1
|
Rocher EE, Luly KM, Tzeng SY, Sunshine JC, Green JJ. Efficient Polymeric Nanoparticle Gene Delivery Enabled Via Tri- and Tetrafunctional Branching. Biomacromolecules 2024; 25:7260-7273. [PMID: 39466232 DOI: 10.1021/acs.biomac.4c00954] [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: 10/29/2024]
Abstract
Poly(β-amino ester) (PBAE) nanoparticles (NPs) show great promise for nonviral gene delivery. Recent studies suggest branched PBAEs (BPBAEs) offer advantages over linear counterparts, but the effect of polymer structure has not been well investigated across many chemical constituents. Here, a library of BPBAEs was synthesized with tri- and tetrafunctional branching. These polymers self-assemble with DNA to form highly cationic, monodisperse NPs with notably small size (∼50 nm). Optimal transfection occurred with polymer structures that featured moderate PBAE branching, enabling complete DNA encapsulation, rapid NP uptake, and robust expression at low DNA doses and polymer amounts. Optimized NPs enabled efficient DNA delivery to diverse cell types in vitro while maintaining high cellular viability, demonstrating significant improvements over a well-performing linear PBAE counterpart. BPBAEs also facilitated efficient mRNA and siRNA delivery, highlighting the versatility of these structures and demonstrating the broad utility of BPBAE NPs as vectors for nucleic acid delivery.
Collapse
|
2
|
Yang X, Zhang H, Wu Z, Chen Q, Zheng W, Shen Q, Wei Q, Shen JW, Guo Y. Tumor therapy utilizing dual-responsive nanoparticles: A multifaceted approach integrating calcium-overload and PTT/CDT/chemotherapy. J Control Release 2024; 376:646-658. [PMID: 39427774 DOI: 10.1016/j.jconrel.2024.10.029] [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: 07/13/2024] [Revised: 10/08/2024] [Accepted: 10/15/2024] [Indexed: 10/22/2024]
Abstract
The advancement of rational nano drug delivery systems offers robust tools for achieving synergistic therapeutic outcomes in tumor treatment. In this study, we present the development of pH and near-infrared laser dual-responsive nanoparticles (DOX-CuS@CaCO3@PL-PEG, DCCP NPs) based on calcium carbonate, utilizing a one-pot gas diffusion reaction. These nanoparticles enable combined photothermal therapy (PTT), chemodynamic therapy (CDT), chemotherapy, and Ca2+-overloading synergistic therapy. Doxorubicin (DOX) and copper sulfide (CuS) NPs were co-loaded in CaCO3, followed by PEG surface functionalization. The presence of PEG enhanced the stability of DCCP NPs in aqueous environments. Controlled release of DOX, CuS NPs, and Ca2+ occurs specifically in the acidic tumor microenvironment. Released DOX enhances chemotherapy efficiency, while CuS NPs, upon laser irradiation, induce thermal damage, promoting further drug release and cellular uptake. Additionally, CuS NPs in our system consume excess GSH and generate toxic hydroxyl radicals (·OH) through a Fenton-like reaction, contributing to CDT. These radicals not only directly eliminate tumor cells but also disrupt mitochondrial Ca2+ buffering capacity. Furthermore, Ca2+ released from CaCO3 induces Ca2+-overloading, intensifying mitochondrial disruption and oxidative damage. The synergistic combination of PTT, CDT, chemotherapy, and Ca2+-overloading showcases significant therapeutic potential, indicating broad applications in tumor therapy. This multifaceted approach holds promise for advancing the field of tumor therapeutics.
Collapse
Affiliation(s)
- Xiaorong Yang
- School of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China
| | - Hong Zhang
- School of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China
| | - Zehua Wu
- School of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China
| | - Qin Chen
- School of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China
| | - Wei Zheng
- School of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China; Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China
| | - Qiying Shen
- School of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China; Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China
| | - Qiaolin Wei
- School of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China; Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China; State Key Lab of Silicon Materials, Zhejiang University, Hangzhou 310027, China.
| | - Jia-Wei Shen
- School of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China; Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China.
| | - Yong Guo
- School of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China; Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China.
