systems where the pDNA delivery to DCs is important.
Collapse
Affiliation(s)
- Yue Zhang
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, QLD 4072, Australia
| | - He Xian
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Ekaterina Strounina
- Centre for Advanced Imaging, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Kimberley S Gunther
- Institute for Molecular Bioscience (IMB), IMB Centre for Inflammation and Disease Research, and Australian Infectious Diseases Research Centre, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Matthew J Sweet
- Institute for Molecular Bioscience (IMB), IMB Centre for Inflammation and Disease Research, and Australian Infectious Diseases Research Centre, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Chen Chen
- School of Biomedical Sciences, Faculty of Medicine, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Chengzhong Yu
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Yue Wang
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, QLD 4072, Australia
| |
Collapse
|
28
|
Eswaran L, Kazimirsky G, Yehuda R, Byk G. A New Strategy for Nucleic Acid Delivery and Protein Expression Using Biocompatible Nanohydrogels of Predefined Sizes. Pharmaceutics 2023; 15:pharmaceutics15030961. [PMID: 36986821 PMCID: PMC10058534 DOI: 10.3390/pharmaceutics15030961] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Revised: 03/12/2023] [Accepted: 03/14/2023] [Indexed: 03/19/2023] Open
Abstract
We have developed new formulations of nanohydrogels (NHGs) complexed with DNA devoid of cell toxicity, which, together with their tuned sizes, makes them of great interest for delivering DNA/RNA for foreign protein expression. Transfection results demonstrate that, unlike classical lipo/polyplexes, the new NHGs can be incubated indefinitely with cells without apparent cellular toxicity, resulting in the high expression of foreign proteins for long periods of time. Although protein expression starts with a delay as compared to classical systems, it is sustained for a long period of time, even after passing cells without observation of toxicity. A fluorescently labelled NHG used for gene delivery was detected inside cells very early after incubation, but the protein expression was delayed by many days, demonstrating that there is a time-dependent release of genes from the NHGs. We suggest that this delay is due to the slow but continuous release of DNA from the particles concomitantly with slow but continuous protein expression. Additionally, results obtained after the in vivo administration of m-Cherry/NHG complexes indicated a delayed but prolonged expression of the marker gene in the tissue of administration. Overall, we have demonstrated gene delivery and foreign protein expression using GFP and m-Cherry marker genes complexed with biocompatible nanohydrogels.
Collapse
|
29
|
Yi Y, Yu M, Li W, Zhu D, Mei L, Ou M. Vaccine-like nanomedicine for cancer immunotherapy. J Control Release 2023; 355:760-778. [PMID: 36822241 DOI: 10.1016/j.jconrel.2023.02.015] [Citation(s) in RCA: 69] [Impact Index Per Article: 34.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Revised: 02/07/2023] [Accepted: 02/09/2023] [Indexed: 02/25/2023]
Abstract
The successful clinical application of immune checkpoint blockade (ICB) and chimeric antigen receptor T cells (CAR-T) therapeutics has attracted extensive attention to immunotherapy, however, their drawbacks such as limited specificity, persistence and toxicity haven't met the high expectations on efficient cancer treatments. Therapeutic cancer vaccines which instruct the immune system to capture tumor specific antigens, generate long-term immune memory and specifically eliminate cancer cells gradually become the most promising strategies to eradicate tumor. However, the disadvantages of some existing vaccines such as weak immunogenicity and in vivo instability have restricted their development. Nanotechnology has been recently incorporated into vaccine fabrication and exhibited promising results for cancer immunotherapy. Nanoparticles promote the stability of vaccines, as well as enhance antigen recognition and presentation owing to their nanometer size which promotes internalization of antigens by phagocytic cells. The surface modification with targeting units further permits the delivery of vaccines to specific cells. Meanwhile, nanocarriers with adjuvant effect can improve the efficacy of vaccines. In addition to classic vaccines composed of antigens and adjuvants, the nanoparticle-mediated chemotherapy, radiotherapy and certain other therapeutics could induce the release of tumor antigens in situ, which therefore effectively simulate antitumor immune responses. Such vaccine-like nanomedicine not only kills primary tumors, but also prevents tumor recurrence and helps eliminate metastatic tumors. Herein, we introduce recent developments in nanoparticle-based delivery systems for antigen delivery and in situ antitumor vaccination. We will also discuss the remaining opportunities and challenges of nanovaccine in clinical translation towards cancer treatment.
Collapse
Affiliation(s)
- Yunfei Yi
- Tianjin Key Laboratory of Biomedical Materials, Key Laboratory of Biomaterials and Nanotechnology for Cancer Immunotherapy, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 300192, China; School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen 518107, China
| | - Mian Yu
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen 518107, China
| | - Wen Li
- Tianjin Key Laboratory of Biomedical Materials, Key Laboratory of Biomaterials and Nanotechnology for Cancer Immunotherapy, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 300192, China
| | - Dunwan Zhu
- Tianjin Key Laboratory of Biomedical Materials, Key Laboratory of Biomaterials and Nanotechnology for Cancer Immunotherapy, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 300192, China.
| | - Lin Mei
- Tianjin Key Laboratory of Biomedical Materials, Key Laboratory of Biomaterials and Nanotechnology for Cancer Immunotherapy, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 300192, China.
| | - Meitong Ou
- Tianjin Key Laboratory of Biomedical Materials, Key Laboratory of Biomaterials and Nanotechnology for Cancer Immunotherapy, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 300192, China.
| |
Collapse
|
30
|
Wong KH, Guo Z, Law MK, Chen M. Functionalized PAMAM constructed nanosystems for biomacromolecule delivery. Biomater Sci 2023; 11:1589-1606. [PMID: 36692071 DOI: 10.1039/d2bm01677j] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Polyamidoamines (PAMAMs) are a class of dendrimer with monodispersity and controlled topology, which can deliver biologically active macromolecules (e.g., genes and proteins) to specific regions with high efficiency and minimum side effects. In detail, PAMAMs can be functionalized easily by core modification or surface amendment to encapsulate a wide range of biomacromolecules. Besides, self-assembled, cross-linked and hybrid PAMAMs with customized therapeutic purposes are developed as delivery vehicles, which makes PAMAMs promising for biomacromolecule therapy. In this review, we comprehensively summarize the application of PAMAMs in biomacromolecule delivery from the synthesis of functionalized PAMAM carriers to the development of PAMAM-based drug delivery systems. The underlying strategies for PAMAM functionalization and assembly are first systematically discussed, and then the current applications of PAMAMs for biomacromolecule delivery are reviewed. Finally, a brief perspective on the further applications of PAMAMs concludes, aiming to provide insights into developing PAMAM-based biomacromolecule delivery systems.
