1
|
Li M, Li Y, Yao X, Liu Y, Cai K, Yang H, Luo Z. Optically controllable nanoregulators enable tumor-specific pro-ferroptosis lipometabolic reprogramming for in-situ adjuvant-free vaccination. Biomaterials 2025; 317:123096. [PMID: 39805186 DOI: 10.1016/j.biomaterials.2025.123096] [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: 10/29/2024] [Revised: 12/26/2024] [Accepted: 01/07/2025] [Indexed: 01/16/2025]
Abstract
In-situ tumor vaccination remains challenging due to difficulties in the exposure and presentation of tumor-associated neoantigens (TANs). In view of the central role of lipid metabolism in cell fate determination and tumor-immune cell communication, here we report a photo-controlled lipid metabolism nanoregulator (PLMN) to achieve robust in-situ adjuvant-free vaccination, which is constructed through hierarchically integrating photothermal-inducible arachidonate 15-lipoxygenase (ALOX15)-expressing plasmids, cypate and FIN56 into cationic liposomes. Near-infrared light (NIR) stimulation triggers on-demand ALOX15 editing and causes excessive accumulation of downstream pro-ferroptosis lipid metabolites. PLMN treatment enables efficient TAN release through ferroptosis-dependent membrane perturbation and facilitates their capture and processing by antigen-presenting cells via cationic lipid-mediated TAN enrichment. Meanwhile, upregulation of ALOX15-associated lipid metabolites also enhances M2-to-M1 phenotypic transition of tumor-associated macrophages through regulating tumor-macrophage metabolic crosstalk. PLMN treatment significantly enhance the robustness and durability of adaptive antitumor immunity in vivo, offering an approach for in-situ tumor vaccination in the clinic.
Collapse
Affiliation(s)
- Menghuan Li
- School of Life Science, Chongqing University, Chongqing, 400044, China
| | - Yanan Li
- School of Life Science, Chongqing University, Chongqing, 400044, China
| | - Xuemei Yao
- School of Life Science, Chongqing University, Chongqing, 400044, China
| | - Yingqi Liu
- School of Life Science, Chongqing University, Chongqing, 400044, China
| | - Kaiyong Cai
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, Chongqing University, Chongqing, 400044, China
| | - Huocheng Yang
- School of Life Science, Chongqing University, Chongqing, 400044, China.
| | - Zhong Luo
- School of Life Science, Chongqing University, Chongqing, 400044, China.
| |
Collapse
|
2
|
Huang Y, Li H, Qi L, Wang Z, Liu Z, Wu R, Chen Q, Zhu C, Sun D, Liu L, Zhang L, Feng G. NanoCRISPR-assisted biomimetic tissue-equivalent patch regenerates the intervertebral disc by inhibiting endothelial-to-mesenchymal transition. Biomaterials 2025; 322:123404. [PMID: 40398216 DOI: 10.1016/j.biomaterials.2025.123404] [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: 09/14/2024] [Revised: 04/18/2025] [Accepted: 05/09/2025] [Indexed: 05/23/2025]
Abstract
The integrity of the intervertebral disc (IVD), an immune-privileged organ protected by the blood-disc barrier, is compromised following annulus fibrosus (AF) injury. This breach facilitates angiogenesis, immune cell infiltration, and inflammation, accelerating intervertebral disc degeneration (IDD) and resulting in various clinical disorders. Current treatments fail to adequately address biological repair of AF defects and angiogenesis. Single-cell RNA sequencing analyses reveal that vascular endothelial growth factor (VEGF), secreted by IDD-associated fibrochondrocytes, is crucial in promoting angiogenesis by inducing endothelial-to-mesenchymal transition (EndoMT). This study proposes a nano-clustered regularly interspaced short palindromic repeats (CRISPR)-assisted AF patch with an aligned, polydopamine-modified nano-lamellae nanofibrous scaffold that replicates the hierarchical structure of natural AF, providing a conducive microenvironment for AF repair. A zeolitic imidazolate framework-8-based nanoCRISPR system encapsulates the CRISPR/CRISPR-associated protein 9 complex to target and eliminate VEGF-mediated angiogenic factors. In vitro studies demonstrate that the nanoCRISPR-assisted patch can enhance AF cell adhesion and migration, promote extracellular matrix deposition, knock out VEGF expression, and inhibit EndoMT. In vivo studies show its significant efficacy in promoting AF repair, inhibiting abnormal angiogenesis, and delaying IDD progression. This study presents a promising approach for structural and biological AF regeneration, addressing physical and angiogenic barriers in IVD regeneration.
Collapse
Affiliation(s)
- Yong Huang
- Department of Orthopedics Surgery and Orthopedic Research Institute, Analytical & Testing Center, West China Hospital, Sichuan University, Chengdu, 610065, China
| | - Hao Li
- Department of Orthopedics Surgery and Orthopedic Research Institute, Analytical & Testing Center, West China Hospital, Sichuan University, Chengdu, 610065, China
| | - Lin Qi
- Department of Orthopedics Surgery and Orthopedic Research Institute, Analytical & Testing Center, West China Hospital, Sichuan University, Chengdu, 610065, China
| | - Zhe Wang
- Department of Orthopedics Surgery and Orthopedic Research Institute, Analytical & Testing Center, West China Hospital, Sichuan University, Chengdu, 610065, China
| | - Zheng Liu
- Department of Orthopedics Surgery and Orthopedic Research Institute, Analytical & Testing Center, West China Hospital, Sichuan University, Chengdu, 610065, China
| | - Ruibang Wu
- Department of Orthopedics Surgery and Orthopedic Research Institute, Analytical & Testing Center, West China Hospital, Sichuan University, Chengdu, 610065, China
| | - Qian Chen
- Department of Orthopedics Surgery and Orthopedic Research Institute, Analytical & Testing Center, West China Hospital, Sichuan University, Chengdu, 610065, China
| | - Ce Zhu
- Department of Orthopedics Surgery and Orthopedic Research Institute, Analytical & Testing Center, West China Hospital, Sichuan University, Chengdu, 610065, China
| | - Dan Sun
- Advanced Composite Research Group (ACRG), School of Mechanical and Aerospace Engineering, Queens University Belfast, Belfast, BT9 5AH, UK
| | - Limin Liu
- Department of Orthopedics Surgery and Orthopedic Research Institute, Analytical & Testing Center, West China Hospital, Sichuan University, Chengdu, 610065, China
| | - Li Zhang
- Department of Orthopedics Surgery and Orthopedic Research Institute, Analytical & Testing Center, West China Hospital, Sichuan University, Chengdu, 610065, China.
| | - Ganjun Feng
- Department of Orthopedics Surgery and Orthopedic Research Institute, Analytical & Testing Center, West China Hospital, Sichuan University, Chengdu, 610065, China.
| |
Collapse
|
3
|
Xian H, Song Y, Qu J, Shi Y, Zhang Y, Wu W, Kim M, Wang Y, Yu C. CaClOH-Modified Silica Nanoparticles for mRNA Delivery. NANO LETTERS 2025; 25:6365-6373. [PMID: 40214747 DOI: 10.1021/acs.nanolett.4c05615] [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: 04/24/2025]
Abstract
Messenger RNA (mRNA) technology has attracted wide attention in biomedical applications; its success relies heavily on the development of effective delivery tools. Herein, we report the synthesis of a novel CaClOH-modified silica nanoparticle (SNP-CaClOH) with a spiky surface for mRNA delivery. SNP-CaClOH is obtained by a rationally designed thermal decomposition process of hydrated CaCl2 inside the silanol-rich mesopores of preformed spiky SNPs. When used as a carrier for the cellular delivery of mRNA, the unique composition of CaClOH offers alkalinity to SNP-CaClOH that promotes endosomal escape via the proton sponge effect. Moreover, SNP-CaClOH leads to an increased intracellular Ca2+ level to activate the mammalian target of rapamycin complex 1 (mTORC1) by interacting with calmodulin (CaM) for enhanced mRNA translation. Taking further advantage of the spiky nanotopography, the superior mRNA delivery performance of SNP-CaClOH is demonstrated both in vitro and in vivo, providing useful delivery tools for mRNA technology development.
Collapse
Affiliation(s)
- He Xian
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Yaping Song
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Jingjing Qu
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Yiru Shi
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Yue Zhang
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Weixi Wu
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Minjun Kim
- 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
| | - Chengzhong Yu
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, QLD 4072, Australia
| |
Collapse
|
4
|
Singh AK, Goel K, Dhanawat M. Plasmid DNA and mRNA delivery: Approaches and challenges. Adv Immunol 2025; 165:63-87. [PMID: 40449975 DOI: 10.1016/bs.ai.2024.12.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/03/2025]
Abstract
for delivery of plasmid DNA and mRNA transform biology and medicine, offering powerful tools for gene therapy, vaccine development, cancer immunotherapy, and regenerative medicine. Plasmid DNA provides a relatively stable and sustained expression of the genes which also provides the basic groundwork for long-lasting therapeutic. At the same time, mRNA has also demonstrated more appropriateness for dynamic and time-sensitive applications due to its short-lived and accurate translation capabilities, such as during the development of mRNA-based COVID-19 vaccines. Despite their unique advantages, however, the efficient delivery of these biomolecules poses challenges including immune system activation, enzymatic degradation, and limited cellular uptake. The structural and functional features of plasmid DNA and mRNA highlighted the positive functions that underpin their complementary roles in next-generation biomedical applications. In addition, it highlights the novel delivery routes across lipid nanoparticles, polymeric systems, biomimetic carriers, and hybrid applied sciences which can resolve long-standing challenges to efficient distribution. Emerging technologies such as CRISPR gene editing, self-amplifying RNA, and multiplexed nanoparticles are also increasing the utility of these systems. Significant advances in the delivery of plasmid DNA and mRNA molecules have revolutionized vaccine development, opened new avenues in personalized medicine, and have also inspired a future with engineerable tissues. As these innovations develop, they are predicted to go beyond current limitations and bring around a fresh era of accurate medication taking on one of the global healthcare's most complex challenges. Our revolutionary delivery methods provide stability and simplicity, transforming medical advances.