| |
Collapse
|
3
|
Al Hoque A, Kannaboina P, Abraham Y, Mehedi M, Sibi MP, Quadir M. Furan-rich, biobased transfection agents as potential oligomeric candidates for intracellular plasmid DNA delivery. RSC Adv 2024; 14:32637-32647. [PMID: 39411251 PMCID: PMC11476585 DOI: 10.1039/d4ra05978f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2024] [Accepted: 09/23/2024] [Indexed: 10/19/2024] Open
Abstract
Biobased, DNA delivery vectors have been synthesized with a core motif composed of 2,5-bishydroxymethylfuran (BHMF) readily available from an important biomass feedstock 5-hydroxymethyl furfural (HMF). To generate the product, BHMF was first converted to 2,5-furan bishydroxymethyl diacrylate (2,5-FDA), which was later conjugated with different types of secondary amines. Rich in tertiary nitrogen, these oligomeric FDA-amino esters demonstrated stable electrostatic interactions with negatively charged plasmid DNA in an aqueous environment. We evaluated synthetic routes toward these plasmid DNA-binding amino esters (pFASTs), identified their nanoscale features, and attempted to establish their structure-property relationship in the context of the DNA delivery. Our preliminary studies show that the pFASTs formed stable complexes with the plasmid DNA. Dynamic light scattering indicated that the DNA polyplexes of pFASTs have hydrodynamic diameters within the size range of 100-150 nm with a surface charge (ζ-potential) ranging from -10 to +33 mV, depending on pFAST type. These oligomeric amino esters rich in furan motif were also found to successfully transfect the GFP-expressing plasmid DNA intracellularly. Collectively, this study establishes a new route to produce DNA transfection agents from sustainable resources that can be used for transferring genetic materials for humans, veterinary, and agrochemical purposes.
Collapse
Affiliation(s)
- Ashique Al Hoque
- Department of Coatings and Polymeric Materials, North Dakota State University Fargo ND 58108 USA +1-701-231-6283
- Department of Pharmaceutical Technology, Jadavpur University Kolkata India
| | - Prakash Kannaboina
- Department of Chemistry Biochemistry, North Dakota State University Fargo ND 58108 USA
| | - Yeabstega Abraham
- Department of Biomedical Sciences, University of North Dakota, School of Medicine & Health Grand Forks ND 58202 USA
| | - Masfique Mehedi
- Department of Biomedical Sciences, University of North Dakota, School of Medicine & Health Grand Forks ND 58202 USA
| | - Mukund P Sibi
- Department of Chemistry Biochemistry, North Dakota State University Fargo ND 58108 USA
| | - Mohiuddin Quadir
- Department of Coatings and Polymeric Materials, North Dakota State University Fargo ND 58108 USA +1-701-231-6283
| |
Collapse
|
4
|
Wang C, Wang F, Zheng Y, Bo T, Zhao Y, Yong H, Li Z, Xu W, Zhao Q, Zhou D. Enhancing Gene Transfection Efficiency of Star-Shaped Poly(β-amino ester)s via "Top-down" Hydrolysis. ACS Macro Lett 2024; 13:1218-1225. [PMID: 39236076 DOI: 10.1021/acsmacrolett.4c00399] [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: 09/07/2024]
Abstract
Gene therapy has emerged as a potent tool for treating a wide range of hereditary and acquired disorders. However, the development of high-performance nonviral gene delivery vectors remains a significant challenge. Here we report the development of a new type of star-shaped poly(β-amino ester) (SPAE) through a "top-down" hydrolysis approach and demonstrate its exceptional DNA transfection efficiency and safety profiles. Two SPAEs with different monomer combinations are first synthesized using an "arm first" strategy and then hydrolyzed sequentially to produce h-SPAEs with varied chemical compositions and molecular weights. Results demonstrate that hydrolysis significantly influences the physiological characteristics of the resulting h-SPAEs and h-SPAE/DNA polyplexes. Dependent on the chemical composition, h-SPAEs with low to moderate hydrolysis degrees exhibit superior gene transfection efficiency and cell viability across various cell types. Notably, the leading candidate, h-SPAE-1-5h, achieves up to 88.8% gene transfection efficiency, which was 154-257% higher compared to SPAE-1. This study not only establishes an easy-to-operate "top-down" approach for reshaping the topological structure and chemical composition of SPAEs, but also identifies promising candidates for effective gene transfection. This strategy can be applied to other cationic polymers to enhance their gene transfection performance.