Collapse
Affiliation(s)
- Ka Hong Wong
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau SAR, China.
| | - Zhaopei Guo
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau SAR, China.
| | - Man-Kay Law
- State Key Laboratory of Analog and Mixed-Signal VLSI, IME and FST-ECE, University of Macau, Macau SAR, China
| | - Meiwan Chen
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau SAR, China.
| |
Collapse
|
31
|
Wang N, Liu C, Li Y, Huang D, Wu X, Kou X, Wang X, Wu Q, Gong C. A cooperative nano-CRISPR scaffold potentiates immunotherapy via activation of tumour-intrinsic pyroptosis. Nat Commun 2023; 14:779. [PMID: 36774382 PMCID: PMC9922300 DOI: 10.1038/s41467-023-36550-9] [Citation(s) in RCA: 42] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Accepted: 02/07/2023] [Indexed: 02/13/2023] Open
Abstract
Efficient cancer immunotherapy depends on selective targeting of high bioactivity therapeutic agents to the tumours. However, delivering exogenous medication might prove difficult in clinical practice. Here we report a cooperative Nano-CRISPR scaffold (Nano-CD) that utilizes a specific sgRNA, selected from a functional screen for triggering endogenous GDSME expression, while releasing cisplatin to initiate immunologic cell death. Mechanistically, cascade-amplification of the antitumor immune response is prompted by the adjuvantic properties of the lytic intracellular content and enhanced by the heightened GDSME expression, resulting in pyroptosis and the release of tumor associated antigens. Neither of the single components provide efficient tumour control, while tumor growth is efficiently inhibited in primary and recurrent melanomas due to the combinatorial effect of cisplatin and self-supplied GSDME. Moreover, Nano-CD in combination with checkpoint blockade creates durable immune memory and strong systemic anti-tumor immune response, leading to disease relapse prevention, lung metastasis inhibition and increased survival in mouse melanomas. Taken together, our therapeutic approach utilizes CRISPR-technology to enable cell-intrinsic protein expression for immunotherapy, using GDSME as prototypic immune modulator. This nanoplatform thus can be applied to modulate further immunological processes for therapeutic benefit.
Collapse
Affiliation(s)
- Ning Wang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Chao Liu
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Yingjie Li
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Dongxue Huang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Xinyue Wu
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Xiaorong Kou
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Xiye Wang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Qinjie Wu
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, 610041, China.
| | - Changyang Gong
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, 610041, China.
| |
Collapse
|
32
|
Influence of DNA Type on the Physicochemical and Biological Properties of Polyplexes Based on Star Polymers Bearing Different Amino Functionalities. Polymers (Basel) 2023; 15:polym15040894. [PMID: 36850178 PMCID: PMC9966362 DOI: 10.3390/polym15040894] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Revised: 01/20/2023] [Accepted: 02/07/2023] [Indexed: 02/16/2023] Open
Abstract
The interactions of two star polymers based on poly (2-(dimethylamino)ethyl methacrylate) with different types of nucleic acids are investigated. The star polymers differ only in their functionality to bear protonable amino or permanently charged quaternary ammonium groups, while DNAs of different molar masses, lengths and topologies are used. The main physicochemical parameters of the resulting polyplexes are determined. The influence of the polymer' functionality and length and topology of the DNA on the structure and properties of the polyelectrolyte complexes is established. The quaternized polymer is characterized by a high binding affinity to DNA and formed strongly positively charged, compact and tight polyplexes. The parent, non-quaternized polymer exhibits an enhanced buffering capacity and weakened polymer/DNA interactions, particularly upon the addition of NaCl, resulting in the formation of less compact and tight polyplexes. The cytotoxic evaluation of the systems indicates that they are sparing with respect to the cell lines studied including osteosarcoma, osteoblast and human adipose-derived mesenchymal stem cells and exhibit good biocompatibility. Transfection experiments reveal that the non-quaternized polymer is effective at transferring DNA into cells, which is attributed to its high buffering capacity, facilitating the endo-lysosomal escape of the polyplex, the loose structure of the latter one and weakened polymer/DNA interactions, benefitting the DNA release.
Collapse
|
33
|
Li F, Sun X, Yang J, Ren J, Huang M, Wang S, Yang D. A Thermal and Enzymatic Dual-Stimuli Responsive DNA-Based Nanomachine for Controlled mRNA Delivery. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2204905. [PMID: 36461751 PMCID: PMC9896069 DOI: 10.1002/advs.202204905] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Revised: 11/03/2022] [Indexed: 06/17/2023]
Abstract
The extreme instability of mRNA makes the practical application of mRNA-based vaccines heavily rely on efficient delivery system and cold chain transportation. Herein, a DNA-based nanomachine, which achieves programmed capture, long-term storage without cryopreservation, and efficient delivery of mRNA in cells, is developed. The polythymidine acid (Poly-T) functionalized poly(N-isopropylacrylamide) (DNA-PNIPAM) is synthesized and assembled as the central compartment of the nanomachine. The DNA-PNIPAM nano-assembly exhibits reversible thermal-responsive dynamic property: when lower than the low critical solution temperature (LCST, ≈32 °C) of PNIPAM, the DNA-PNIPAM transforms into extension state to expose the poly-T, facilitating the hybridization with polyadenylic acid (Poly-A) tail of mRNA; when higher than LCST, DNA-PNIPAM re-assembles and achieves an efficient encapsulation of mRNA. It is remarkable that the DNA-PNIPAM nano-assembly realizes long-term storage of mRNA (≈7 days) at 37 °C. Biodegradable 2-hydroxypropyltrimethyl ammonium chloride chitosan is assembled on the outside of DNA-PNIPAM to facilitate the endocytosis of mRNA, RNase-H mediating mRNA release occurs in cytoplasm, and efficient mRNA translation is achieved. This work provides a new disign principle of nanosystem for mRNA delivery.
Collapse
Affiliation(s)
- Feng Li
- Frontiers Science Center for Synthetic BiologyKey Laboratory of Systems Bioengineering (MOE)Institute of Biomolecular and Biomedical EngineeringSchool of Chemical Engineering and TechnologyTianjin UniversityTianjin300350P. R. China
| | - Xiaolei Sun
- Frontiers Science Center for Synthetic BiologyKey Laboratory of Systems Bioengineering (MOE)Institute of Biomolecular and Biomedical EngineeringSchool of Chemical Engineering and TechnologyTianjin UniversityTianjin300350P. R. China
| | - Jing Yang
- Beijing Institute of Microbiology and EpidemiologyBeijing100850P. R. China
| | - Jin Ren
- Beijing Institute of Microbiology and EpidemiologyBeijing100850P. R. China
| | - Mengxue Huang
- Frontiers Science Center for Synthetic BiologyKey Laboratory of Systems Bioengineering (MOE)Institute of Biomolecular and Biomedical EngineeringSchool of Chemical Engineering and TechnologyTianjin UniversityTianjin300350P. R. China
| | - Shengqi Wang
- Beijing Institute of Microbiology and EpidemiologyBeijing100850P. R. China
| | - Dayong Yang
- Frontiers Science Center for Synthetic BiologyKey Laboratory of Systems Bioengineering (MOE)Institute of Biomolecular and Biomedical EngineeringSchool of Chemical Engineering and TechnologyTianjin UniversityTianjin300350P. R. China
| |
Collapse
|
34
|
Lee D, Kwak G, Johnson TV, Suk JS. Formulation and Evaluation of Polymer-Based Nanoparticles for Intravitreal Gene-Delivery Applications. Curr Protoc 2022; 2:e607. [PMID: 36469609 PMCID: PMC9731353 DOI: 10.1002/cpz1.607] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The advent of the first-ever retinal gene therapy product, involving subretinal administration of a virus-based gene delivery platform, has garnered hope that this state-of-the-art therapeutic modality may benefit a broad spectrum of patients with diverse retinal disorders. On the other hand, clinical studies have revealed limitations of the applied delivery strategy that may restrict its universal use. To this end, intravitreal administration of synthetic gene-delivery platforms, such as polymer-based nanoparticles (PNPs), has emerged as an attractive alternative to the current mainstay. To achieve success, however, it is imperative that synthetic platforms overcome key biological barriers in human eyes encountered following intravitreal administration, including the vitreous gel and inner limiting membrane (ILM). Here, we introduce a series of experiments, from the fabrication of PNPs to a comprehensive evaluation in relevant experimental models, to determine whether PNPs overcome these barriers and efficiently deliver therapeutic gene payloads to retinal cells. We conclude the article by discussing a few important considerations for successful implementation of the strategy. © 2022 Wiley Periodicals LLC. Basic Protocol 1: Preparation and characterization of PNPs Basic Protocol 2: Evaluation of in vitro transfection efficacy Basic Protocol 3: Evaluation of PNP diffusion in vitreous gel Basic Protocol 4: Ex vivo assessment of PNP penetration within vitreoretinal explant culture Basic Protocol 5: Assessment of in vivo transgene expression mediated by intravitreally administered PNPs.