Collapse
Affiliation(s)
- Arun Kumar Singh
- Department of Pharmacy, Vivekananda Global University, Jaipur, Rajasthan, India.
| | - Karan Goel
- M.M College of Pharmacy, Maharishi Markandeshwar (Deemed to be University), Mullana-Ambala, Haryana, India
| | - Meenakshi Dhanawat
- Amity Institute of Pharmacy, Amity University Haryana, Amity Education Valley, Panchgaon, Manesar, Gurugram, Haryana, India
| |
Collapse
|
5
|
Mamidi N, Franco De Silva F, Orash Mahmoudsalehi A. Advanced disease therapeutics using engineered living drug delivery systems. NANOSCALE 2025; 17:7673-7696. [PMID: 40040419 DOI: 10.1039/d4nr05298f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/06/2025]
Abstract
Biological barriers significantly impede the delivery of nanotherapeutics to diseased tissues, diminishing therapeutic efficacy across pathologies such as cancer and inflammatory disorders. Although conventional strategies integrate multifunctional designs and molecular components into nanomaterials (NMs), many approaches remain insufficient to overcome these barriers. Key challenges, including inadequate drug accumulation at target sites and nonspecific biodistribution, persist in nanotherapeutic development. NMs, which harness the ability to precisely modulate drug delivery spatiotemporally and control release kinetics, represent a transformative platform for targeted cancer therapy. In this review, we highlight the biological obstacles limiting effective cancer treatment and evaluate how stimuli-responsive NMs address these constraints. By leveraging exogenous and endogenous stimuli, such NMs improve therapeutic specificity, reduce off-target effects, and amplify drug activity within pathological microenvironments. We systematically analyze the rational design and synthesis of stimuli-responsive NMs, driven by advances in oncology, biomaterials science, and nanoscale engineering. Furthermore, we highlight advances across NM classes-including polymeric, lipid-based, inorganic, and hybrid systems and explore functionalization approaches using targeting ligands, antibodies, and biomimetic coatings. Diverse delivery strategies are evaluated, such as small-molecule prodrug activation, peptide- and protein-based targeting, nucleic acid payloads, and engineered cell-mediated transport. Despite the promise of stimuli-responsive NMs, challenges such as biocompatibility, scalable fabrication, and clinical translation barriers must be addressed. By elucidating structure-function relationships and refining stimulus-triggered mechanisms, these NMs pave the way for transformative precision oncology strategies, enabling patient-specific therapies with enhanced efficacy and safety. This synthesis of interdisciplinary insights aims to catalyze innovation in next-generation nanomedicine for cancer treatment.
Collapse
Affiliation(s)
- Narsimha Mamidi
- Wisconsin Center for Nanobiosystems, School of Pharmacy, University of Wisconsin-Madison, Wisconsin-53705, USA.
| | - Fátima Franco De Silva
- Department of Food Engineering, Tecnologico de Monterrey, Monterrey, Nuevo Leon-64849, Mexico
| | - Amin Orash Mahmoudsalehi
- Tecnologico de Monterrey, Escuela de Ingeniería y Ciencias, Av. Eugenio Garza Sada 2501 Sur, Monterrey, Nuevo Leon-64849, Mexico
| |
Collapse
|
6
|
Feng S, Li Y, Tan Z, Shen S. Current landscape of metal-organic framework-mediated nucleic acid delivery and therapeutics. Int J Pharm 2025; 672:125295. [PMID: 39914507 DOI: 10.1016/j.ijpharm.2025.125295] [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: 11/12/2024] [Revised: 01/16/2025] [Accepted: 01/27/2025] [Indexed: 02/13/2025]
Abstract
Nucleic acid drugs utilize DNA or RNA molecules to modulate abnormal gene expression or protein translation in cells, enabling precise treatment for specific conditions. In recent years, nucleic acid drugs have demonstrated tremendous potential in vaccine development and treating genetic disorders. Currently, the primary carriers for clinically approved nucleic acid therapies include lipid nanoparticles and viral vectors. Beyond that, metal-organic frameworks (MOFs) are highly ordered, porous nanomaterials formed through the self-assembly of metal ions and organic ligands via coordination bonds. Their porosity structure offers great loading efficiency, stability, tunability, and biocompatibility, making them an attractive option for nucleic acid delivery. Given the research on MOFs as nucleic acid carriers has garnered significant attention in recent years, this review provides an overview of the therapeutic strategies and advancements in MOF-mediated nucleic acid delivery. The unique properties of various MOF carriers are introduced, and different approaches for nucleic acid loading are parallelly compared. Moreover, a systematic classification based on the type of nucleic acid cargo loaded in MOFs and corresponding applications is thoroughly described. This summary outlines the unique mechanisms through MOFs enhance nucleic acid delivery and emphasizes their substantial impact on therapeutic efficacy. In addition, the utilization of MOF-mediated nucleic acid treatment in combination with other therapies against malignant tumors is discussed in particular. Finally, an outlook on the challenges and potential opportunities of this technology in future translational production and clinical implementation is presented and explored.
Collapse
Affiliation(s)
- Shiwei Feng
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Zhongshan 528400, China
| | - Yan Li
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Zhongshan 528400, China
| | - Zheng Tan
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Zhongshan 528400, China.
| | - Shiyang Shen
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Zhongshan 528400, China; Pharmaceutical Preparation Research Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China.
| |
Collapse
|
7
|
Li J, Mao X, Zhao T, Fang W, Jin Y, Liu M, Fan C, Tian Y. Tetrahedral DNA Framework-Based Spherical Nucleic Acids for Efficient siRNA Delivery. Angew Chem Int Ed Engl 2025; 64:e202416988. [PMID: 39497620 DOI: 10.1002/anie.202416988] [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: 09/04/2024] [Indexed: 11/20/2024]
Abstract
Spherical nucleic acids (SNAs) hold substantial therapeutic potential for the delivery of small interfering RNAs (siRNAs). Nevertheless, their potential remains largely untapped due to the challenges of cytosolic delivery. Inspired by the dynamic, spiky architecture of coronavirus, an interface engineering approach based on a tetrahedral DNA framework (tDF) is demonstrated for the development of coronavirus-mimicking SNAs. By exploiting their robustness and precise construction, tDFs are evenly arranged on the surface of core nanoparticles (NPs) with flexible conformations, generating a dynamic, spiky architecture. This spiky architecture in tetrahedral DNA framework-based SNAs (tDF-SNAs) substantially improve siRNAs duplex efficiency from 20 % to 95 %. Meanwhile, tDF-SNAs changed the endocytosis pathway to clathrin-independent cellular engulfment pathway and enhanced the cellular uptake efficiency. Due to these advances, the delivery efficiency of siRNA molecules by tDF-SNAs is 1-2 orders of magnitude higher than that of SNAs, resulting in a 2-fold increase in gene silencing efficacy. These results show promise in the development of bioinspired siRNAs delivery systems for intracellular applications.
Collapse
Affiliation(s)
- Jie Li
- School of Chemistry and Molecular Engineering, Shanghai Key Laboratory of Green Chemistry and Chemical Processes, East China Normal University, Shanghai, 200241, China
| | - Xiuhai Mao
- Shanghai Key Laboratory for Nucleic Acids Chemistry and Nanomedicine, Institute of Molecular Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
| | - Tiantian Zhao
- School of Chemistry and Molecular Engineering, Shanghai Key Laboratory of Green Chemistry and Chemical Processes, East China Normal University, Shanghai, 200241, China
| | - Weina Fang
- School of Chemistry and Molecular Engineering, Shanghai Key Laboratory of Green Chemistry and Chemical Processes, East China Normal University, Shanghai, 200241, China
| | - Yangyang Jin
- Shanghai Key Laboratory for Nucleic Acids Chemistry and Nanomedicine, Institute of Molecular Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
| | - Mengmeng Liu
- School of Chemistry and Molecular Engineering, Shanghai Key Laboratory of Green Chemistry and Chemical Processes, East China Normal University, Shanghai, 200241, China
| | - Chunhai Fan
- Frontiers Science Center for Transformative Molecules, School of Chemistry and Chemical Engineering, Institute of Translational Medicine, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Yang Tian
- School of Chemistry and Molecular Engineering, Shanghai Key Laboratory of Green Chemistry and Chemical Processes, East China Normal University, Shanghai, 200241, China
| |
Collapse
|
8
|
Bian X, Yu X, Lu S, Jia L, Li P, Yin J, Tan S. Chitosan-based nanoarchitectures for siRNA delivery in cancer therapy: A review of pre-clinical and clinical importance. Int J Biol Macromol 2025; 284:137708. [PMID: 39571854 DOI: 10.1016/j.ijbiomac.2024.137708] [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: 10/12/2024] [Revised: 11/01/2024] [Accepted: 11/13/2024] [Indexed: 11/30/2024]
Abstract
The gene therapy has been developed into a new cancer treatment option. Now that we know which molecular components contribute to carcinogenesis, we may use gene therapy to target particular signalling pathways in cancer treatment. Problems with gene therapy include genetic tool degradation in blood, off-targeting features, and inadequate tumor site accumulation; new delivery mechanisms are needed to address these issues. A polysaccharide made from chitin, chitosan has found extensive use in the creation of nanoparticles. The delivery of genes in the treatment of illnesses, particularly cancer, has made use of nanostructures modified with chitosan. Topics covered in this review center on cancer treatment using chitosan-based polymers for siRNA delivery. This study aims to assess the potential of chitosan nanoparticles for the simultaneous administration of siRNA and anti-cancer medications. In cancer treatment, these nanoparticles can transport phytochemicals or chemotherapeutics together with siRNA. In addition, chitosan nanoparticles loaded with siRNA can inhibit the growth and spread of human malignancies by delivering siRNA that targets particular genes. Chitosan nanoparticles loaded with siRNA can heighten the responsiveness of cancer cells. Future therapeutic applications of chitosan nanoparticles may open the path for cancer treatment, thanks to their biocompatibility and biosafety.
Collapse
Affiliation(s)
- Xiaobo Bian
- Department of Oncology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Xiaopeng Yu
- Department of Oncology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Shiyang Lu
- Department of Urology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Linan Jia
- Department of Urology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Ping Li
- Center of Reproductive Medicine, Shengjing Hospital of China Medical University, Shenyang, China.
| | - Jianqiao Yin
- Department of Oncology, Shengjing Hospital of China Medical University, Shenyang, China.
| | - Shutao Tan
- Department of Urology, Shengjing Hospital of China Medical University, Shenyang, China.
| |
Collapse
|
9
|
Ji L, Huang J, Yu L, Jin H, Hu X, Sun Y, Yin F, Cai Y. Recent advances in nanoagents delivery system-based phototherapy for osteosarcoma treatment. Int J Pharm 2024; 665:124633. [PMID: 39187032 DOI: 10.1016/j.ijpharm.2024.124633] [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: 04/02/2024] [Revised: 08/09/2024] [Accepted: 08/22/2024] [Indexed: 08/28/2024]
Abstract
Osteosarcoma (OS) is a prevalent and highly malignant bone tumor, characterized by its aggressive nature, invasiveness, and rapid progression, contributing to a high mortality rate, particularly among adolescents. Traditional treatment modalities, including surgical resection, radiotherapy, and chemotherapy, face significant challenges, especially in addressing chemotherapy resistance and managing postoperative recurrence and metastasis. Phototherapy (PT), encompassing photodynamic therapy (PDT) and photothermal therapy (PTT), offers unique advantages such as low toxicity, minimal drug resistance, selective destruction, and temporal control, making it a promising approach for the clinical treatment of various malignant tumors. Constructing multifunctional delivery systems presents an opportunity to effectively combine tumor PDT, PTT, and chemotherapy, creating a synergistic anti-tumor effect. This review aims to consolidate the progress in the application of novel delivery system-mediated phototherapy in osteosarcoma. By summarizing advancements in this field, the objective is to propose a rational combination therapy involving targeted delivery systems and phototherapy for tumors, thereby expanding treatment options and enhancing the prognosis for osteosarcoma patients. In conclusion, the integration of innovative delivery systems with phototherapy represents a promising avenue in osteosarcoma treatment, offering a comprehensive approach to overcome challenges associated with conventional treatments and improve patient outcomes.