Collapse
Affiliation(s)
- Chenfei Wang
- School of Chemical Engineering and Technology, Xi'an Jiaotong University, Xi'an 710049, China
- Department of Dermatology, Children's Hospital of Fudan University, National Children's Medical Center, Shanghai 201102, China
| | - Feifei Wang
- Department of Anesthesiology, The Second Affiliated Hospital of Air Force Medical University, Xi'an 710032, China
| | - Yuxing Zheng
- School of Chemical Engineering and Technology, Xi'an Jiaotong University, Xi'an 710049, China
| | - Tao Bo
- Department of Dermatology, Children's Hospital of Fudan University, National Children's Medical Center, Shanghai 201102, China
| | - Yitong Zhao
- Department of Dermatology, Children's Hospital of Fudan University, National Children's Medical Center, Shanghai 201102, China
| | - Haiyang Yong
- School of Chemical Engineering and Technology, Xi'an Jiaotong University, Xi'an 710049, China
| | - Zhili Li
- School of Chemical Engineering and Technology, Xi'an Jiaotong University, Xi'an 710049, China
| | - Weiyi Xu
- Department of Dermatology, Children's Hospital of Fudan University, National Children's Medical Center, Shanghai 201102, China
| | - Qiang Zhao
- Department of Dermatology, The Second Affiliated Hospital, Xi'an Jiaotong University, Xi'an 710004, China
| | - Dezhong Zhou
- School of Chemical Engineering and Technology, Xi'an Jiaotong University, Xi'an 710049, China
| |
Collapse
|
5
|
Leyden MC, Oviedo F, Saxena S, Kumar R, Le N, Reineke TM. Synergistic Polymer Blending Informs Efficient Terpolymer Design and Machine Learning Discerns Performance Trends for pDNA Delivery. Bioconjug Chem 2024; 35:897-911. [PMID: 38924453 DOI: 10.1021/acs.bioconjchem.4c00028] [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: 06/28/2024]
Abstract
Cationic polymers offer an alternative to viral vectors in nucleic acid delivery. However, the development of polymer vehicles capable of high transfection efficiency and minimal toxicity has remained elusive, and continued exploration of the vast design space is required. Traditional single polymer syntheses with large monomer bases are very time-intensive, limiting the speed at which new formulations are identified. In this work, we present an experimental method for the quick probing of the design space, utilizing a combinatorial set of 90 polymer blends, derived from 6 statistical copolymers, to deliver pDNA. This workflow facilitated rapid screening of polyplex compositions, successfully tailoring polyplex hydrophobicity, particle size, and payload binding affinity. This workflow identified blended polyplexes with high levels of transfection efficiency and cell viability relative to single copolymer controls and commercial JetPEI, indicating synergistic benefits from copolymer blending. Polyplex composition was coupled with biological outputs to guide the synthesis of single terpolymer vehicles, with high-performing polymers P10 and M20, providing superior transfection of HEK293T cells in serum-free and serum-containing media, respectively. Machine learning coupled with SHapley Additive exPlanations (SHAP) was used to identify polymer/polyplex attributes that most impact transfection efficiency, viability, and overall effective efficiency. Subsequent transfections on ARPE-19 and HDFn cells found that P10 and M20 were surpassed in performance by M10, contrasting with results in HEK293T cells. This cell type dependency reinforced the need to evaluate transfection conditions with multiple cell models to potentially identify moieties more beneficial to delivery in certain tissues. Overall, the workflow employed can be used to expedite the exploration of the polymer design space, bypassing extensive synthesis, and to develop improved polymer delivery vehicles more readily for nucleic acid therapies.
Collapse
Affiliation(s)
- Michael C Leyden
- Department of Chemical Engineering and Materials Science, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Felipe Oviedo
- Nanite Inc., Boston, Massachusetts 02109, United States
| | - Sonashree Saxena
- Department of Chemistry, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Ramya Kumar
- Department of Chemical and Biological Engineering, Colorado School of Mines, Golden, Colorado 80401, United States
| | - Ngoc Le
- 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
|
6
|
Yong H, Lin L, Li Z, Guo R, Wang C, Liu S, Zhou D. Tailoring Highly Branched Poly(β-amino ester)s for Efficient and Organ-Selective mRNA Delivery. NANO LETTERS 2024. [PMID: 39013032 DOI: 10.1021/acs.nanolett.4c02440] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/18/2024]
Abstract
Development of mRNA therapeutics necessitates targeted delivery technology, while the clinically advanced lipid nanoparticles face difficulty for extrahepatic delivery. Herein, we design highly branched poly(β-amino ester)s (HPAEs) for efficacious organ-selective mRNA delivery through tailoring their chemical compositions and topological structures. Using an "A2+B3+C2" Michael addition platform, a combinatorial library of 219 HPAEs with varied backbone structures, terminal groups, and branching degrees are synthesized. The branched topological structures of HPAEs provide enhanced serum resistance and significantly higher mRNA expression in vivo. The terminal amine structures of HPAEs determine the organ-selectivity of mRNA delivery following systemic administration: morpholine facilitates liver targeting, ethylenediamine favors spleen delivery, while methylpentane enables mRNA delivery to the liver, spleen, and lungs simultaneously. This study represents a comprehensive exploration of the structure-activity relationship governing both the efficiency and organ-selectivity of mRNA delivery by HPAEs, suggesting promising candidates for treating various organ-related diseases.