Collapse
Affiliation(s)
- Daiheon Lee
- Department of Ophthalmology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Center for Nanomedicine, Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- These authors contributed equally to this work
| | - Gijung Kwak
- Department of Ophthalmology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Center for Nanomedicine, Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- These authors contributed equally to this work
| | - Thomas V. Johnson
- Department of Ophthalmology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Jung Soo Suk
- Department of Ophthalmology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Center for Nanomedicine, Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Chemical & Biomolecular Engineering, Johns Hopkins University, Baltimore, MD, USA
| |
Collapse
|
35
|
Alavijeh HN, Baltus RE. Can Hindered Transport Models for Rigid Spheres Predict the Rejection of Single Stranded DNA from Porous Membranes? MEMBRANES 2022; 12:1099. [PMID: 36363653 PMCID: PMC9694696 DOI: 10.3390/membranes12111099] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Revised: 10/29/2022] [Accepted: 11/01/2022] [Indexed: 06/16/2023]
Abstract
In this paper, predictions from a theoretical model describing the rejection of a rigid spherical solute from porous membranes are compared to experimental results for a single stranded DNA (ssDNA) with 60 thymine nucleotides. Experiments were conducted with different pore size track-etched membranes at different transmembrane pressures and different NaCl concentrations. The model includes both hydrodynamic and electrostatic solute-pore wall interactions; predictions were made using different size parameters for the ssDNA (radius of gyration, hydrodynamic radius, and root mean square end-to-end distance). At low transmembrane pressures, experimental results are in good agreement with rejection predictions made using the hard sphere model for the ssDNA when the solute size is described using its root mean square end-to-end distance. When the ssDNA size is characterized using the radius of gyration or the hydrodynamic radius, the hard sphere model under-predicts rejection. Not surprisingly, the model overestimates ssDNA rejection at conditions where flow induced elongation of the DNA is expected. The results from this study are encouraging because they mean that a relatively simple hindered transport model can be used to estimate the rejection of a small DNA from porous membranes.
Collapse
Affiliation(s)
- Hossein Nouri Alavijeh
- Department of Chemical Engineering, University of Virginia, Charlottesville, VA 22904-4259, USA
| | - Ruth E. Baltus
- Department of Chemical and Biomolecular Engineering, Clarkson University, Potsdam, NY 13699-5705, USA
| |
Collapse
|
36
|
Corridon PR, Wang X, Shakeel A, Chan V. Digital Technologies: Advancing Individualized Treatments through Gene and Cell Therapies, Pharmacogenetics, and Disease Detection and Diagnostics. Biomedicines 2022; 10:biomedicines10102445. [PMID: 36289707 PMCID: PMC9599083 DOI: 10.3390/biomedicines10102445] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Accepted: 09/25/2022] [Indexed: 11/28/2022] Open
Abstract
Digital technologies are shifting the paradigm of medicine in a way that will transform the healthcare industry. Conventional medical approaches focus on treating symptoms and ailments for large groups of people. These approaches can elicit differences in treatment responses and adverse reactions based on population variations, and are often incapable of treating the inherent pathophysiology of the medical conditions. Advances in genetics and engineering are improving healthcare via individualized treatments that include gene and cell therapies, pharmacogenetics, disease detection, and diagnostics. This paper highlights ways that artificial intelligence can help usher in an age of personalized medicine.
Collapse
Affiliation(s)
- Peter R. Corridon
- Department of Immunology and Physiology, College of Medicine and Health Sciences, Khalifa University, Abu Dhabi P.O. Box 127788, United Arab Emirates
- Biomedical Engineering and Healthcare Engineering Innovation Center, Khalifa University, Abu Dhabi P.O. Box 127788, United Arab Emirates
- Center for Biotechnology, Khalifa University, Abu Dhabi P.O. Box 127788, United Arab Emirates
- Correspondence:
| | - Xinyu Wang
- Department of Immunology and Physiology, College of Medicine and Health Sciences, Khalifa University, Abu Dhabi P.O. Box 127788, United Arab Emirates
- Biomedical Engineering and Healthcare Engineering Innovation Center, Khalifa University, Abu Dhabi P.O. Box 127788, United Arab Emirates
| | - Adeeba Shakeel
- Department of Immunology and Physiology, College of Medicine and Health Sciences, Khalifa University, Abu Dhabi P.O. Box 127788, United Arab Emirates
| | - Vincent Chan
- Biomedical Engineering and Healthcare Engineering Innovation Center, Khalifa University, Abu Dhabi P.O. Box 127788, United Arab Emirates
| |
Collapse
|
37
|
Wu W, Ngo A, Ban W, Zhong Y, Cheng D, Gu Z, Yu C, Song H. Tailoring head-tail mesoporous silica nanoparticles for enhanced gene transfection. J Mater Chem B 2022; 10:7995-8002. [PMID: 36128923 DOI: 10.1039/d2tb01737g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Plasmid DNA (pDNA) delivery has attracted extensive research interest due to its great potential in gene therapy. The design of efficient nano-vectors to promote cellular delivery and transfection of gene molecules is the key to success. Compared to conventional nanocarriers with spherical geometry, asymmetric nanoparticles have been well documented showing enhanced cellular uptake and drug delivery capability. However, the impact of asymmetric nanostructures on pDNA binding and following intracellular delivery performance has been less reported. Herein, asymmetric head-tail mesoporous silica nanoparticles (HTMSNs) with tailored tail lengths were synthesized and employed as nano-vectors for pDNA delivery. The nanostructures of HTMSNs were carefully characterized by electron tomography. The pDNA binding, cellular uptake and gene transfection capabilities of engineered asymmetric nanoparticles were compared with symmetric dendritic mesoporous silica nanoparticles (DMSNs). The results showed that the asymmetric morphology of nanoparticles promoted pDNA binding and cell internalization, where HTMSNs-66 with a specific tail length of 66 nm achieved the highest transfection efficiency. This study reveals the impact of asymmetric nanostructure on DNA interaction, and provides guidance in future designs of non-viral nano-vectors for efficient gene delivery.