Collapse
Affiliation(s)
- Lichen Ji
- Zhejiang Chinese Medical University, Hangzhou 310053, China; Center for Rehabilitation Medicine Rehabilitation & Sports Medicine Research Institute of Zhejiang Province Department of Rehabilitation Medicine, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou 310014, China; Department of Joint Surgery, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200092, China
| | - Jiaqing Huang
- Zhejiang Chinese Medical University, Hangzhou 310053, China; Center for Rehabilitation Medicine Rehabilitation & Sports Medicine Research Institute of Zhejiang Province Department of Rehabilitation Medicine, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou 310014, China; Department of Hematology, Hangzhou First People's Hospital, Hangzhou 310003, China
| | - Liting Yu
- Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Huihui Jin
- Zhejiang Chinese Medical University, Hangzhou 310053, China; Center for Rehabilitation Medicine Rehabilitation & Sports Medicine Research Institute of Zhejiang Province Department of Rehabilitation Medicine, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou 310014, China
| | - Xuanhan Hu
- Zhejiang Chinese Medical University, Hangzhou 310053, China; Center for Rehabilitation Medicine Rehabilitation & Sports Medicine Research Institute of Zhejiang Province Department of Rehabilitation Medicine, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou 310014, China
| | - Yuan Sun
- College of Chemistry Engineering, Zhejiang University of Technology, Hangzhou, 310014, China.
| | - Feng Yin
- Department of Joint Surgery, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200092, China.
| | - Yu Cai
- Center for Rehabilitation Medicine Rehabilitation & Sports Medicine Research Institute of Zhejiang Province Department of Rehabilitation Medicine, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou 310014, China.
| |
Collapse
|
10
|
Wang Y, Wang C, Lu Y. Spleen Targeting Nucleic Acid Delivery Vector Based on Metal-Organic Frameworks. ACS APPLIED MATERIALS & INTERFACES 2024; 16:56826-56836. [PMID: 39390629 DOI: 10.1021/acsami.4c13519] [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: 10/12/2024]
Abstract
Nucleic acids have attracted increasing attention as drugs due to their fascinating advantages, such as long-term efficacy and ease of preparation compared to proteins. The nucleic acid therapy relies heavily on delivery vectors, which can prevent the degradation of nucleic acids while assisting them in cellular internalization. However, commonly used nonviral vector liposomes easily accumulate in the liver, which can limit their application in extrahepatic diseases. Herein, a potential spleen targeting vector for nucleic acids is developed based on the metal-organic frameworks. The plasmids are encapsulated inside the nanoscale zeolitic imidazolate framework (ZIF) via coprecipitation. The co-encapsulation of the cationic polymer poly(ether imide) (PEI) and the stabilizer polyvinylpyrrolidone (PVP) can significantly improve particle dispersion and stability. The prepared nanoparticles allow efficient transfection in vitro, mainly through clathrin-mediated and caveolae-mediated endocytosis. The biodistribution in mice shows that 46% of the nanoparticles accumulate in the spleen, which is much higher than that of the liposomes. The vector can successfully deliver plasmids to extrahepatic organs for protein synthesis and even induce an immune response. The elaborate ZIF-based nanoparticle may offer a new route for extrahepatic, especially spleen targeting delivery for the nucleic acids.
Collapse
Affiliation(s)
- Yiming Wang
- Department of Chemical Engineering, Tsinghua University, Beijing 100084, China
- Key Laboratory of Industrial Biocatalysis, Ministry of Education, Tsinghua University, Beijing 100084, China
| | - Chen Wang
- Department of Chemical Engineering, Tsinghua University, Beijing 100084, China
- Key Laboratory of Industrial Biocatalysis, Ministry of Education, Tsinghua University, Beijing 100084, China
| | - Yuan Lu
- Department of Chemical Engineering, Tsinghua University, Beijing 100084, China
- Key Laboratory of Industrial Biocatalysis, Ministry of Education, Tsinghua University, Beijing 100084, China
| |
Collapse
|
11
|
Lawson JL, Sekar RP, Wright ARE, Wheeler G, Yanes J, Estridge J, Johansen CG, Farnsworth NL, Kumar P, Tay JW, Kumar R. The Spatial Distribution of Lipophilic Cations in Gradient Copolymers Regulates Polymer-pDNA Complexation, Polyplex Aggregation, and Intracellular pDNA Delivery. Biomacromolecules 2024; 25:6855-6870. [PMID: 39318335 PMCID: PMC12020213 DOI: 10.1021/acs.biomac.4c01101] [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] [Indexed: 09/26/2024]
Abstract
Here, we demonstrate that the spatial distribution of lipophilic cations governs the complexation pathways, serum stability, and biological performance of polymer-pDNA complexes (polyplexes). Previous research focused on block/statistical copolymers, whereas gradient copolymers, where the density of lipophilic cations diminishes (gradually or steeply) along polymer backbones, remain underexplored. We engineered gradient copolymers that combine the polyplex colloidal stability of block copolymers with the transfection efficiency of statistical copolymers. We synthesized length- and compositionally equivalent gradient copolymers (G1-G3) along with statistical (S) and block (B) copolymers of 2-(diisopropylamino)ethyl methacrylate and 2-hydroxyethyl methacrylate. We mapped how polymer microstructure governs pDNA loading per polyplex, pDNA conformational changes, and polymer-pDNA binding thermodynamics via static light scattering, circular dichroism spectroscopy, and isothermal titration calorimetry, respectively. While gradient steepness is a powerful design handle to improve polyplex physical properties, augment pDNA delivery capacity, and attenuate polycation-triggered hemolysis, microstructural contrasts did not elicit differences in complement activation.
Collapse
Affiliation(s)
- Jessica L Lawson
- Materials Science, Colorado School of Mines, Golden, Colorado 80401, United States
| | - Ram Prasad Sekar
- Chemical and Biological Engineering, Colorado School of Mines, Golden, Colorado 80401, United States
| | - Aryelle R E Wright
- Quantitative Biosciences and Engineering, Colorado School of Mines, Golden, Colorado 80401, United States
| | - Grant Wheeler
- Quantitative Biosciences and Engineering, Colorado School of Mines, Golden, Colorado 80401, United States
| | - Jillian Yanes
- Quantitative Biosciences and Engineering, Colorado School of Mines, Golden, Colorado 80401, United States
| | - Jordan Estridge
- Chemical and Biological Engineering, Colorado School of Mines, Golden, Colorado 80401, United States
| | - Chelsea G Johansen
- Chemical and Biological Engineering, Colorado School of Mines, Golden, Colorado 80401, United States
| | - Nikki L Farnsworth
- Chemical and Biological Engineering, Colorado School of Mines, Golden, Colorado 80401, United States
- Quantitative Biosciences and Engineering, Colorado School of Mines, Golden, Colorado 80401, United States
| | - Praveen Kumar
- Shared Instrumentation Facility, Colorado School of Mines, Golden, Colorado 80401, United States
| | - Jian Wei Tay
- Biofrontiers Institute, University of Colorado, Boulder, Colorado 80309, United States
| | - Ramya Kumar
- Materials Science, Colorado School of Mines, Golden, Colorado 80401, United States
- Chemical and Biological Engineering, Colorado School of Mines, Golden, Colorado 80401, United States
- Quantitative Biosciences and Engineering, Colorado School of Mines, Golden, Colorado 80401, United States
| |
Collapse
|
12
|
Guan JX, Wang YL, Wang JL. How Advanced are Nanocarriers for Effective Subretinal Injection? Int J Nanomedicine 2024; 19:9273-9289. [PMID: 39282576 PMCID: PMC11401526 DOI: 10.2147/ijn.s479327] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Accepted: 08/28/2024] [Indexed: 09/19/2024] Open
Abstract
Subretinal injection (SR injection) is a commonly used method of ocular drug delivery and has been mainly applied for the treatment of neovascular age-associated macular degeneration (nAMD) and sub-macular hemorrhage (SMH) caused by nAMD, as well as various types of hereditary retinopathies (IRD) such as Stargardt's disease (STGD), retinitis pigmentosa (RP), and a series of fundus diseases such as Leber's congenital dark haze (LCA), choroidal defects, etc. The commonly used carriers of SR injection are mainly divided into viral and non-viral vectors. Leber's congenital amaurosis (LCA), choroidal agenesis, and a series of other fundus diseases are also commonly treated using SR injection. The commonly used vectors for SR injection are divided into two categories: viral vectors and non-viral vectors. Viral vectors are a traditional class of SR injection drug carriers that have been extensively studied in clinical treatment, but they still have many limitations that cannot be ignored, such as poor reproduction efficiency, small loading genes, and triggering of immune reactions. With the rapid development of nanotechnology in the treatment of ocular diseases, nanovectors have become a research hotspot in the field of non-viral vectors. Nanocarriers have numerous attractive properties such as low immunogenicity, robust loading capacity, stable structure, and easy modification. These valuable features imply greater safety, improved therapeutic efficacy, longer duration, and more flexible indications. In recent years, there has been a growing interest in nanocarriers, which has led to significant advancements in the treatment of ocular diseases. Nanocarriers have not only successfully addressed clinical problems that viral vectors have failed to overcome but have also introduced new therapeutic possibilities for certain classical disease types. Nanocarriers offer undeniable advantages over viral vectors. This review discusses the advantages of subretinal (SR) injection, the current status of research, and the research hotspots of gene therapy with viral vectors. It focuses on the latest progress of nanocarriers in SR injection and enumerates the limitations and future perspectives of nanocarriers in the treatment of fundus lesions. Furthermore, this review also covers the research progress of nanocarriers in the field of subretinal injection and highlights the value of nanocarrier-mediated SR injection in the treatment of fundus disorders. Overall, it provides a theoretical basis for the application of nanocarriers in SR injection.