Collapse
Affiliation(s)
- Haiyang Yong
- School of Chemical Engineering and Technology, Xi'an Jiaotong University, Xi'an 710049, China
| | - Lixin Lin
- College of Pharmaceutical Sciences, Liangzhu Laboratory, Zhejiang University, Hangzhou 310058, China
| | - Zhili Li
- School of Chemical Engineering and Technology, Xi'an Jiaotong University, Xi'an 710049, China
| | - Rui Guo
- School of Chemical Engineering and Technology, Xi'an Jiaotong University, Xi'an 710049, China
| | - Chenfei Wang
- School of Chemical Engineering and Technology, Xi'an Jiaotong University, Xi'an 710049, China
| | - Shuai Liu
- College of Pharmaceutical Sciences, Liangzhu Laboratory, Zhejiang University, Hangzhou 310058, China
| | - Dezhong Zhou
- School of Chemical Engineering and Technology, Xi'an Jiaotong University, Xi'an 710049, China
| |
Collapse
|
7
|
Pan C, Wang C, Zhao Y, Bo T, Han L, Yao D, Wang Y, Wang X, Shi L, Zhao A, Cao Q, Chen F, He W, Ye Y, Zhang S, Li M. Superior COL7A1 and TGM1 gene expression in difficult-to-transfect skin cell mediated by highly branched poly(β-amino esters) through stepwise fractionation. J Control Release 2024; 370:82-94. [PMID: 38643938 DOI: 10.1016/j.jconrel.2024.04.030] [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: 11/29/2023] [Revised: 03/24/2024] [Accepted: 04/18/2024] [Indexed: 04/23/2024]
Abstract
Delivering functional gene into targeted skin cells or tissues to modulate the genes expression, has the potential to treat various hereditary cutaneous disorders. Nevertheless, the lack of safe and effective gene delivery vehicles greatly limits the clinical translation of gene therapy for inherited skin diseases. Herein, we developed a facile elution fractionation strategy to isolate eight HPAEs with Mw ranging from 7.6 to 131.8 kg/mol and Đ < 2.0 from the one crude HPAE23.7k, and investigated the expression efficiency for TGM1 and COL7A1 plasmids. Gene transfection results revealed that the intermediate MW HPAEs, HPAE20.6k, exhibited the highest gene transfection efficiency (46.4%) and the strongest mean fluorescence intensity (143,032 RLU), compared to other isolated components and the crude product. Importantly, best-performing isolated HPAE effectively delivered COL7A1 (15,974 bp) and TGM1 (7181 bp) plasmids, promoting the efficient expression of type VII collagen (C7) and transglutaminase-1 proteins in cutaneous cells. Our study establishes a straightforward step-by-step elution fractionation strategy for the development of HPAEs gene delivery vectors, expediting their clinical translation in inherited skin diseases.
Collapse
Affiliation(s)
- Chaolan Pan
- Department of Dermatology, Children's Hospital of Fudan University, National Children's Medical Center, Shanghai 201102, China
| | - Chenfei Wang
- Department of Dermatology, Children's Hospital of Fudan University, National Children's Medical Center, Shanghai 201102, China.