Collapse
Affiliation(s)
- Weixi Wu
- Australian Institute for Bioengineering and Nanotechnology, the University of Queensland, Brisbane, Queensland 4072, Australia.
| | - Anh Ngo
- Australian Institute for Bioengineering and Nanotechnology, the University of Queensland, Brisbane, Queensland 4072, Australia.
| | - Wenhuang Ban
- Australian Institute for Bioengineering and Nanotechnology, the University of Queensland, Brisbane, Queensland 4072, Australia.
| | - Yuening Zhong
- Australian Institute for Bioengineering and Nanotechnology, the University of Queensland, Brisbane, Queensland 4072, Australia.
| | - Dan Cheng
- Australian Institute for Bioengineering and Nanotechnology, the University of Queensland, Brisbane, Queensland 4072, Australia.
| | - Zhengying Gu
- School of Chemistry and Molecular Engineering, East China Normal University, Shanghai, 200241, P. R. China
| | - Chengzhong Yu
- Australian Institute for Bioengineering and Nanotechnology, the University of Queensland, Brisbane, Queensland 4072, Australia. .,School of Chemistry and Molecular Engineering, East China Normal University, Shanghai, 200241, P. R. China
| | - Hao Song
- Australian Institute for Bioengineering and Nanotechnology, the University of Queensland, Brisbane, Queensland 4072, Australia.
| |
Collapse
|
38
|
Liu L, Yang Z, Liu C, Wang M, Chen X. Preparation of PEI-modified nanoparticles by dopamine self-polymerization for efficient DNA delivery. Biotechnol Appl Biochem 2022; 70:824-834. [PMID: 36070708 DOI: 10.1002/bab.2402] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Accepted: 08/27/2022] [Indexed: 11/09/2022]
Abstract
Achieving efficient and safe gene delivery is great of significance to promote the development of gene therapy. In this work, a polydopamine (PDA) layer was coated on the surface of Fe3 O4 nanoparticles (NPs) by dopamine (DA) self-polymerization, and then magnetic Fe3 O4 NPs were prepared by the Michael addition between amino groups in polyethyleneimine (PEI) and PDA. The prepared Fe3 O4 NPs (named Fe3 O4 @PDA@PEI) were characterized by FTIR, atomic force microscopy (AFM) and scanning electron microscope (SEM). As an efficient and safe gene carrier, the potential of Fe3 O4 @PDA@PEI was evaluated by agarose gel electrophoresis, MTT assay, fluorescence microscopy, flow cytometry. The results shows that the Fe3 O4 @PDA@PEI NPs is stable hydrophilic nanoparticles with a particle size of 50-150 nm. It can efficiently condense DNA at low N/P ratios and protect it from nuclease degradation. In addition, the Fe3 O4 @PDA@PEI NPs has higher safety than PEI. Further, the Fe3 O4 @PDA@PEI/DNA polyplexes could be effectively absorbed by cells and successfully transfected, and exhibit higher cellular uptake and gene transfection efficiency than PEI/DNA polyplexes. The findings indicate that the Fe3 O4 @PDA@PEI NPs has the potential to be developed into a novel gene vector. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Liang Liu
- School of Life and Biology, Wuhan Polytechnic University, Wuhan, 430023, China
| | - Zhaojun Yang
- School of Life and Biology, Wuhan Polytechnic University, Wuhan, 430023, China
| | - Chaobing Liu
- School of Life and Biology, Wuhan Polytechnic University, Wuhan, 430023, China
| | - Mengying Wang
- School of Life and Biology, Wuhan Polytechnic University, Wuhan, 430023, China
| | - Xin Chen
- School of Life and Biology, Wuhan Polytechnic University, Wuhan, 430023, China
| |
Collapse
|
39
|
Zhang Q, Cai J, Wang Z, Wang Z, Lin B, Zhao J, Mao J, Li Y, Li J, Yang X, Shuai X, Lu L, Shen J. Upregulating microRNA‐210 to Inhibit Apoptosis of Neural Stem Cells with an MRI–Visible Nanomedicine for Stroke Therapy. SMALL STRUCTURES 2022; 3. [DOI: 10.1002/sstr.202200035] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/22/2025]
Abstract
Transplantation of neural stem cells (NSCs) is a promising paradigm for treating stroke. However, the poor survival of transplanted NSCs greatly limits the therapeutic potential. microRNA‐210 (miR‐210), a key hypoxia‐regulated miRNA, can enhance cell survival by targeting the expression of multiple apoptosis‐related genes, such as caspase‐8‐associated protein‐2 (casp8ap2), Bax, and Bcl‐2. Meanwhile, a noninvasive cell‐tracking method is also indispensable for monitoring the in vivo cell‐based therapy. Herein, an MRI–visible nanomedicine is developed to codeliver superparamagnetic iron oxide (SPIO) nanoparticles and miR‐210 into NSCs. This therapeutic nanomedicine not only promotes the survival of NSCs via upregulating miR‐210 to inhibit NSCs apoptosis but also allows an in vivo tracking of transplanted NSCs with MRI. The enhanced NSCs survivability significantly promotes the structural and functional recovery after stroke onset, which highlights the great potential of the multifunctional nanomedicine to improve the therapeutic efficacy of NSCs for stroke treatment.
Collapse
Affiliation(s)
- Qinyuan Zhang
- Department of Radiology Sun Yat-Sen Memorial Hospital Sun Yat-Sen University Guangzhou 510120 China
| | - Jiali Cai
- PCFM Lab of Ministry of Education Guangzhou Key Laboratory of Flexible Electronic Materials and Wearable Devices School of Materials Science and Engineering Sun Yat-Sen University Guangzhou 510275 China
| | - Zhiyong Wang
- PCFM Lab of Ministry of Education Guangzhou Key Laboratory of Flexible Electronic Materials and Wearable Devices School of Materials Science and Engineering Sun Yat-Sen University Guangzhou 510275 China
| | - Zhe Wang
- Department of Radiology Sun Yat-Sen Memorial Hospital Sun Yat-Sen University Guangzhou 510120 China
| | - Bingling Lin
- Department of Radiology Peking University Shenzhen Hospital Shenzhen 518000 China
| | - Junya Zhao
- Department of Radiology Sun Yat-Sen Memorial Hospital Sun Yat-Sen University Guangzhou 510120 China
| | - Jiaji Mao
- Department of Radiology Sun Yat-Sen Memorial Hospital Sun Yat-Sen University Guangzhou 510120 China
| | - Yunhua Li
- Department of Radiology Sun Yat-Sen Memorial Hospital Sun Yat-Sen University Guangzhou 510120 China
| | - Jianing Li
- Department of Radiology Sun Yat-Sen Memorial Hospital Sun Yat-Sen University Guangzhou 510120 China
| | - Xieqing Yang
- Department of Radiology Sun Yat-Sen Memorial Hospital Sun Yat-Sen University Guangzhou 510120 China
| | - Xintao Shuai
- PCFM Lab of Ministry of Education School of Materials Science and Engineering Sun Yat-Sen University Guangzhou 510275 China
| | - Liejing Lu
- Department of Radiology Sun Yat-Sen Memorial Hospital Sun Yat-Sen University Guangzhou 510120 China
| | - Jun Shen
- Department of Radiology Sun Yat-Sen Memorial Hospital Sun Yat-Sen University Guangzhou 510120 China
| |
Collapse
|
40
|
Hao M, Zhang L, Chen P. Membrane Internalization Mechanisms and Design Strategies of Arginine-Rich Cell-Penetrating Peptides. Int J Mol Sci 2022; 23:ijms23169038. [PMID: 36012300 PMCID: PMC9409441 DOI: 10.3390/ijms23169038] [Citation(s) in RCA: 48] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2022] [Revised: 08/05/2022] [Accepted: 08/05/2022] [Indexed: 11/29/2022] Open
Abstract
Cell-penetrating peptides (CPPs) have been discovered to deliver chemical drugs, nucleic acids, and macromolecules to permeate cell membranes, creating a novel route for exogenous substances to enter cells. Up until now, various sequence structures and fundamental action mechanisms of CPPs have been established. Among them, arginine-rich peptides with unique cell penetration properties have attracted substantial scientific attention. Due to the positively charged essential amino acids of the arginine-rich peptides, they can interact with negatively charged drug molecules and cell membranes through non-covalent interaction, including electrostatic interactions. Significantly, the sequence design and the penetrating mechanisms are critical. In this brief synopsis, we summarize the transmembrane processes and mechanisms of arginine-rich peptides; and outline the relationship between the function of arginine-rich peptides and the number of arginine residues, arginine optical isomers, primary sequence, secondary and ternary structures, etc. Taking advantage of the penetration ability, biomedical applications of arginine-rich peptides have been refreshed, including drug/RNA delivery systems, biosensors, and blood-brain barrier (BBB) penetration. Understanding the membrane internalization mechanisms and design strategies of CPPs will expand their potential applications in clinical trials.