Collapse
Affiliation(s)
- Jia-Xin Guan
- Department of Ophthalmology, Beijing Friendship Hospital, Capital Medical University, Beijing, People's Republic of China
- Institute of Ophthalmology, Capital Medical University, Beijing, People's Republic of China
| | - Yan-Ling Wang
- Department of Ophthalmology, Beijing Friendship Hospital, Capital Medical University, Beijing, People's Republic of China
- Institute of Ophthalmology, Capital Medical University, Beijing, People's Republic of China
| | - Jia-Lin Wang
- Department of Ophthalmology, Beijing Friendship Hospital, Capital Medical University, Beijing, People's Republic of China
- Institute of Ophthalmology, Capital Medical University, Beijing, People's Republic of China
| |
Collapse
|
13
|
Zhang Y, Huang C, Xiong R. Advanced materials for intracellular delivery of plant cells: Strategies, mechanisms and applications. MATERIALS SCIENCE AND ENGINEERING: R: REPORTS 2024; 160:100821. [DOI: 10.1016/j.mser.2024.100821] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
|
14
|
Godakhindi V, Tarannum M, Dam SK, Vivero-Escoto JL. Mesoporous Silica Nanoparticles as an Ideal Platform for Cancer Immunotherapy: Recent Advances and Future Directions. Adv Healthc Mater 2024; 13:e2400323. [PMID: 38653190 PMCID: PMC11305940 DOI: 10.1002/adhm.202400323] [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: 01/27/2024] [Revised: 04/10/2024] [Indexed: 04/25/2024]
Abstract
Cancer immunotherapy recently transforms the traditional approaches against various cancer malignancies. Immunotherapy includes systemic and local treatments to enhance immune responses against cancer and involves strategies such as immune checkpoints, cancer vaccines, immune modulatory agents, mimetic antigen-presenting cells, and adoptive cell therapy. Despite promising results, these approaches still suffer from several limitations including lack of precise delivery of immune-modulatory agents to the target cells and off-target toxicity, among others, that can be overcome using nanotechnology. Mesoporous silica nanoparticles (MSNs) are investigated to improve various aspects of cancer immunotherapy attributed to the advantageous structural features of this nanomaterial. MSNs can be engineered to alter their properties such as size, shape, porosity, surface functionality, and adjuvanticity. This review explores the immunological properties of MSNs and the use of MSNs as delivery vehicles for immune-adjuvants, vaccines, and mimetic antigen-presenting cells (APCs). The review also details the current strategies to remodel the tumor microenvironment to positively reciprocate toward the anti-tumor immune cells and the use of MSNs for immunotherapy in combination with other anti-tumor therapies including photodynamic/thermal therapies to enhance the therapeutic effect against cancer. Last, the present demands and future scenarios for the use of MSNs for cancer immunotherapy are discussed.
Collapse
Affiliation(s)
- Varsha Godakhindi
- Department of Chemistry, University of North Carolina at Charlotte, Charlotte, NC, 28223, USA
- Nanoscale Science Program, The University of North Carolina at Charlotte, Charlotte, NC, 28223, USA
| | - Mubin Tarannum
- Division of Medical Oncology, Dana Farber Cancer Institute, Harvard Medical School, Boston, MA, 02215, USA
| | - Sudip Kumar Dam
- Department of Chemistry, University of North Carolina at Charlotte, Charlotte, NC, 28223, USA
- Nanoscale Science Program, The University of North Carolina at Charlotte, Charlotte, NC, 28223, USA
| | - Juan L Vivero-Escoto
- Department of Chemistry, University of North Carolina at Charlotte, Charlotte, NC, 28223, USA
- Nanoscale Science Program, The University of North Carolina at Charlotte, Charlotte, NC, 28223, USA
| |
Collapse
|
15
|
Liu J, Han Y, Zhao M, Wang L, Hu H, Chen D. Unlocking the power of immunotherapy: Combinatorial delivery of plasmid IL-15 and gemcitabine to synergistically remodeling the tumor microenvironment. Int J Pharm 2024; 655:124027. [PMID: 38554742 DOI: 10.1016/j.ijpharm.2024.124027] [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: 11/30/2023] [Revised: 03/03/2024] [Accepted: 03/19/2024] [Indexed: 04/02/2024]
Abstract
Cancer immunotherapy has emerged as a promising clinical treatment strategy in recent years. Unfortunately, the satisfactory antitumor therapeutic efficacy of immunotherapy is limited by intricate immunosuppressive tumor microenvironment (ITM). To remodel the ITM and alleviate the immune evasion, we constructed FA-PEG-modified liposomes to deliver plasmid IL-15 (pIL-15) and gemcitabine (GEM) (FPCL@pIL-15 + FPGL), respectively. The FPCL@pIL-15 (150 nm) and FPGL (120 nm) exhibited symmetrically spherical structures as well as desirable penetration and accumulation on tumor tissue depending on folic acid (FA) specialized targeting function. The transfected expression of IL-15 efficiently fosters the proliferation and co-activation of Natural killer (NK) cells and CD8+T cells through binding to IL-15R. FPGL upregulated the expression of Natural killer group 2 member D ligands (NKG2DLs) and reinforced recognition by NK cells to alleviate the immune evasion, and simultaneously promoted activation of CD8+T cells through immunogenic cell death (ICD) effects. More importantly, the combinatorial administration achieved intended anti-tumor efficacy in the subcutaneous 4T1 tumor model. In essence, we demonstrated that combining FPCL@pIL-15 with FPGL synergistically stimulates and mobilizes the immune system to reverse the ITM and trigger an anti-tumor immune response, indicating a tremendous potential for application in immunotherapy.
Collapse
Affiliation(s)
- Jingwen Liu
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, No. 103, Wenhua Road, Shenyang, 110016, PR China
| | - Yanyan Han
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, No. 103, Wenhua Road, Shenyang, 110016, PR China
| | - Ming Zhao
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, No. 103, Wenhua Road, Shenyang, 110016, PR China
| | - Leyuan Wang
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, No. 103, Wenhua Road, Shenyang, 110016, PR China
| | - Haiyang Hu
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, No. 103, Wenhua Road, Shenyang, 110016, PR China.
| | - Dawei Chen
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, No. 103, Wenhua Road, Shenyang, 110016, PR China.
| |
Collapse
|
16
|
Shen X, Zhang Y, Wang D, Huang Y, Song Y, Wang S. Mediator Monomer Regulated Emulsion Interfacial Polymerization to Synthesize Nanofractal Magnetic Particles for Nucleic Acid Separation. SMALL METHODS 2024; 8:e2300531. [PMID: 37491768 DOI: 10.1002/smtd.202300531] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/22/2023] [Revised: 07/14/2023] [Indexed: 07/27/2023]
Abstract
Polymer-based magnetic particles have been widely used for the separation of biological samples including nucleic acids, proteins, virus, and cells. Existing magnetic particles are almost prepared by coating polymers on magnetic nanoparticles (NPs). However, this strategy usually encounters the problem of poor magnetic NPs loading capacity. Here, a series of nanofractal magnetic particles (nanoFMPs) synthesized by a strategy of mediator monomer regulated emulsion interfacial polymerization is presented, which allows effective magnetic NPs loading and show efficient nucleic acid separation performance. The mediator monomers facilitate the dispersion of magnetic NPs in internal phase to achieve higher loading, and the hydrophilic monomers use electrostatic interactions to form surface nanofractal structures with functional groups. Compared with magnetic particles without nanofractal structure, nanoFMPs exhibit a higher nucleic acid extraction capability. This strategy offers an effective and versatile way for the synthesis of nanoFMPs toward efficient separation in various fields from clinical diagnosis to food safety and environmental monitoring.
Collapse
Affiliation(s)
- Xinyi Shen
- CAS Key Laboratory of Bio-inspired Materials and Interfacial Science, Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Beijing, 100190, P. R. China
- University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Yue Zhang
- CAS Key Laboratory of Bio-inspired Materials and Interfacial Science, Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Beijing, 100190, P. R. China
- University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Duanda Wang
- CAS Key Laboratory of Bio-inspired Materials and Interfacial Science, Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Beijing, 100190, P. R. China
- University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Yanling Huang
- CAS Key Laboratory of Bio-inspired Materials and Interfacial Science, Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Beijing, 100190, P. R. China
| | - Yongyang Song
- CAS Key Laboratory of Bio-inspired Materials and Interfacial Science, Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Beijing, 100190, P. R. China
- University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
- Binzhou Institute of Technology, Weiqiao-UCAS Science and Technology Park, Binzhou City, Shandong Province, 256606, P. R. China
| | - Shutao Wang
- CAS Key Laboratory of Bio-inspired Materials and Interfacial Science, Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Beijing, 100190, P. R. China
- University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
- Binzhou Institute of Technology, Weiqiao-UCAS Science and Technology Park, Binzhou City, Shandong Province, 256606, P. R. China
| |
Collapse
|
17
|
Zhao Y, Xu C, Liu Q, Lei X, Deng L, Wang F, Yang J. pH-responsive interface conversion efficient oral drug delivery platform for alleviating inflammatory bowel disease. Front Chem 2024; 12:1365880. [PMID: 38532806 PMCID: PMC10963395 DOI: 10.3389/fchem.2024.1365880] [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: 01/05/2024] [Accepted: 03/04/2024] [Indexed: 03/28/2024] Open
Abstract
A key challenge for the effective treatment of intestinal diseases, including inflammatory bowel disease (IBD), is to develop an oral drug delivery system that can resist gastric acid erosion and efficiently release drugs after rapid entry into the intestine. In the present work, we developed oral composite nanoparticles (MSZ@PRHS) consisting of a rough mesoporous silica (RHS) loaded with Mesalazine (MSZ) and a CAP polymer membrane for targeted relief of inflammation in colitis. At the pH values of the simulated stomach and small intestine, the release rate of MSZ from MSZ@PRHS was low, while at the pH values of the simulated colon, the release rate of MSZ was high. In dextran sulfate sodium salt (DSS)-induced acute colitis mouse model, compared with oral administration of the drug Mesalazine in the equivalent solution form, oral administration of PRHS loaded with drug-loaded nanoparticles can significantly alleviate the symptoms of inflammatory bowel disease, and improve the therapeutic effect. We propose that the intestinal microenvironment provides an interface for nanocomposites switch and a promising drug delivery platform for the management and treatment of many intestinal diseases, where controlled drug release and prolonged residence time are required.
Collapse
Affiliation(s)
- Yingying Zhao
- Department of Gastroenterology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong, China
| | - Changqing Xu
- Department of Gastroenterology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong, China
| | - Qing Liu
- Department of Clinical Laboratory, Shanghai Gongli Hospital, The Second Military Medical University, Shanghai, China
| | - Xiaofei Lei
- Department of Gastroenterology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong, China
| | - Li Deng
- Department of Gastroenterology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong, China
| | - Fengyan Wang
- Department of Gastroenterology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong, China
| | - Jing Yang
- Department of Gastroenterology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong, China
| |
Collapse
|
18
|
Gholap AD, Kapare HS, Pagar S, Kamandar P, Bhowmik D, Vishwakarma N, Raikwar S, Garkal A, Mehta TA, Rojekar S, Hatvate N, Mohanto S. Exploring modified chitosan-based gene delivery technologies for therapeutic advancements. Int J Biol Macromol 2024; 260:129581. [PMID: 38266848 DOI: 10.1016/j.ijbiomac.2024.129581] [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: 12/26/2023] [Accepted: 01/06/2024] [Indexed: 01/26/2024]
Abstract
One of the critical steps in gene therapy is the successful delivery of the genes. Immunogenicity and toxicity are major issues for viral gene delivery systems. Thus, non-viral vectors are explored. A cationic polysaccharide like chitosan could be used as a nonviral gene delivery vector owing to its significant interaction with negatively charged nucleic acid and biomembrane, providing effective cellular uptake. However, the native chitosan has issues of targetability, unpacking ability, and solubility along with poor buffer capability, hence requiring modifications for effective use in gene delivery. Modified chitosan has shown that the "proton sponge effect" involved in buffering the endosomal pH results in osmotic swelling owing to the accumulation of a greater amount of proton and chloride along with water. The major challenges include limited exploration of chitosan as a gene carrier, the availability of high-purity chitosan for toxicity reduction, and its immunogenicity. The genetic drugs are in their infancy phase and require further exploration for effective delivery of nucleic acid molecules as FDA-approved marketed formulations soon.