| | - Yitong Zhao
- School of Medicine, Anhui University of Science and Technology, Huainan, Anhui 232000, China
| | - Tao Bo
- NHC Key Laboratory of Glycoconjugate Research, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, Shanghai 200032, China
| | - Liping Han
- NHC Key Laboratory of Glycoconjugate Research, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, Shanghai 200032, China; Department of Blood Transfusion, Huashan Hospital, Fudan University, Shanghai, China
| | - Dingjin Yao
- Shanghai EditorGene Technology Co., Ltd, Shanghai, 200000, China
| | - Yumeng Wang
- Department of Dermatology, Children's Hospital of Fudan University, National Children's Medical Center, Shanghai 201102, China
| | - Xiaoxiao Wang
- Department of Dermatology, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200092, China
| | - Linjing Shi
- Department of Dermatology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, China
| | - Anqi Zhao
- Department of Dermatology, Children's Hospital of Fudan University, National Children's Medical Center, Shanghai 201102, China
| | - Qiaoyu Cao
- Department of Dermatology, Children's Hospital of Fudan University, National Children's Medical Center, Shanghai 201102, China
| | - Fuying Chen
- Department of Dermatology, Children's Hospital of Fudan University, National Children's Medical Center, Shanghai 201102, China
| | - Wei He
- Department of Dermatology, Children's Hospital of Fudan University, National Children's Medical Center, Shanghai 201102, China
| | - Ying Ye
- Department of Dermatology, Children's Hospital of Fudan University, National Children's Medical Center, Shanghai 201102, China
| | - Si Zhang
- NHC Key Laboratory of Glycoconjugate Research, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, Shanghai 200032, China.
| | - Ming Li
- Department of Dermatology, Children's Hospital of Fudan University, National Children's Medical Center, Shanghai 201102, China.
| |
Collapse
|
8
|
Li Z, Guo R, Zhang Z, Yong H, Guo L, Chen Z, Huang D, Zhou D. Enhancing gene transfection of poly(β-amino ester)s through modulation of amphiphilicity and chain sequence. J Control Release 2024; 368:131-139. [PMID: 38331003 DOI: 10.1016/j.jconrel.2024.02.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2023] [Revised: 01/31/2024] [Accepted: 02/05/2024] [Indexed: 02/10/2024]
Abstract
Poly(β-amino ester)s (PAEs) have emerged as a type of highly safe and efficient non-viral DNA delivery vectors. However, the influence of amphiphilicity and chain sequence on DNA transfection efficiency and safety profile remain largely unexplored. In this study, four PAEs with distinct amphiphilicity and chain sequences were synthesized. Results show that both amphiphilicity and chain sequence significantly affect the DNA binding and condensation ability of PAEs, as well as size, zeta potential and cellular uptake of PAE/DNA polyplexes. PAEs with different amphiphilicity and chain sequence exhibit cell type-dependent transfection capabilities: in human bladder transitional cell carcinoma (UM-UC-3), hydrophilic PAE (P-Philic) and amphiphilic PAE random copolymer (R-Amphilic) exhibit relatively higher gene transfection efficiency, while in human bladder epithelial immortalized cells (SV-HUC-1), hydrophobic PAE (P-Phobic), R-Amphilic, and amphiphilic PAE block copolymer (B-Amphilic) demonstrate higher transfection capability. Regardless of cell types, amphiphilic PAE block copolymer (B-Amphilic) always exhibits much lower gene transfection efficiency. In addition, in human colon cancer cells (HCT-116), P-Philic and R-Amphilic achieved superior gene transfection efficiency at high and low polymer/DNA weight ratios, respectively. Importantly, R-Amphilic can effectively deliver the gene encoding tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) to human chondrosarcoma cells SW1353 to induce their apoptosis, highlighting its potential application in cancer gene therapy. This study not only establishes a new paradigm for enhancing the gene transfection efficiency of PAEs by modulating their amphiphilicity and chain sequence but also identifies R-Amphilic as a potential candidate for the effective delivery of TRAIL gene in cancer gene therapy.
Collapse
Affiliation(s)
- Zhili Li
- School of Chemical Engineering and Technology, Xi'an Jiaotong University, Xi'an 710049, China
| | - Rui Guo
- School of Chemical Engineering and Technology, Xi'an Jiaotong University, Xi'an 710049, China
| | - Zhiyong Zhang
- Department of Colorectal and Anal Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China.
| | - Haiyang Yong
- School of Chemical Engineering and Technology, Xi'an Jiaotong University, Xi'an 710049, China.
| | - Lei Guo
- Pooling Institute of Translational Medicine, Hangzhou 311100, China
| | - Zhengju Chen
- Pooling Medical Research Institutes of 100Biotech, Beijing 100006, China
| | - Dongdong Huang
- Pooling Institute of Translational Medicine, Hangzhou 311100, China
| | - Dezhong Zhou
- School of Chemical Engineering and Technology, Xi'an Jiaotong University, Xi'an 710049, China.