Collapse
Affiliation(s)
- Minglu Hao
- Advanced Materials Institute, Qilu University of Technology (Shandong Academy of Sciences), Jinan 250014, China
| | - Lei Zhang
- Department of Chemical Engineering, Waterloo Institute for Nanotechnology, University of Waterloo, 200 University Avenue West, Waterloo, ON N2L3G1, Canada
- Correspondence: (L.Z.); (P.C.)
| | - Pu Chen
- Advanced Materials Institute, Qilu University of Technology (Shandong Academy of Sciences), Jinan 250014, China
- Department of Chemical Engineering, Waterloo Institute for Nanotechnology, University of Waterloo, 200 University Avenue West, Waterloo, ON N2L3G1, Canada
- Correspondence: (L.Z.); (P.C.)
| |
Collapse
|
41
|
Upconversion Nanostructures Applied in Theranostic Systems. Int J Mol Sci 2022; 23:ijms23169003. [PMID: 36012269 PMCID: PMC9409402 DOI: 10.3390/ijms23169003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Revised: 08/05/2022] [Accepted: 08/10/2022] [Indexed: 11/30/2022] Open
Abstract
Upconversion (UC) nanostructures, which can upconvert near-infrared (NIR) light with low energy to visible or UV light with higher energy, are investigated for theranostic applications. The surface of lanthanide (Ln)-doped UC nanostructures can be modified with different functional groups and bioconjugated with biomolecules for therapeutic systems. On the other hand, organic molecular-based UC nanostructures, by using the triplet-triplet annihilation (TTA) UC mechanism, have high UC quantum yields and do not require high excitation power. In this review, the major UC mechanisms in different nanostructures have been introduced, including the Ln-doped UC mechanism and the TTA UC mechanism. The design and fabrication of Ln-doped UC nanostructures and TTA UC-based UC nanostructures for theranostic applications have been reviewed and discussed. In addition, the current progress in the application of UC nanostructures for diagnosis and therapy has been summarized, including tumor-targeted bioimaging and chemotherapy, image-guided diagnosis and phototherapy, NIR-triggered controlled drug releasing and bioimaging. We also provide insight into the development of emerging UC nanostructures in the field of theranostics.
Collapse
|
42
|
Cheng D, Theivendran S, Tang J, Cai L, Zhang J, Song H, Yu C. Surface chemistry of spiky silica nanoparticles tailors polyethyleneimine binding and intracellular DNA delivery. J Colloid Interface Sci 2022; 628:297-305. [PMID: 35998455 DOI: 10.1016/j.jcis.2022.08.038] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2022] [Revised: 07/27/2022] [Accepted: 08/06/2022] [Indexed: 11/17/2022]
Abstract
Cellular delivery of DNA using silica nanoparticles has attracted great attention. Typically, polyethyleneimine (PEI) is used to form a silica/PEI composite vector. Understanding the interactions at the silica and PEI interface is important for successful DNA delivery and transfection, especially for silica with different surface functionality. Herein, we report that a higher content of hydrogen boning formed between PEI molecules and phosphonate modified silica nanoparticles could slow down the PEI dissolution from the freeze-dried solid composites into aqueous solution than the bare silica counterpart. The pronounced PEI retention ability through phosphonation of silica nanoparticles effectively improves the transfection efficiency due to the high DNA binding affinity extracellularly, effective lysosome escape and high nuclear entry of both PEI and DNA intracellularly. Our study provides a fundamental understanding on designing effective silica-PEI-based nano-vectors for DNA delivery applications.
Collapse
Affiliation(s)
- Dan Cheng
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Shevanuja Theivendran
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Jie Tang
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Larry Cai
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Jun Zhang
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Hao Song
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, QLD 4072, Australia.
| | - Chengzhong Yu
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, QLD 4072, Australia.
| |
Collapse
|
43
|
Soltani Dehnavi S, Eivazi Zadeh Z, Harvey AR, Voelcker NH, Parish CL, Williams RJ, Elnathan R, Nisbet DR. Changing Fate: Reprogramming Cells via Engineered Nanoscale Delivery Materials. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2022; 34:e2108757. [PMID: 35396884 DOI: 10.1002/adma.202108757] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/31/2021] [Revised: 04/02/2022] [Indexed: 06/14/2023]
Abstract
The incorporation of nanotechnology in regenerative medicine is at the nexus of fundamental innovations and early-stage breakthroughs, enabling exciting biomedical advances. One of the most exciting recent developments is the use of nanoscale constructs to influence the fate of cells, which are the basic building blocks of healthy function. Appropriate cell types can be effectively manipulated by direct cell reprogramming; a robust technique to manipulate cellular function and fate, underpinning burgeoning advances in drug delivery systems, regenerative medicine, and disease remodeling. Individual transcription factors, or combinations thereof, can be introduced into cells using both viral and nonviral delivery systems. Existing approaches have inherent limitations. Viral-based tools include issues of viral integration into the genome of the cells, the propensity for uncontrollable silencing, reduced copy potential and cell specificity, and neutralization via the immune response. Current nonviral cell reprogramming tools generally suffer from inferior expression efficiency. Nanomaterials are increasingly being explored to address these challenges and improve the efficacy of both viral and nonviral delivery because of their unique properties such as small size and high surface area. This review presents the state-of-the-art research in cell reprogramming, focused on recent breakthroughs in the deployment of nanomaterials as cell reprogramming delivery tools.