Collapse
Affiliation(s)
- Amol D Gholap
- Department of Pharmaceutics, St. John Institute of Pharmacy and Research, Palghar 401404, Maharashtra, India
| | - Harshad S Kapare
- Department of Pharmaceutics, Dr. D. Y. Patil Institute of Pharmaceutical Sciences and Research, Pune 411018, Maharashtra, India
| | - Sakshi Pagar
- Department of Pharmaceutical Sciences and Technology, Institute of Chemical Technology, Mumbai 400019, India
| | - Pallavi Kamandar
- Institute of Chemical Technology, Mumbai, Marathwada Campus, Jalna 431203, India
| | - Deblina Bhowmik
- Institute of Chemical Technology, Mumbai, Marathwada Campus, Jalna 431203, India
| | - Nikhar Vishwakarma
- Department of Pharmacy, Gyan Ganga Institute of Technology and Sciences, Jabalpur 482003, Madhya Pradesh, India
| | - Sarjana Raikwar
- Department of Pharmaceutical Sciences, Dr. Harisingh Gour Central University, Sagar 470003, Madhya Pradesh, India
| | - Atul Garkal
- Department of Pharmaceutics, Institute of Pharmacy, Nirma University, Ahmedabad 382481, Gujrat, India
| | - Tejal A Mehta
- Department of Pharmaceutics, Institute of Pharmacy, Nirma University, Ahmedabad 382481, Gujrat, India
| | - Satish Rojekar
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA.
| | - Navnath Hatvate
- Institute of Chemical Technology, Mumbai, Marathwada Campus, Jalna 431203, India.
| | - Sourav Mohanto
- Department of Pharmaceutics, Yenepoya Pharmacy College & Research Centre, Yenepoya (Deemed to be University), Mangaluru, Karnataka 575018, India
| |
Collapse
|
19
|
Zhang Y, Yu Y, Yang Y, Wang Y, Yu C. Engineered Silica Nanoparticles for Nucleic Acid Delivery. SMALL METHODS 2024; 8:e2300812. [PMID: 37906035 DOI: 10.1002/smtd.202300812] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Revised: 09/14/2023] [Indexed: 11/02/2023]
Abstract
The development of nucleic acid-based drugs holds great promise for therapeutic applications, but their effective delivery into cells is hindered by poor cellular membrane permeability and inherent instability. To overcome these challenges, delivery vehicles are required to protect and deliver nucleic acids efficiently. Silica nanoparticles (SiNPs) have emerged as promising nanovectors and recently bioregulators for gene delivery due to their unique advantages. In this review, a summary of recent advancements in the design of SiNPs for nucleic acid delivery and their applications is provided, mainly according to the specific type of nucleic acids. First, the structural characteristics and working mechanisms of various types of nucleic acids are introduced and classified according to their functions. Subsequently, for each nucleic acid type, the use of SiNPs for enhancing delivery performance and their biomedical applications are summarized. The tailored design of SiNPs for selected type of nucleic acid delivery will be highlighted considering the characteristics of nucleic acids. Lastly, the limitations in current research and personal perspectives on future directions in this field are presented. It is expected this opportune review will provide insights into a burgeoning research area for the development of next-generation SiNP-based nucleic acid delivery systems.
Collapse
Affiliation(s)
- Yue Zhang
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, Queensland, 4072, Australia
| | - Yingjie Yu
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, Queensland, 4072, Australia
| | - Yannan Yang
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, Queensland, 4072, Australia
- Shanghai Frontiers Science Research Base of Intelligent Optoelectronics and Perception, Institute of Optoelectronics, Fudan University, Shanghai, 200433, P. R. China
| | - Yue Wang
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, Queensland, 4072, Australia
| | - 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
| |
Collapse
|
20
|
Xu J, Zhong X, Fan M, Xu Y, Xu Y, Wang S, Luo Z, Huang Y. Enhancing intracellular mRNA precise imaging-guided photothermal therapy with a nucleic acid-based polydopamine nanoprobe. Anal Bioanal Chem 2024; 416:849-859. [PMID: 38006441 DOI: 10.1007/s00216-023-05062-2] [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: 10/29/2023] [Revised: 11/15/2023] [Accepted: 11/17/2023] [Indexed: 11/27/2023]
Abstract
Despite significant advancements in cancer research, real-time monitoring and effective treatment of cancer through non-invasive techniques remain a challenge. Herein, a novel polydopamine (PDA) nucleic acid nanoprobe has been developed for imaging signal amplification of intracellular mRNA and precise photothermal therapy guidance in cancer cells. The PDA nucleic acid nanoprobe (PDA@DNA) is constructed by assembling an aptamer hairpin (H1) labeled with the Cy5 fluorophore and another nucleic acid recognition hairpin (H2) onto PDA nanoparticles (PDA NPs), which have exceptionally high fluorescence quenching ability and excellent photothermal conversion properties. The nanoprobe could facilitate cellular uptake of DNA molecules and their protection from nuclease degradation. Upon recognition and binding to the intracellular mRNA target, a catalytic hairpin assembly (CHA) reaction occurs. The stem of H1 unfolds upon binding, allowing the exposed H1 to hybridize with H2, forming a flat and sturdy DNA double-stranded structure that detaches from the surface of PDA NPs. At the same time, the target mRNA is displaced and engages in a new cyclic reaction, resulting in the recovery and significant amplification of Cy5 fluorescence. Using thymidine kinase1 (TK1) mRNA as a model mRNA, this nanoprobe enables the analysis of TK1 mRNA with a detection limit of 9.34 pM, which is at least two orders of magnitude lower than that of a non-amplifying imaging nucleic acid probe. Moreover, with its outstanding performance for in vitro detection, this nanoprobe excels in precisely imaging tumor cells. Through live-cell TK1 mRNA imaging, it can accurately distinguish between tumor cells and normal cells. Furthermore, when exposed to 808-nm laser irradiation, the nanoprobe fully harnesses exceptional photothermal conversion properties of PDA NPs. This results in a localized temperature increase within tumor cells, which ultimately triggers apoptosis in these tumor cells. The integration of PDA@DNA presents innovative prospects for tumor diagnosis and image-guided tumor therapy, offering the potential for high-precision diagnosis and treatment of tumors.
Collapse
Affiliation(s)
- Jiayao Xu
- Guangxi Key Lab of Agricultural Resources Chemistry and Biotechnology, College of Chemistry and Food Science, Yulin Normal University, Yulin, 537000, People's Republic of China
| | - Xiaohong Zhong
- Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin, 541004, People's Republic of China
| | - Mingzhu Fan
- Guangxi Key Lab of Agricultural Resources Chemistry and Biotechnology, College of Chemistry and Food Science, Yulin Normal University, Yulin, 537000, People's Republic of China
| | - Yang Xu
- Guangxi Key Lab of Agricultural Resources Chemistry and Biotechnology, College of Chemistry and Food Science, Yulin Normal University, Yulin, 537000, People's Republic of China
| | - Yiqi Xu
- Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin, 541004, People's Republic of China
| | - Shulong Wang
- Guangxi Key Lab of Agricultural Resources Chemistry and Biotechnology, College of Chemistry and Food Science, Yulin Normal University, Yulin, 537000, People's Republic of China.
| | - Zhihui Luo
- Guangxi Key Lab of Agricultural Resources Chemistry and Biotechnology, College of Chemistry and Food Science, Yulin Normal University, Yulin, 537000, People's Republic of China.
| | - Yong Huang
- Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin, 541004, People's Republic of China.
| |
Collapse
|
21
|
Salehi Abar E, Vandghanooni S, Torab A, Jaymand M, Eskandani M. A comprehensive review on nanocomposite biomaterials based on gelatin for bone tissue engineering. Int J Biol Macromol 2024; 254:127556. [PMID: 37884249 DOI: 10.1016/j.ijbiomac.2023.127556] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2023] [Revised: 10/09/2023] [Accepted: 10/18/2023] [Indexed: 10/28/2023]
Abstract
The creation of a suitable scaffold is a crucial step in the process of bone tissue engineering (BTE). The scaffold, acting as an artificial extracellular matrix, plays a significant role in determining the fate of cells by affecting their proliferation and differentiation in BTE. Therefore, careful consideration should be given to the fabrication approach and materials used for scaffold preparation. Natural polypeptides such as gelatin and collagen have been widely used for this purpose. The unique properties of nanoparticles, which vary depending on their size, charge, and physicochemical properties, have demonstrated potential in solving various challenges encountered in BTE. Therefore, nanocomposite biomaterials consisting of polymers and nanoparticles have been extensively used for BTE. Gelatin has also been utilized in combination with other nanomaterials to apply for this purpose. Composites of gelatin with various types of nanoparticles are particularly promising for creating scaffolds with superior biological and physicochemical properties. This review explores the use of nanocomposite biomaterials based on gelatin and various types of nanoparticles together for applications in bone tissue engineering.
Collapse
Affiliation(s)
- Elaheh Salehi Abar
- Department of Prosthodontics, Faculty of Dentistry, Tabriz University of Medical Sciences, Tabriz, Iran; Research Center for Pharmaceutical Nanotechnology, Biomedicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Somayeh Vandghanooni
- Hematology and Oncology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ali Torab
- Department of Prosthodontics, Faculty of Dentistry, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mehdi Jaymand
- Nano Drug Delivery Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran; Student Research Committee, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Morteza Eskandani
- Research Center for Pharmaceutical Nanotechnology, Biomedicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
22
|
Wang Y, Wu P, Wang Y, He H, Huang L. Dendritic mesoporous nanoparticles for the detection, adsorption, and degradation of hazardous substances in the environment: State-of-the-art and future prospects. JOURNAL OF ENVIRONMENTAL MANAGEMENT 2023; 345:118629. [PMID: 37499417 DOI: 10.1016/j.jenvman.2023.118629] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Revised: 07/03/2023] [Accepted: 07/11/2023] [Indexed: 07/29/2023]
Abstract
Equipped with hierarchical pores and three-dimensional (3D) center-radial channels, dendritic mesoporous nanoparticles (DMNs) make their pore volumes extremely large, specific surface areas super-high, internal spaces especially accessible, and so on. Other entities (like organic moieties or nanoparticles) can be modified onto the interfaces or skeletons of DMNs, accomplishing their functionalization for desirable applications. This comprehensive review emphasizes on the design and construction of DMNs-based systems which serve as sensors, adsorbents and catalysts for the detection, adsorption, and degradation of hazardous substances, mainly including the construction procedures of brand-new DMNs-based materials and the involved hazardous substances (like industrial chemicals, chemical dyes, heavy metal ions, medicines, pesticides, and harmful gases). The sensitive, adsorptive, or catalytic performances of various DMNs have been compared; correspondingly, the reaction mechanisms have been revealed strictly. It is honestly anticipated that the profound discussion could offer scientists certain enlightenment to design novel DMNs-based systems towards the detection, adsorption, and degradation of hazardous substances, respectively or comprehensively.