| |
Collapse
|
9
|
Wang X, Li Y, A S, Lyu J, Wang X, He Z, Lara-Sáez I, Li M, Wang W. Cyclization-enhanced poly(β-amino ester)s vectors for efficient CRISPR gene editing therapy. J Control Release 2024; 368:444-452. [PMID: 38401849 DOI: 10.1016/j.jconrel.2024.02.032] [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: 11/28/2023] [Revised: 01/29/2024] [Accepted: 02/21/2024] [Indexed: 02/26/2024]
Abstract
Among non-viral gene delivery vectors, poly(β-amino ester)s (PAEs) are one of the most versatile candidates because of their wide monomer availability, high polymer flexibility, and superior gene transfection performance both in vitro and in vivo. Over two decades, PAEs have evolved from linear to highly branched structures, significantly enhancing gene delivery efficacy. Building on the proven efficient sets of monomers in highly branched PAEs (HPAEs), this work introduced a new class of cyclic PAEs (CPAEs) constructed via an A2 + B4 + C2 cyclization synthesis strategy and identified their markedly improved gene transfection capabilities in gene delivery applications. Two sets of cyclic PAEs (CPAEs) with rings of different sizes and topologies were obtained. Their chemical structures were confirmed via two-dimensional nuclear magnetic resonance and the photoluminescence phenomena, and their DNA delivery behaviours were investigated and compared with the HPAE counterparts. In vitro assessments demonstrated that the CPAEs with a macrocyclic architecture (MCPAEs), significantly enhanced DNA intracellular uptake and facilitated efficient gene expression while maintaining perfect biocompatibility. The top-performance MCPAEs have been further employed to deliver a plasmid coding dual single guide RNA-guided CRISPR-Cas9 machinery to delete COL7A1 exon 80 containing the c.6527dupC mutation. In recessive dystrophic epidermolysis bullosa (RDEB) patient-derived epidermal keratinocytes, MCPAEs facilitated the CRISPR plasmid delivery and achieved efficient targeted gene editing in multiple colonies.
Collapse
Affiliation(s)
- Xianqing Wang
- Charles Institute of Dermatology, School of Medicine, University College Dublin, D04 V1W8 Dublin, Ireland
| | - Yinghao Li
- Charles Institute of Dermatology, School of Medicine, University College Dublin, D04 V1W8 Dublin, Ireland
| | - Sigen A
- School of Medicine, Anhui University of Science and Technology, Huainan 232001, China
| | - Jing Lyu
- Charles Institute of Dermatology, School of Medicine, University College Dublin, D04 V1W8 Dublin, Ireland.
| | - Xi Wang
- Institute of Precision Medicine (AUST-IPM), Anhui University of Science and Technology, Huainan 232001, China
| | - Zhonglei He
- Institute of Precision Medicine (AUST-IPM), Anhui University of Science and Technology, Huainan 232001, China
| | - Irene Lara-Sáez
- Charles Institute of Dermatology, School of Medicine, University College Dublin, D04 V1W8 Dublin, Ireland
| | - Ming Li
- Department of Dermatology, Children's Hospital of Fudan University, National Children's Medical Center, 399 Wanyuan Road, Shanghai, China
| | - Wenxin Wang
- Charles Institute of Dermatology, School of Medicine, University College Dublin, D04 V1W8 Dublin, Ireland.
| |
Collapse
|
10
|
Chu Z, Li Z, Yong H, Che D, Li B, Yan C, Zhou T, Wang X, Feng Y, Guo K, Geng S. Enhanced gene transfection and induction of apoptosis in melanoma cells by branched poly(β-amino ester)s with uniformly distributed branching units. J Control Release 2024; 367:197-208. [PMID: 38246205 DOI: 10.1016/j.jconrel.2024.01.026] [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: 11/09/2023] [Revised: 01/05/2024] [Accepted: 01/13/2024] [Indexed: 01/23/2024]
Abstract
Melanoma, one of the most devastating forms of skin cancer, currently lacks effective clinical treatments. Delivery of functional genes to modulate specific protein expression to induce melanoma cell apoptosis could be a promising therapeutic approach. However, transfecting melanoma cells using non-viral methods, particularly with cationic polymers, presents significant challenges. In this study, we synthesized three branched poly(β-amino ester)s (HPAEs) with evenly distributed branching units but varying space lengths through a two-step "oligomer combination" strategy. The unique topological structure enables HPAEs to condense DNA to form nano-sized polyplexes with favorable physiochemical properties. Notably, HPAEs, especially HPAE-2 with intermediate branching unit space length, demonstrated significantly higher gene transfection efficiency than the leading commercial gene transfection reagent, jetPRIME, in human melanoma cells. Furthermore, HPAE-2 efficiently delivered the Bax-encoding plasmid into melanoma cells, leading to a pronounced pro-apoptotic effect without causing noticeable cytotoxicity. This study establishes a potent non-viral platform for gene transfection of melanoma cells by harnessing the distribution of branching units, paving the way for potential clinical applications of gene therapy in melanoma treatment.