Collapse
Affiliation(s)
- Shiva Soltani Dehnavi
- ACRF Department of Cancer Biology and Therapeutics, The John Curtin School of Medical Research, ANU College of Health & Medicine, Canberra, ACT, 2601, Australia
- Research School of Chemistry, ANU College of Science, Canberra, ACT, 2601, Australia
- ANU College of Engineering & Computer Science, Canberra, ACT, 2601, Australia
| | - Zahra Eivazi Zadeh
- Biomedical Engineering Department, Amirkabir University of Technology, Tehran, 15875-4413, Iran
- The Graeme Clark Institute, The University of Melbourne, Melbourne, VIC, 3010, Australia
- Department of Biomedical Engineering, Faculty of Engineering and Information Technology, The University of Melbourne, Melbourne, VIC, 3010, Australia
| | - Alan R Harvey
- School of Human Sciences, The University of Western Australia, and Perron Institute for Neurological and Translational Science, Perth, WA, 6009, Australia
| | - Nicolas H Voelcker
- Faculty of Pharmacy and Pharmaceutical Sciences, Monash University, Parkville, VIC, 3052, Australia
- Melbourne Centre for Nanofabrication, Victorian Node of the Australian National Fabrication Facility, 151 Wellington Road, Clayton, VIC, 3168, Australia
- CSIRO Manufacturing, Bayview Avenue, Clayton, VIC, 3168, Australia
| | - Clare L Parish
- The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, Melbourne, VIC, 3010, Australia
| | - Richard J Williams
- iMPACT, School of Medicine, Deakin University, Waurn Ponds, VIC, 3216, Australia
| | - Roey Elnathan
- Faculty of Pharmacy and Pharmaceutical Sciences, Monash University, Parkville, VIC, 3052, Australia
- Melbourne Centre for Nanofabrication, Victorian Node of the Australian National Fabrication Facility, 151 Wellington Road, Clayton, VIC, 3168, Australia
- CSIRO Manufacturing, Bayview Avenue, Clayton, VIC, 3168, Australia
- iMPACT, School of Medicine, Deakin University, Waurn Ponds, VIC, 3216, Australia
| | - David R Nisbet
- ACRF Department of Cancer Biology and Therapeutics, The John Curtin School of Medical Research, ANU College of Health & Medicine, Canberra, ACT, 2601, Australia
- Research School of Chemistry, ANU College of Science, Canberra, ACT, 2601, Australia
- The Graeme Clark Institute, The University of Melbourne, Melbourne, VIC, 3010, Australia
- Department of Biomedical Engineering, Faculty of Engineering and Information Technology, The University of Melbourne, Melbourne, VIC, 3010, Australia
- Melbourne Medical School, Faculty of Medicine, Dentistry and Health Science, The University of Melbourne, Melbourne, VIC, 3010, Australia
| |
Collapse
|
44
|
Yi S, Kim SY, Vincent MP, Yuk SA, Bobbala S, Du F, Scott EA. Dendritic peptide-conjugated polymeric nanovectors for non-toxic delivery of plasmid DNA and enhanced non-viral transfection of immune cells. iScience 2022; 25:104555. [PMID: 35769884 PMCID: PMC9234717 DOI: 10.1016/j.isci.2022.104555] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Revised: 04/29/2022] [Accepted: 06/02/2022] [Indexed: 10/26/2022] Open
Abstract
Plasmid DNA (pDNA) transfection is advantageous for gene therapies requiring larger genetic elements, including "all-in-one" CRISPR/Cas9 plasmids, but is limited by toxicity as well as poor intracellular release and transfection efficiency in immune cell populations. Here, we developed a synthetic non-viral gene delivery platform composed of poly(ethylene glycol)-b-poly(propylene sulfide) copolymers linked to a cationic dendritic peptide (DP) via a reduceable bond, PEG-b-PPS-ss-DP (PPDP). A library of self-assembling PPDP polymers was synthesized and screened to identify optimal constructs capable of transfecting macrophages with small (pCMV-DsRed, 4.6 kb) and large (pL-CRISPR.EFS.tRFP, 11.7 kb) plasmids. The optimized PPDP construct transfected macrophages, fibroblasts, dendritic cells, and T cells more efficiently and with less toxicity than a commercial Lipo2K reagent, regardless of pDNA size and under standard culture conditions in the presence of serum. The PPDP technology described herein is a stimuli-responsive polymeric nanovector that can be leveraged to meet diverse challenges in gene delivery.
Collapse
Affiliation(s)
- Sijia Yi
- Department of Biomedical Engineering, Northwestern University, Evanston, IL 60208, USA
| | - Sun-Young Kim
- Department of Biomedical Engineering, Northwestern University, Evanston, IL 60208, USA
- SKKU Advanced Institute of Nanotechnology (SAINT), Sungkyunkwan University (SKKU), 2066 Seobu-ro, Jangan-gu, Suwon-si, Gyeonggi-do 16419, Republic of Korea
| | - Michael P. Vincent
- Department of Biomedical Engineering, Northwestern University, Evanston, IL 60208, USA
| | - Simseok A. Yuk
- Department of Biomedical Engineering, Northwestern University, Evanston, IL 60208, USA
| | - Sharan Bobbala
- Department of Pharmaceutical Sciences, School of Pharmacy, West Virginia University, Morgantown, WV 26505, USA
| | - Fanfan Du
- Department of Biomedical Engineering, Northwestern University, Evanston, IL 60208, USA
| | - Evan Alexander Scott
- Department of Biomedical Engineering, Northwestern University, Evanston, IL 60208, USA
- Simpson Querrey Institute, Northwestern University, Chicago, IL 60611, USA
- Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago, IL 60611, USA
- Department of Microbiology-Immunology, Northwestern University, Chicago, IL 60611, USA
| |
Collapse
|
45
|
Feng C, Zhang YJ, Ren CL. pH-Regulated Single and Double Charge Inversions on PEI-Coated Surfaces. ACS Macro Lett 2022; 11:773-779. [PMID: 35653775 DOI: 10.1021/acsmacrolett.2c00227] [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
The pH-regulated charge inversions on polyethylenimine (PEI)-coated surfaces are indispensable to their applications in biomaterials and nanomaterials. Various PEI-coated surfaces, where single charge inversion happens, have been extensively investigated, while the surfaces where double charge inversion appears are less reported. Here, using a molecular theory, we systematically study the pH-regulated charge density of PEI-coated surfaces. The results suggest whether single or double charge inversion happens depends on PEI affinity to the surface and the bare surface charge density. The region of double charge inversion is much smaller than that of single charge inversion, revealing the reason why double charge inversion is less observed in experiments. Besides, the charge inversions are significantly influenced by the solution condition. The present work provides a useful guideline to the selection of the coated materials and the parameters of PEI solution in the design of PEI-coated surfaces aiming to promote their applications in multifunctional nanomaterials.
Collapse
Affiliation(s)
- Chao Feng
- State Key Laboratory of Metastable Materials Science and Technology and Hebei Key Laboratory of Microstructural Material Physics, School of Science, Yanshan University, Qinhuangdao, 066004, China
| | - Yun-jian Zhang
- State Key Laboratory of Metastable Materials Science and Technology and Hebei Key Laboratory of Microstructural Material Physics, School of Science, Yanshan University, Qinhuangdao, 066004, China
| | - Chun-lai Ren
- National Laboratory of Solid State Microstructures and Department of Physics, Nanjing University, Nanjing, 210093, China
- Collaborative Innovation Center of Advanced Microstructures, Nanjing University, Nanjing, 210093, China
| |
Collapse
|
46
|
Białkowska K, Komorowski P, Gomez-Ramirez R, de la Mata FJ, Bryszewska M, Miłowska K. Interaction of Cationic Carbosilane Dendrimers and Their siRNA Complexes with MCF-7 Cells Cultured in 3D Spheroids. Cells 2022; 11:cells11101697. [PMID: 35626734 PMCID: PMC9140188 DOI: 10.3390/cells11101697] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Revised: 05/18/2022] [Accepted: 05/19/2022] [Indexed: 02/01/2023] Open
Abstract
Cationic dendrimers are effective carriers for the delivery of siRNA into cells; they can penetrate cell membranes and protect nucleic acids against RNase degradation. Two types of dendrimers (CBD-1 and CBD-2) and their complexes with pro-apoptotic siRNA (Mcl-1 and Bcl-2) were tested on MCF-7 cells cultured as spheroids. Cytotoxicity of dendrimers and dendriplexes was measured using the live–dead test and Annexin V-FITC Apoptosis Detection Kit (flow cytometry). Uptake of dendriplexes was examined using flow cytometry and confocal microscopy. The live–dead test showed that for cells in 3D, CBD-2 is more toxic than CBD-1, contrasting with the data for 2D cultures. Attaching siRNA to a dendrimer molecule did not lead to increased cytotoxic effect in cells, either after 24 or 48 h. Measurements of apoptosis did not show a high increase in the level of the apoptosis marker after 24 h exposure of spheroids to CBD-2 and its dendriplexes. Measurements of the internalization of dendriplexes and microscopy images confirmed that the dendriplexes were transported into cells of the spheroids. Flow cytometry analysis of internalization indicated that CBD-2 transported siRNAs more effectively than CBD-1. Cytotoxic effects were visible after incubation with 3 doses of complexes for CBD-1 and both siRNAs.