Collapse
Affiliation(s)
- Yabin Wang
- College of Chemistry and Chemical Engineering, Yan'an University, Yan'an, 716000, Shaanxi, PR China; Institute for Triazine Compounds & Hierarchical Porous Materials, Shaanxi, PR China.
| | - Peng Wu
- College of Chemistry and Chemical Engineering, Yan'an University, Yan'an, 716000, Shaanxi, PR China
| | - Yanni Wang
- College of Chemistry and Chemical Engineering, Yan'an University, Yan'an, 716000, Shaanxi, PR China
| | - Hua He
- Institute for Triazine Compounds & Hierarchical Porous Materials, Shaanxi, PR China
| | - Liangzhu Huang
- College of Chemistry and Chemical Engineering, Yan'an University, Yan'an, 716000, Shaanxi, PR China; Institute for Triazine Compounds & Hierarchical Porous Materials, Shaanxi, PR China
| |
Collapse
|
23
|
Cheng D, Zhang J, Fu J, Song H, Yu C. A hierarchical spatial assembly approach of silica-polymer composites leads to versatile silica/carbon nanoparticles. SCIENCE ADVANCES 2023; 9:eadi7502. [PMID: 37792932 PMCID: PMC10550229 DOI: 10.1126/sciadv.adi7502] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Accepted: 08/31/2023] [Indexed: 10/06/2023]
Abstract
Assembly of silica and polymer in the absence of surfactant templates is an emerging strategy to construct intricate nanostructures, whereas the underlying mechanism and structural versatility remain largely unexplored. We report a hierarchical spatial assembly strategy of silica-polymer composites to produce silica and carbon nanoparticles with unprecedented structures. The assembly hierarchy involves a higher length scale asymmetric A-B-A core-shell-type spatial assembly in a composite sphere, and a nanoscale assembly in the middle layer B in which the silica/polymer ratio governs the assembled structures of silica nanodomains. Through an in-depth understanding of the hierarchical spatial assembly mechanism, a series of silica and carbon nanoparticles with intriguing and controllable architectures are obtained that cannot be easily achieved via conventional surfactant-templating approaches. This work opens an avenue toward the designed synthesis of nanoparticles with precisely regulated structures.
Collapse
Affiliation(s)
- Dan Cheng
- 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
| | - Jianye Fu
- College of Chemistry and Chemical Engineering, China University of Petroleum, Qingdao 266555, China
| | - 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
|
24
|
Yang C, Nguyen DD, Lai J. Poly(l-Histidine)-Mediated On-Demand Therapeutic Delivery of Roughened Ceria Nanocages for Treatment of Chemical Eye Injury. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2302174. [PMID: 37430140 PMCID: PMC10502830 DOI: 10.1002/advs.202302174] [Citation(s) in RCA: 87] [Impact Index Per Article: 43.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Revised: 06/10/2023] [Indexed: 07/12/2023]
Abstract
Development of topical bioactive formulations capable of overcoming the low bioavailability of conventional eye drops is critically important for efficient management of ocular chemical burns. Herein, a nanomedicine strategy is presented to harness the surface roughness-controlled ceria nanocages (SRCNs) and poly(l-histidine) surface coatings for triggering multiple bioactive roles of intrinsically therapeutic nanocarriers and promoting transport across corneal epithelial barriers as well as achieving on-demand release of dual drugs [acetylcholine chloride (ACh) and SB431542] at the lesion site. Specifically, the high surface roughness helps improve cellular uptake and therapeutic activity of SRCNs while exerting a negligible impact on good ocular biocompatibility of the nanomaterials. Moreover, the high poly(l-histidine) coating amount can endow the SRCNs with an ≈24-fold enhancement in corneal penetration and an effective smart release of ACh and SB431542 in response to endogenous pH changes caused by tissue injury/inflammation. In a rat model of alkali burn, topical single-dose nanoformulation can efficaciously reduce corneal wound areas (19-fold improvement as compared to a marketed eye drops), attenuate ≈93% abnormal blood vessels, and restore corneal transparency to almost normal at 4 days post-administration, suggesting great promise for designing multifunctional metallic nanotherapeutics for ocular pharmacology and tissue regenerative medicine.
Collapse
Affiliation(s)
- Chia‐Jung Yang
- Department of Biomedical EngineeringChang Gung UniversityTaoyuan33302Taiwan
| | - Duc Dung Nguyen
- Department of Biomedical EngineeringChang Gung UniversityTaoyuan33302Taiwan
| | - Jui‐Yang Lai
- Department of Biomedical EngineeringChang Gung UniversityTaoyuan33302Taiwan
- Department of OphthalmologyChang Gung Memorial Hospital, LinkouTaoyuan33305Taiwan
- Department of Materials EngineeringMing Chi University of TechnologyNew Taipei City24301Taiwan
- Research Center for Chinese Herbal MedicineCollege of Human EcologyChang Gung University of Science and TechnologyTaoyuan33303Taiwan
| |
Collapse
|
25
|
Sun B, Wu W, Narasipura EA, Ma Y, Yu C, Fenton OS, Song H. Engineering nanoparticle toolkits for mRNA delivery. Adv Drug Deliv Rev 2023; 200:115042. [PMID: 37536506 DOI: 10.1016/j.addr.2023.115042] [Citation(s) in RCA: 44] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 07/26/2023] [Accepted: 07/31/2023] [Indexed: 08/05/2023]
Abstract
The concept of using mRNA to produce its own medicine in situ in the body makes it an ideal drug candidate, holding great potential to revolutionize the way we approach medicine. The unique characteristics of mRNA, as well as its customizable biomedical functions, call for the rational design of delivery systems to protect and transport mRNA molecules. In this review, a nanoparticle toolkit is presented for the development of mRNA-based therapeutics from a drug delivery perspective. Nano-delivery systems derived from either natural systems or chemical synthesis, in the nature of organic or inorganic materials, are summarised. Delivery strategies in controlling the tissue targeting and mRNA release, as well as the role of nanoparticles in building and boosting the activity of mRNA drugs, have also been introduced. In the end, our insights into the clinical and translational development of mRNA nano-drugs are presented.
Collapse
Affiliation(s)
- Bing Sun
- Australian Institute for Bioengineering and Nanotechnology, the University of Queensland, Brisbane, QLD 4072, Australia
| | - Weixi Wu
- Australian Institute for Bioengineering and Nanotechnology, the University of Queensland, Brisbane, QLD 4072, Australia
| | - Eshan A Narasipura
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Yutian Ma
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Chengzhong Yu
- Australian Institute for Bioengineering and Nanotechnology, the University of Queensland, Brisbane, QLD 4072, Australia
| | - Owen S Fenton
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA.
| | - Hao Song
- Australian Institute for Bioengineering and Nanotechnology, the University of Queensland, Brisbane, QLD 4072, Australia.
| |
Collapse
|
26
|
Zhou H, He Y, Xiong W, Jing S, Duan X, Huang Z, Nahal GS, Peng Y, Li M, Zhu Y, Ye Q. MSC based gene delivery methods and strategies improve the therapeutic efficacy of neurological diseases. Bioact Mater 2023; 23:409-437. [PMCID: PMC9713256 DOI: 10.1016/j.bioactmat.2022.11.007] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Revised: 11/08/2022] [Accepted: 11/13/2022] [Indexed: 12/05/2022] Open
|
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; 15:1013. [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
|
28
|
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
|
29
|
Zhang J, Kothalawala S, Yu C. Engineered silica nanomaterials in pesticide delivery: Challenges and perspectives. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2023; 320:121045. [PMID: 36639042 DOI: 10.1016/j.envpol.2023.121045] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Revised: 12/04/2022] [Accepted: 01/06/2023] [Indexed: 06/17/2023]
Abstract
Over the past decade, nanopesticide has been developed rapidly for exploring effective and safe alternatives to conventional pesticides with significant drawbacks and risks. Many nanotechnologies, including pesticide nanoemulsions, polymer-based nanopesticides, and metal/metal oxide nanoparticle-based pesticides have emerged and are extensively reviewed. Engineered silica nanomaterials (ESNs) have also shown promising potential as carriers in nanopesticides for modern agriculture. However, there are limited reviews specifically on ESN-based nanopesticides. Herein, we provide a comprehensive review on the recent progress of ESN-based nanopesticide technologies. An introduction of synthetic technology, formation mechanism, and surface engineering technology is firstly presented. Then, the advantages of ESN-based pesticide formulation and their structure-function-relationship are illustrated in detail. Finally, our perspectives on challenges and future research in ESN-based nanopesticide development are discussed.
Collapse
Affiliation(s)
- Jun Zhang
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, Queensland, 4072, Australia
| | - Sukitha Kothalawala
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, Queensland, 4072, Australia
| | - Chengzhong Yu
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, Queensland, 4072, Australia.
| |
Collapse
|
30
|
Sun J, Nie H, Pan P, Jiang Q, Liu C, Wang M, Deng Y, Yan B. Combined Anti-Angiogenic and Anti-Inflammatory Nanoformulation for Effective Treatment of Ocular Vascular Diseases. Int J Nanomedicine 2023; 18:437-453. [PMID: 36718193 PMCID: PMC9884055 DOI: 10.2147/ijn.s387428] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Accepted: 01/16/2023] [Indexed: 01/24/2023] Open
Abstract
Background Ocular vascular diseases are the major causes of visual impairment, which are characterized by retinal vascular dysfunction and robust inflammatory responses. Traditional anti-angiogenic or anti-inflammatory drugs still have limitations due to the short-acting effects. To improve the anti-angiogenic or anti-inflammatory efficiency, a dual-drug nanocomposite formulation was proposed for combined anti-angiogenic and anti-inflammatory treatment of ocular vascular diseases. Methods CBC-MCC@hMSN(SM) complex nanoformulation was prepared by integrating conbercept (CBC, an anti-angiogenic drug) and MCC950 (MCC, an inhibitor of inflammation) into the surface-modified hollow mesoporous silica nanoparticles (hMSN(SM)). CBC-MCC@hMSN(SM) complex nanoformulation was then characterized by Fourier transform infrared spectroscopy, transmission electron microscopy, zeta potentials, and nitrogen adsorption-desorption measurement. CBC and MCC release profile, cytotoxicity, tissue toxicity, anti-angiogenic effects, and anti-inflammatory effects of CBC-MCC@hMSN(SM) were estimated using the in vitro and in vivo experiments. Results CBC-MCC@hMSN(SM) complex had no obvious cytotoxicity and tissue toxicity and did not cause a detectable ocular inflammatory responses. CBC-MCC@hMSN(SM) complex was more effective than free CBC or MCC in suppressing endothelial angiogenic effects and inflammatory responses in vitro. A single intraocular injection of CBC-MCC@hMSN(SM) complex potently suppressed diabetes-induced retinal vascular dysfunction, choroidal neovascularization, and inflammatory responses for up to 6 months. Conclusion Combined CBC and MCC nanoformulation provides a promising strategy for sustained suppression of pathological angiogenesis and inflammatory responses to improve the treatment outcomes of ocular vascular diseases.