Collapse
Affiliation(s)
- Zhaowei Chu
- Department of Dermatology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, China
| | - Zhili Li
- School of Chemical Engineering and Technology, Xi'an Jiaotong University, Xi'an 710049, China.
| | - Haiyang Yong
- School of Chemical Engineering and Technology, Xi'an Jiaotong University, Xi'an 710049, China
| | - Delu Che
- Department of Dermatology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, China
| | - Bingjie Li
- Department of Dermatology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, China
| | - Cong Yan
- Department of Dermatology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, China
| | - Tong Zhou
- Department of Dermatology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, China
| | - Xi Wang
- Department of Dermatology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, China
| | - Yuqing Feng
- Department of Dermatology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, China
| | - Kun Guo
- Department of Dermatology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, China
| | - Songmei Geng
- Department of Dermatology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, China.
| |
Collapse
|
11
|
Li Y, Qiu B, Li Z, Wang X, He Z, Sandoval DM, Song R, Sigen A, Zhao C, Johnson M, Lyu J, Lara-Sáez I, Wang W. Backbone cationized highly branched poly(β-amino ester)s as enhanced delivery vectors in non-viral gene therapy. J Control Release 2024; 367:327-338. [PMID: 38272397 DOI: 10.1016/j.jconrel.2024.01.046] [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: 11/28/2023] [Revised: 01/19/2024] [Accepted: 01/22/2024] [Indexed: 01/27/2024]
Abstract
Gene therapy holds great potential for treating Lung Cystic Fibrosis (CF) which is a fatal hereditary condition arising from mutations in the CF transmembrane conductance regulator (CFTR) gene, resulting in dysfunctional CFTR protein. However, the advancement and clinical application of CF gene therapy systems have been hindered due to the absence of a highly efficient delivery vector. In this work, we introduce a new generation of highly branched poly(β-amino ester) (HPAE) gene delivery vectors for CF treatment. Building upon the classical chemical composition of HPAE, a novel backbone cationization strategy was developed to incorporate additional functional amine groups into HPAE without altering their branching degree. By carefully adjusting the type, proportion, and backbone distribution of the added cationic groups, a series of highly effective HPAE gene delivery vectors were successfully constructed for CF disease gene therapy. In vitro assessment results showed that the backbone cationized HPAEs with randomly distributed 10% proportion of 1-(3-aminopropyl)-4-methylpiperazine (E7) amine groups exhibited superior transfection performance than their counterparts. Furthermore, the top-performed backbone cationized HPAEs, when loaded with therapeutic plasmids, successfully reinstated CFTR protein expression in the CFBE41o- disease model, achieving levels 20-23 times higher than that of normal human bronchial epithelial (HBE) cells. Their therapeutic effectiveness significantly surpassed that of the currently advanced commercial vectors, Xfect and Lipofectamine 3000.