Collapse
Affiliation(s)
- Kamila Białkowska
- Department of General Biophysics, Faculty of Biology and Environmental Protection, University of Lodz, 141/143 Pomorska St., 90-236 Lodz, Poland; (M.B.); (K.M.)
- Molecular and Nanostructural Biophysics Laboratory, “Bionanopark” Ldt., 114/116 Dubois St., 93-465 Lodz, Poland;
- Correspondence:
| | - Piotr Komorowski
- Molecular and Nanostructural Biophysics Laboratory, “Bionanopark” Ldt., 114/116 Dubois St., 93-465 Lodz, Poland;
- Department of Biophysics, Institute of Materials Science, Lodz University of Technology, 1/15 Stefanowskiego St., 90-924 Lodz, Poland
| | - Rafael Gomez-Ramirez
- Department of Organic and Inorganic Chemistry, IQAR, University of Alcalá, 28805 Madrid, Spain; (R.G.-R.); (F.J.d.l.M.)
- Networking Research Center on Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), 28029 Madrid, Spain
- Ramón y Cajal Health Research Institute (IRYCIS), 28034 Madrid, Spain
| | - Francisco Javier de la Mata
- Department of Organic and Inorganic Chemistry, IQAR, University of Alcalá, 28805 Madrid, Spain; (R.G.-R.); (F.J.d.l.M.)
- Networking Research Center on Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), 28029 Madrid, Spain
- Ramón y Cajal Health Research Institute (IRYCIS), 28034 Madrid, Spain
| | - Maria Bryszewska
- Department of General Biophysics, Faculty of Biology and Environmental Protection, University of Lodz, 141/143 Pomorska St., 90-236 Lodz, Poland; (M.B.); (K.M.)
| | - Katarzyna Miłowska
- Department of General Biophysics, Faculty of Biology and Environmental Protection, University of Lodz, 141/143 Pomorska St., 90-236 Lodz, Poland; (M.B.); (K.M.)
| |
Collapse
|
47
|
Sarode A, Fan Y, Byrnes AE, Hammel M, Hura GL, Fu Y, Kou P, Hu C, Hinz FI, Roberts J, Koenig SG, Nagapudi K, Hoogenraad CC, Chen T, Leung D, Yen CW. Predictive high-throughput screening of PEGylated lipids in oligonucleotide-loaded lipid nanoparticles for neuronal gene silencing. NANOSCALE ADVANCES 2022; 4:2107-2123. [PMID: 36133441 PMCID: PMC9417559 DOI: 10.1039/d1na00712b] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/25/2021] [Accepted: 01/22/2022] [Indexed: 05/25/2023]
Abstract
Lipid nanoparticles (LNPs) are gaining traction in the field of nucleic acid delivery following the success of two mRNA vaccines against COVID-19. As one of the constituent lipids on LNP surfaces, PEGylated lipids (PEG-lipids) play an important role in defining LNP physicochemical properties and biological interactions. Previous studies indicate that LNP performance is modulated by tuning PEG-lipid parameters including PEG size and architecture, carbon tail type and length, as well as the PEG-lipid molar ratio in LNPs. Owing to these numerous degrees of freedom, a high-throughput approach is necessary to fully understand LNP behavioral trends over a broad range of PEG-lipid variables. To this end, we report a low-volume, automated, high-throughput screening (HTS) workflow for the preparation, characterization, and in vitro assessment of LNPs loaded with a therapeutic antisense oligonucleotide (ASO). A library of 54 ASO-LNP formulations with distinct PEG-lipid compositions was prepared using a liquid handling robot and assessed for their physiochemical properties as well as gene silencing efficacy in murine cortical neurons. Our results show that the molar ratio of anionic PEG-lipid in LNPs regulates particle size and PEG-lipid carbon tail length controls ASO-LNP gene silencing activity. ASO-LNPs formulated using PEG-lipids with optimal carbon tail lengths achieved up to 5-fold lower mRNA expression in neurons as compared to naked ASO. Representative ASO-LNP formulations were further characterized using dose-response curves and small-angle X-ray scattering to understand structure-activity relationships. Identified hits were also tested for efficacy in primary murine microglia and were scaled-up using a microfluidic formulation technique, demonstrating a smooth translation of ASO-LNP properties and in vitro efficacy. The reported HTS workflow can be used to screen additional multivariate parameters of LNPs with significant time and material savings, therefore guiding the selection and scale-up of optimal formulations for nucleic acid delivery to a variety of cellular targets.