Collapse
Affiliation(s)
- Jianguo Sun
- Eye Institute and Department of Ophthalmology, Eye & ENT Hospital, State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai, People’s Republic of China
| | - Huiling Nie
- The Affiliated Eye Hospital and The Fourth School of Clinical Medicine, Nanjing Medical University, Nanjing, People’s Republic of China
| | - Panpan Pan
- Department of Chemistry, Fudan University, Shanghai, People’s Republic of China
| | - Qin Jiang
- The Affiliated Eye Hospital and The Fourth School of Clinical Medicine, Nanjing Medical University, Nanjing, People’s Republic of China
| | - Chang Liu
- Eye Institute and Department of Ophthalmology, Eye & ENT Hospital, State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai, People’s Republic of China
| | - Min Wang
- Eye Institute and Department of Ophthalmology, Eye & ENT Hospital, State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai, People’s Republic of China
| | - Yonghui Deng
- Department of Chemistry, Fudan University, Shanghai, People’s Republic of China,Department of Gastroenterology and Hepatology, Zhongshan Hospital, Institute of Biomedical Sciences, Fudan University, Shanghai, People’s Republic of China,Correspondence: Yonghui Deng; Biao Yan, Email ;
| | - Biao Yan
- Eye Institute and Department of Ophthalmology, Eye & ENT Hospital, State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai, People’s Republic of China
| |
Collapse
|
31
|
Shafi S, Zafar S, Hassan Gilliani MR, Hussain D, Aitani A, Majeed S. Silica-based nanocarriers. BIONANOCATALYSIS : FROM DESIGN TO APPLICATIONS 2023:179-195. [DOI: 10.1016/b978-0-323-91760-5.00010-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
|
32
|
Yang T, Xia L, Li G, Zhao J, Li J, Ge J, Yuan Q, Zhang J, He K, Xia Q. Novel bionic inspired nanosystem construction for precise delivery of mRNA. Front Bioeng Biotechnol 2023; 11:1160509. [PMID: 36937761 PMCID: PMC10018395 DOI: 10.3389/fbioe.2023.1160509] [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: 02/07/2023] [Accepted: 02/20/2023] [Indexed: 03/06/2023] Open
Abstract
The intracellular delivery of messenger (m)RNA holds great potential for the discovery and development of vaccines and therapeutics. Yet, in many applications, a major obstacle to clinical translation of mRNA therapy is the lack of efficient strategy to precisely deliver RNA sequence to liver tissues and cells. In this study, we synthesized virus-like mesoporous silica (V-SiO2) nanoparticles for effectively deliver the therapeutic RNA. Then, the cationic polymer polyethylenimine (PEI) was included for the further silica surface modification (V-SiO2-P). Negatively charged mRNA motifs were successfully linked on the surface of V-SiO2 through electrostatic interactions with PEI (m@V-SiO2-P). Finally, the supported lipid bilayer (LB) was completely wrapped on the bionic inspired surface of the nanoparticles (m@V-SiO2-P/LB). Importantly, we found that, compared with traditional liposomes with mRNA loading (m@LNPs), the V-SiO2-P/LB bionic-like morphology effectively enhanced mRNA delivery effect to hepatocytes both in vitro and in vivo, and PEI modification concurrently promoted mRNA binding and intracellular lysosomal escape. Furthermore, m@V-SiO2-P increased the blood circulation time (t1/2 = 7 h) to be much longer than that of the m@LNPs (4.2 h). Understanding intracellular delivery mediated by the V-SiO2-P/LB nanosystem will inspire the next-generation of highly efficient and effective mRNA therapies. In addition, the nanosystem can also be applied to the oral cavity, forehead, face and other orthotopic injections.
Collapse
Affiliation(s)
- Taihua Yang
- Department of Liver Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Lei Xia
- Department of Liver Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Gen Li
- Department of Orthopedics, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Jie Zhao
- Department of Liver Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Jie Li
- Department of Liver Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Jiahao Ge
- Department of Liver Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Qinggong Yuan
- Department of Gastroenterology, Hepatology and Endocrinology, Hannover Medical School, Hannover, Germany
| | - Jianjun Zhang
- Department of Liver Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- *Correspondence: Jianjun Zhang, ; Kang He, ; Qiang Xia,
| | - Kang He
- Department of Liver Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- *Correspondence: Jianjun Zhang, ; Kang He, ; Qiang Xia,
| | - Qiang Xia
- Department of Liver Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Engineering Research Center of Transplantation and Immunology, Shanghai, China
- Shanghai Institute of Transplantation, Shanghai, China
- *Correspondence: Jianjun Zhang, ; Kang He, ; Qiang Xia,
| |
Collapse
|
33
|
Fu J, Han W, Zhang X, Sun Y, Bhadane R, Wei B, Li L, Yu L, Yang J, Rosenholm JM, Salo-Ahen OMH, Fan T, Zhang B, Swelm W, Al-Ghamdi AA, Xia L, Zhang H, Qiu M, Zhang H, Wang X. Silica Nanoparticles with Virus-Mimetic Spikes Enable Efficient siRNA Delivery In Vitro and In Vivo. RESEARCH (WASHINGTON, D.C.) 2022; 2022:0014. [PMID: 39290973 PMCID: PMC11407519 DOI: 10.34133/research.0014] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Accepted: 11/01/2022] [Indexed: 09/19/2024]
Abstract
Oligonucleotide-based therapy has experienced remarkable development in the past 2 decades, but its broad applications are severely hampered by delivery vectors. Widely used viral vectors and lipid nanovectors are suffering from immune clearance after repeating usage or requiring refrigerated transportation and storage, respectively. In this work, amino-modified virus-mimetic spike silica nanoparticles (NH2-SSNs) were fabricated using a 1-pot surfactant-free approach with controlled spike lengths, which were demonstrated with excellent delivery performance and biosafety in nearly all cell types and mice. It indicated that NH2-SSNs entered cells by spike-dependent cell membrane docking and dynamin-dependent endocytosis. The positively charged spikes with proper length on the surface can facilitate the efficient encapsulation of RNAs, protect the loaded RNAs from degradation, and trigger an early endosome escape during intracellular trafficking, similarly to the cellular internalization mechanism of virions. Regarding the fantastic properties of NH2-SSNs in nucleic acid delivery, it revealed that nanoparticles with solid spikes on the surface would be excellent vehicles for gene therapy, presenting self-evident advantages in storage, transportation, modification, and quality control in large-scale production compared to lipid nanovectors.
Collapse
Affiliation(s)
- Jianye Fu
- Key Laboratory of Marine Drugs, Chinese Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, China
- Collaborative Innovation Center for Optoelectronic Science & Technology, International Collaborative Laboratory of 2D Materials for Optoelectronics Science and Technology of Ministry of Education, Institute of Microscale Optoelectronics, College of Physics and Optoelectronic Engineering, Shenzhen University, Shenzhen 518060, China
- Key Laboratory of Marine Chemistry Theory and Technology (Ocean University of China), Ministry of Education, Qingdao 266100, China
- College of Chemistry and Chemical Engineering, China University of Petroleum, Qingdao 266555, China
| | - Wenwei Han
- Key Laboratory of Marine Drugs, Chinese Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, China
- Key Laboratory of Marine Chemistry Theory and Technology (Ocean University of China), Ministry of Education, Qingdao 266100, China
| | - Xue Zhang
- Key Laboratory of Marine Drugs, Chinese Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, China
| | - Yutong Sun
- Key Laboratory of Marine Drugs, Chinese Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, China
| | - Rajendra Bhadane
- Pharmaceutical Sciences Laboratory, Åbo Akademi University, 20520 Turku, Finland
- Structural Bioinformatics Laboratory, Biochemistry, Åbo Akademi University, 20520 Turku, Finland
| | - Bo Wei
- Key Laboratory of Marine Drugs, Chinese Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, China
| | - Li Li
- Key Laboratory of Marine Drugs, Chinese Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, China
- Center for Innovation Marine Drug Screening & Evaluation, Pilot National Laboratory for Marine Science and Technology (Qingdao), Qingdao 266237, China
- Marine Biomedical Research Institute of Qingdao, Qingdao 266100, China
| | - Liangmin Yu
- Key Laboratory of Marine Chemistry Theory and Technology (Ocean University of China), Ministry of Education, Qingdao 266100, China
| | - Jinbo Yang
- Key Laboratory of Marine Drugs, Chinese Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, China
- Center for Innovation Marine Drug Screening & Evaluation, Pilot National Laboratory for Marine Science and Technology (Qingdao), Qingdao 266237, China
- Marine Biomedical Research Institute of Qingdao, Qingdao 266100, China
| | - Jessica M Rosenholm
- Pharmaceutical Sciences Laboratory, Åbo Akademi University, 20520 Turku, Finland
| | - Outi M H Salo-Ahen
- Pharmaceutical Sciences Laboratory, Åbo Akademi University, 20520 Turku, Finland
- Structural Bioinformatics Laboratory, Biochemistry, Åbo Akademi University, 20520 Turku, Finland
| | - Taojian Fan
- Collaborative Innovation Center for Optoelectronic Science & Technology, International Collaborative Laboratory of 2D Materials for Optoelectronics Science and Technology of Ministry of Education, Institute of Microscale Optoelectronics, College of Physics and Optoelectronic Engineering, Shenzhen University, Shenzhen 518060, China
| | - Bin Zhang
- Collaborative Innovation Center for Optoelectronic Science & Technology, International Collaborative Laboratory of 2D Materials for Optoelectronics Science and Technology of Ministry of Education, Institute of Microscale Optoelectronics, College of Physics and Optoelectronic Engineering, Shenzhen University, Shenzhen 518060, China
| | - Wageh Swelm
- Department of Physics, Faculty of Science, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Ahmed A Al-Ghamdi
- Department of Physics, Faculty of Science, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Lin Xia
- Hangzhou No. 14 High School, Hangzhou 310000, China
| | - Han Zhang
- Collaborative Innovation Center for Optoelectronic Science & Technology, International Collaborative Laboratory of 2D Materials for Optoelectronics Science and Technology of Ministry of Education, Institute of Microscale Optoelectronics, College of Physics and Optoelectronic Engineering, Shenzhen University, Shenzhen 518060, China
| | - Meng Qiu
- Key Laboratory of Marine Chemistry Theory and Technology (Ocean University of China), Ministry of Education, Qingdao 266100, China
| | - Hongbo Zhang
- Pharmaceutical Sciences Laboratory, Åbo Akademi University, 20520 Turku, Finland
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, 20520 Turku, Finland
| | - Xin Wang
- Key Laboratory of Marine Drugs, Chinese Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, China
- Center for Innovation Marine Drug Screening & Evaluation, Pilot National Laboratory for Marine Science and Technology (Qingdao), Qingdao 266237, China
- Marine Biomedical Research Institute of Qingdao, Qingdao 266100, China
| |
Collapse
|
34
|
An effective VEGF-siRNA delivery via folic acid decorated and pegylated silica nanoparticles. J Drug Deliv Sci Technol 2022. [DOI: 10.1016/j.jddst.2022.103828] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
|
35
|
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
|
36
|
Hinchliffe BA, Turner P, J H Cant D, De Santis E, Aggarwal P, Harris R, Templeton D, Shard AG, Hodnett M, Minelli C. Deagglomeration of DNA nanomedicine carriers using controlled ultrasonication. ULTRASONICS SONOCHEMISTRY 2022; 89:106141. [PMID: 36067646 PMCID: PMC9463456 DOI: 10.1016/j.ultsonch.2022.106141] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Revised: 08/17/2022] [Accepted: 08/22/2022] [Indexed: 06/15/2023]
Abstract
Control over the agglomeration state of manufactured particle systems for drug and oligonucleotide intracellular delivery is paramount to ensure reproducible and scalable therapeutic efficacy. Ultrasonication is a well-established mechanism for the deagglomeration of bulk powders in dispersion. Its use in manufacturing requires strict control of the uniformity and reproducibility of the cavitation field within the sample volume to minimise within-batch and batch-to-batch variability. In this work, we demonstrate the use of a reference cavitating vessel which provides stable and reproducible cavitation fields over litre-scale volumes to assist the controlled deagglomeration of a novel non-viral particle-based plasmid delivery system. The system is the Nuvec delivery platform, comprising polyethylenimine-coated spiky silica particles with diameters of ∼ 200 nm. We evaluated the use of controlled cavitation at different input powers and stages of preparation, for example before and after plasmid loading. Plasmid loading was confirmed by X-ray photoelectron spectroscopy and gel electrophoresis. The latter was also used to assess plasmid integrity and the ability of the particles to protect plasmid from potential degradation caused by the deagglomeration process. We show the utility of laser diffraction and differential centrifugal sedimentation in quantifying the efficacy of product de-agglomeration in the microscale and nanoscale size range respectively. Transmission electron microscopy was used to assess potential damages to the silica particle structure due to the sonication process.