Collapse
Affiliation(s)
- Yinghao Li
- Institute of Precision Medicine (AUST-IPM), Anhui University of Science and Technology, Huainan 232001, China; Charles Institute of Dermatology, School of Medicine, University College Dublin, D04 V1W8 Dublin, Ireland
| | - Bei Qiu
- Charles Institute of Dermatology, School of Medicine, University College Dublin, D04 V1W8 Dublin, Ireland
| | - Zishan Li
- Charles Institute of Dermatology, School of Medicine, University College Dublin, D04 V1W8 Dublin, Ireland
| | - Xianqing Wang
- Charles Institute of Dermatology, School of Medicine, University College Dublin, D04 V1W8 Dublin, Ireland
| | - Zhonglei He
- Institute of Precision Medicine (AUST-IPM), Anhui University of Science and Technology, Huainan 232001, China
| | - Darío Manzanares Sandoval
- Charles Institute of Dermatology, School of Medicine, University College Dublin, D04 V1W8 Dublin, Ireland
| | - Rijian Song
- Charles Institute of Dermatology, School of Medicine, University College Dublin, D04 V1W8 Dublin, Ireland
| | - A Sigen
- School of Medicine, Anhui University of Science and Technology, Huainan 232001, China
| | - Chunyu Zhao
- Charles Institute of Dermatology, School of Medicine, University College Dublin, D04 V1W8 Dublin, Ireland
| | - Melissa Johnson
- Charles Institute of Dermatology, School of Medicine, University College Dublin, D04 V1W8 Dublin, Ireland
| | - Jing Lyu
- Charles Institute of Dermatology, School of Medicine, University College Dublin, D04 V1W8 Dublin, Ireland..
| | - Irene Lara-Sáez
- Charles Institute of Dermatology, School of Medicine, University College Dublin, D04 V1W8 Dublin, Ireland
| | - Wenxin Wang
- Institute of Precision Medicine (AUST-IPM), Anhui University of Science and Technology, Huainan 232001, China; Charles Institute of Dermatology, School of Medicine, University College Dublin, D04 V1W8 Dublin, Ireland..
| |
Collapse
|
12
|
Zhou YN, Yong H, Guo R, Wang K, Li Z, Hua W, Zhou D. Self-reporting and Biodegradable Thermosetting Solid Polymer Electrolyte. Angew Chem Int Ed Engl 2024; 63:e202319003. [PMID: 38131604 DOI: 10.1002/anie.202319003] [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: 12/10/2023] [Revised: 12/19/2023] [Accepted: 12/21/2023] [Indexed: 12/23/2023]
Abstract
To date, significant efforts have been dedicated to improve their ionic conductivity, thermal stability, and mechanical strength of solid polymer electrolytes (SPEs). However, direct monitoring of physical and chemical changes in SPEs is still lacking. Moreover, existing thermosetting SPEs are hardly degradable. Herein, by overcoming the limitation predicted by Flory theory, self-reporting and biodegradable thermosetting hyperbranched poly(β-amino ester)-based SPEs (HPAE-SPEs) are reported. HPAE is successfully synthesized through a well-controlled "A2+B4" Michael addition strategy and then crosslinked it in situ to produce HPAE-SPEs. The multiple tertiary aliphatic amines at the branching sites confer multicolour luminescence to HPAE-SPEs, enabling direct observation of its physical and chemical damage. After use, HPAE-SPEs can be rapidly hydrolysed into non-hazardous β-amino acids and polyols via self-catalysis. Optimized HPAE-SPE exhibits an ionic conductivity of 1.3×10-4 S/cm at 60 °C, a Na+ transference number (t N a + ${{t}_{Na}^{+}}$ ) of 0.67, a highly stable sodium plating-stripping behaviour and a low overpotential of ≈190 mV. This study establishes a new paradigm for developing SPEs by engineering multifunctional polymers. The self-reporting and biodegradable properties would greatly expand the scope of applications for SPEs.
Collapse
Affiliation(s)
- Ya-Nan Zhou
- School of Chemical Engineering and Technology, Xi'an Jiaotong University, Xi'an, Shaanxi, 710049, P. R. China
| | - Haiyang Yong
- School of Chemical Engineering and Technology, Xi'an Jiaotong University, Xi'an, Shaanxi, 710049, P. R. China
| | - Rui Guo
- School of Chemical Engineering and Technology, Xi'an Jiaotong University, Xi'an, Shaanxi, 710049, P. R. China
| | - Kaixuan Wang
- School of Chemical Engineering and Technology, Xi'an Jiaotong University, Xi'an, Shaanxi, 710049, P. R. China
| | - Zhili Li
- School of Chemical Engineering and Technology, Xi'an Jiaotong University, Xi'an, Shaanxi, 710049, P. R. China
| | - Weibo Hua
- School of Chemical Engineering and Technology, Xi'an Jiaotong University, Xi'an, Shaanxi, 710049, P. R. China
| | - Dezhong Zhou
- School of Chemical Engineering and Technology, Xi'an Jiaotong University, Xi'an, Shaanxi, 710049, P. R. China
| |
Collapse
|