Collapse
Affiliation(s)
- Apoorva Sarode
- Small Molecule Pharmaceutical Sciences, Genentech Inc. 1 DNA Way South San Francisco CA-94080 USA
| | - Yuchen Fan
- Small Molecule Pharmaceutical Sciences, Genentech Inc. 1 DNA Way South San Francisco CA-94080 USA
| | - Amy E Byrnes
- Department of Neuroscience, Genentech, Inc. South San Francisco CA 94080 USA
| | - Michal Hammel
- Molecular Biophysics and Integrated Bioimaging Division, Lawrence Berkeley National Lab Berkeley CA USA
| | - Greg L Hura
- Molecular Biophysics and Integrated Bioimaging Division, Lawrence Berkeley National Lab Berkeley CA USA
- Chemistry and Biochemistry Department, University of California Santa Cruz Santa Cruz CA USA
| | - Yige Fu
- Small Molecule Pharmaceutical Sciences, Genentech Inc. 1 DNA Way South San Francisco CA-94080 USA
| | - Ponien Kou
- Small Molecule Pharmaceutical Sciences, Genentech Inc. 1 DNA Way South San Francisco CA-94080 USA
| | - Chloe Hu
- Small Molecule Pharmaceutical Sciences, Genentech Inc. 1 DNA Way South San Francisco CA-94080 USA
| | - Flora I Hinz
- Department of Neuroscience, Genentech, Inc. South San Francisco CA 94080 USA
| | - Jasmine Roberts
- Department of Neuroscience, Genentech, Inc. South San Francisco CA 94080 USA
| | - Stefan G Koenig
- Small Molecule Pharmaceutical Sciences, Genentech Inc. 1 DNA Way South San Francisco CA-94080 USA
| | - Karthik Nagapudi
- Small Molecule Pharmaceutical Sciences, Genentech Inc. 1 DNA Way South San Francisco CA-94080 USA
| | - Casper C Hoogenraad
- Department of Neuroscience, Genentech, Inc. South San Francisco CA 94080 USA
| | - Tao Chen
- Small Molecule Pharmaceutical Sciences, Genentech Inc. 1 DNA Way South San Francisco CA-94080 USA
| | - Dennis Leung
- Small Molecule Pharmaceutical Sciences, Genentech Inc. 1 DNA Way South San Francisco CA-94080 USA
| | - Chun-Wan Yen
- Small Molecule Pharmaceutical Sciences, Genentech Inc. 1 DNA Way South San Francisco CA-94080 USA
| |
Collapse
|
48
|
Lewis RW, Klemm B, Macchione M, Eelkema R. Fuel-driven macromolecular coacervation in complex coacervate core micelles. Chem Sci 2022; 13:4533-4544. [PMID: 35656128 PMCID: PMC9019912 DOI: 10.1039/d2sc00805j] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Accepted: 03/30/2022] [Indexed: 12/21/2022] Open
Abstract
Fuel-driven macromolecular coacervation is an entry into the transient formation of highly charged, responsive material phases. In this work, we used a chemical reaction network (CRN) to drive the coacervation of macromolecular species readily produced using radical polymerisation methods. The CRN enables transient quaternization of tertiary amine substrates, driven by the conversion of electron deficient allyl acetates and thiol or amine nucleophiles. By incorporating tertiary amine functionality into block copolymers, we demonstrate chemical triggered complex coacervate core micelle (C3M) assembly and disassembly. In contrast to most dynamic coacervate systems, this CRN operates at constant physiological pH without the need for complex biomolecules. By varying the allyl acetate fuel, deactivating nucleophile and reagent ratios, we achieved both sequential signal-induced C3M (dis)assembly, as well as transient non-equilibrium (dis)assembly. We expect that timed and signal-responsive control over coacervate phase formation at physiological pH will find application in nucleic acid delivery, nano reactors and protocell research. We apply an allyl acetate fuelled chemical reaction network (CRN) to control the coacervation of macromolecular species at constant physiological pH without the need for complex biomolecules.![]()
Collapse
Affiliation(s)
- Reece W Lewis
- Department of Chemical Engineering, Delft University of Technology Van der Maasweg 9 2629 HZ Delft The Netherlands
| | - Benjamin Klemm
- Department of Chemical Engineering, Delft University of Technology Van der Maasweg 9 2629 HZ Delft The Netherlands
| | - Mariano Macchione
- Department of Chemical Engineering, Delft University of Technology Van der Maasweg 9 2629 HZ Delft The Netherlands
| | - Rienk Eelkema
- Department of Chemical Engineering, Delft University of Technology Van der Maasweg 9 2629 HZ Delft The Netherlands
| |
Collapse
|
49
|
Santa Chalarca CF, Dalal RJ, Chapa A, Hanson MG, Reineke TM. Cation Bulk and p Ka Modulate Diblock Polymer Micelle Binding to pDNA. ACS Macro Lett 2022; 11:588-594. [PMID: 35575319 DOI: 10.1021/acsmacrolett.2c00015] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Polymer-based gene delivery relies on the binding, protection, and final release of nucleic acid cargo using polycations. Engineering polymeric vectors, by exploring novel topologies and cationic moieties, is a promising avenue to improve their performance, which hinges on the development of simple synthetic methods that allow facile preparation. In this work, we focus on cationic micelles formed from block polymers, which are examined as promising gene compaction agents and carriers. In this study, we report the synthesis and assembly of six amphiphilic poly(n-butyl acrylate)-b-poly(cationic acrylamide) diblock polymers with different types of cationic groups ((dialkyl)amine, morpholine, or imidazole) in their hydrophilic corona. The polycations were obtained through the parallel postpolymerization modification of a poly(n-butyl acrylate)-b-poly(pentafluorophenyl acrylate) reactive scaffold, which granted diblock polymers with equivalent degrees of polymerization and subsequent quantitative functionalization with cations of different pKa. Ultrasound-assisted direct dissolution of the polycations in different aqueous buffers (pH = 1-7) afforded micellar structures with low size dispersities and hydrodynamic radii below 100 nm. The formation and properties of micelle-DNA complexes ("micelleplexes") were explored via DLS, zeta potential, and dye-exclusion assays revealing that binding is influenced by the cation type present in the micelle corona where bulkiness and pKa are the drivers of micelleplex formation. Combining parallel synthesis strategies with simple direct dissolution formulation opens opportunities to optimize and expand the range of micelle delivery vehicles available by facile tuning of the composition of the cationic micelle corona.
Collapse
Affiliation(s)
| | - Rishad J. Dalal
- Department of Chemistry, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Alejandra Chapa
- Department of Biology, University of Texas Rio Grande Valley, Edinburg, Texas 78539, United States
| | - Mckenna G. Hanson
- Department of Chemistry, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Theresa M. Reineke
- Department of Chemistry, University of Minnesota, Minneapolis, Minnesota 55455, United States
| |
Collapse
|
50
|
Tasset A, Bellamkonda A, Wang W, Pyatnitskiy I, Ward D, Peppas N, Wang H. Overcoming barriers in non-viral gene delivery for neurological applications. NANOSCALE 2022; 14:3698-3719. [PMID: 35195645 PMCID: PMC9036591 DOI: 10.1039/d1nr06939j] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/01/2023]
Abstract
Gene therapy for neurological disorders has attracted significant interest as a way to reverse or stop various disease pathologies. Typical gene therapies involving the central and peripheral nervous system make use of adeno-associated viral vectors whose questionable safety and limitations in manufacturing has given rise to extensive research into non-viral vectors. While early research studies have demonstrated limited efficacy with these non-viral vectors, investigation into various vector materials and functionalization methods has provided insight into ways to optimize these non-viral vectors to improve desired characteristics such as improved blood-brain barrier transcytosis, improved perfusion in brain region, enhanced cellular uptake and endosomal escape in neural cells, and nuclear transport of genetic material post- intracellular delivery. Using a combination of various strategies to enhance non-viral vectors, research groups have designed multi-functional vectors that have been successfully used in a variety of pre-clinical applications for the treatment of Parkinson's disease, brain cancers, and cellular reprogramming for neuron replacement. While more work is needed in the design of these multi-functional non-viral vectors for neural applications, much of the groundwork has been done and is reviewed here.
Collapse
Affiliation(s)
- Aaron Tasset
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX, USA.
| | - Arjun Bellamkonda
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX, USA.
| | - Wenliang Wang
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX, USA.
| | - Ilya Pyatnitskiy
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX, USA.
| | - Deidra Ward
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX, USA.
| | - Nicholas Peppas
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX, USA.
- Department of Chemical Engineering, The University of Texas at Austin, Austin, TX, USA
- Institute for Biomaterials, Drug Delivery, and Regenerative Medicine, The University of Texas at Austin, Austin, TX, USA
- Division of Molecular Pharmaceutics and Drug Delivery, College of Pharmacy, The University of Texas at Austin, Austin, TX, USA
- Department of Surgery and Perioperative Care, Dell Medical School, The University of Texas at Austin, Austin, TX, USA
| | - Huiliang Wang
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX, USA.
| |
Collapse
|