Collapse
Affiliation(s)
| | - Piers Turner
- National Physical Laboratory, Hampton Road, Teddington SW11 0LW, UK
| | - David J H Cant
- National Physical Laboratory, Hampton Road, Teddington SW11 0LW, UK
| | | | - Purnank Aggarwal
- National Physical Laboratory, Hampton Road, Teddington SW11 0LW, UK
| | - Rob Harris
- N4 Pharma, Weston House, Bradgate Park View, Chellaston DE73 5UJ, UK
| | - David Templeton
- N4 Pharma, Weston House, Bradgate Park View, Chellaston DE73 5UJ, UK
| | - Alex G Shard
- National Physical Laboratory, Hampton Road, Teddington SW11 0LW, UK
| | - Mark Hodnett
- National Physical Laboratory, Hampton Road, Teddington SW11 0LW, UK
| | - Caterina Minelli
- National Physical Laboratory, Hampton Road, Teddington SW11 0LW, UK.
| |
Collapse
|
37
|
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
|
38
|
Chen W, Yu B, Zhang X, Zhang F, Zan X, Li T. Precisely controlling the surface roughness of silica nanoparticles for enhanced functionalities and applications. J Colloid Interface Sci 2022; 629:173-181. [DOI: 10.1016/j.jcis.2022.08.159] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Revised: 07/31/2022] [Accepted: 08/25/2022] [Indexed: 10/14/2022]
|
39
|
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
|
40
|
Sasso J, Ambrose BJB, Tenchov R, Datta RS, Basel MT, DeLong RK, Zhou QA. The Progress and Promise of RNA Medicine─An Arsenal of Targeted Treatments. J Med Chem 2022; 65:6975-7015. [PMID: 35533054 PMCID: PMC9115888 DOI: 10.1021/acs.jmedchem.2c00024] [Citation(s) in RCA: 50] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2022] [Indexed: 02/08/2023]
Abstract
In the past decade, there has been a shift in research, clinical development, and commercial activity to exploit the many physiological roles of RNA for use in medicine. With the rapid success in the development of lipid-RNA nanoparticles for mRNA vaccines against COVID-19 and with several approved RNA-based drugs, RNA has catapulted to the forefront of drug research. With diverse functions beyond the role of mRNA in producing antigens or therapeutic proteins, many classes of RNA serve regulatory roles in cells and tissues. These RNAs have potential as new therapeutics, with RNA itself serving as either a drug or a target. Here, based on the CAS Content Collection, we provide a landscape view of the current state and outline trends in RNA research in medicine across time, geography, therapeutic pipelines, chemical modifications, and delivery mechanisms.
Collapse
Affiliation(s)
- Janet
M. Sasso
- CAS,
a division of the American Chemical Society 2540 Olentangy River Road, Columbus, Ohio 43202, United States
| | - Barbara J. B. Ambrose
- CAS,
a division of the American Chemical Society 2540 Olentangy River Road, Columbus, Ohio 43202, United States
| | - Rumiana Tenchov
- CAS,
a division of the American Chemical Society 2540 Olentangy River Road, Columbus, Ohio 43202, United States
| | - Ruchira S. Datta
- CAS,
a division of the American Chemical Society 2540 Olentangy River Road, Columbus, Ohio 43202, United States
| | - Matthew T. Basel
- College
of Veterinary Medicine, Kansas State University, Manhattan, Kansas 66506, United States
| | - Robert K. DeLong
- Nanotechnology
Innovation Center Kansas State, Kansas State
University, Manhattan, Kansas 66506, United States
| | - Qiongqiong Angela Zhou
- CAS,
a division of the American Chemical Society 2540 Olentangy River Road, Columbus, Ohio 43202, United States
| |
Collapse
|
41
|
Topography-Mediated Enhancement of Nonviral Gene Delivery in Stem Cells. Pharmaceutics 2022; 14:pharmaceutics14051096. [PMID: 35631682 PMCID: PMC9142897 DOI: 10.3390/pharmaceutics14051096] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Revised: 05/09/2022] [Accepted: 05/18/2022] [Indexed: 02/07/2023] Open
Abstract
Gene delivery holds great promise for bioengineering, biomedical applications, biosensors, diagnoses, and gene therapy. In particular, the influence of topography on gene delivery is considered to be an attractive approach due to low toxicity and localized delivery properties. Even though many gene vectors and transfection systems have been developed to enhance transfection potential and combining it with other forms of stimulations could even further enhance it. Topography is an interesting surface property that has been shown to stimulate differentiation, migration, cell morphology, and cell mechanics. Therefore, it is envisioned that topography might also be able to stimulate transfection. In this study, we tested the hypothesis “topography is able to regulate transfection efficiency”, for which we used nano- and microwave-like topographical substrates with wavelengths ranging from 500 nm to 25 µm and assessed the transfectability of human bone marrow-derived mesenchymal stem cells (hBM-MSCs) and myoblasts. For transfection, Lipofectamine 2000 and a gene encoding plasmid for red-fluorescent protein (m-Cherry) were used and topography-induced cell morphology and transfection efficiency was analyzed. As a result, topography directs cell spreading, elongation, and proliferation as well as the transfection efficiency, which were investigated but were found not to be correlated and dependent on the cell type. A 55% percent improvement of transfection efficiency was identified for hBM-MSCs grown on 2 µm wrinkles (24.3%) as compared to hBM-MSCs cultured on flat controls (15.7%). For myoblast cells, the highest gene-expression efficiency (46.1%) was observed on the 10 µm topography, which enhanced the transfection efficiency by 64% as compared to the flat control (28.1%). From a qualitative assessment, it was observed that the uptake capacity of cationic complexes of TAMRA-labeled oligodeoxynucleotides (ODNs) was not topography-dependent but that the intracellular release was faster, as indicated by the positively stained nuclei on 2 μm for hBM-MSCs and 10 μm for myoblasts. The presented results indicate that topography enhances the gene-delivery capacity and that the responses are dependent on cell type. This study demonstrates the important role of topography on cell stimulation for gene delivery as well as understanding the uptake capacity of lipoplexes and may be useful for developing advanced nonviral gene delivery strategies.
Collapse
|
42
|
Ni C, Zhong Y, Wu W, Song Y, Makvandi P, Yu C, Song H. Co-Delivery of Nano-Silver and Vancomycin via Silica Nanopollens for Enhanced Antibacterial Functions. Antibiotics (Basel) 2022; 11:antibiotics11050685. [PMID: 35625329 PMCID: PMC9137463 DOI: 10.3390/antibiotics11050685] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2022] [Revised: 05/14/2022] [Accepted: 05/17/2022] [Indexed: 12/13/2022] Open
Abstract
Infectious diseases caused by bacteria have led to a great threat to public health. With the significant advances in nanotechnology in recent decades, nanomaterials have emerged as a powerful tool to boost antibacterial performance due to either intrinsic bactericidal properties or by enhancing the delivery efficiency of antibiotics for effective pathogen killing. Vancomycin, as one of the most widely employed antimicrobial peptides, has a potent bactericidal activity, but at the same time shows a limited bioavailability. Silver nanoparticles have also been extensively explored and were found to have a well-recognized antibacterial activity and limited resistance potential; however, how to prevent nanosized Ag particles from aggregation in biological conditions is challenging. In this study, we aimed to combine the advantages of both vancomycin and nano-Ag for enhanced bacterial killing, where both antibacterial agents were successfully loaded onto a silica nanoparticle with a pollen-like morphology. The morphology of nano-Ag-decorated silica nanopollens was characterized using transmission electron microscopy and elemental mapping through energy dispersive spectroscopy. Silver nanoparticles with a size of 10–25 nm were observed as well-distributed on the surface of silica nanoparticles of around 200 nm. The unique design of a spiky morphology of silica nano-carriers promoted the adhesion of nanoparticles towards bacterial surfaces to promote localized drug release for bacterial killing, where the bacterial damage was visualized through scanning electron microscopy. Enhanced bactericidal activity was demonstrated through this co-delivery of vancomycin and nano-Ag, decreasing the minimum inhibition concentration (MIC) towards E. coli and S. epidermidis down to 15 and 10 µg/mL. This study provides an efficient antimicrobial nano-strategy to address potential bacterial infections.
Collapse
Affiliation(s)
- Chengang Ni
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane 4072, Australia; (C.N.); (Y.Z.); (W.W.); (Y.S.); (C.Y.)
| | - Yuening Zhong
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane 4072, Australia; (C.N.); (Y.Z.); (W.W.); (Y.S.); (C.Y.)
| | - Weixi Wu
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane 4072, Australia; (C.N.); (Y.Z.); (W.W.); (Y.S.); (C.Y.)
| | - Yaping Song
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane 4072, Australia; (C.N.); (Y.Z.); (W.W.); (Y.S.); (C.Y.)
| | - Pooyan Makvandi
- Center for Materials Interfaces, Istituto Italiano di Tecnologia, Pontedera, 56025 Pisa, Italy;
| | - Chengzhong Yu
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane 4072, Australia; (C.N.); (Y.Z.); (W.W.); (Y.S.); (C.Y.)
| | - Hao Song
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane 4072, Australia; (C.N.); (Y.Z.); (W.W.); (Y.S.); (C.Y.)
- Correspondence:
| |
Collapse
|
43
|
Song G, Lv F, Huang Y, Bai H, Wang S. Conjugated Polymers for Gene Delivery and Photothermal Gene Expression. Chempluschem 2022; 87:e202200073. [DOI: 10.1002/cplu.202200073] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Revised: 04/26/2022] [Indexed: 11/09/2022]
Affiliation(s)
- Gang Song
- Institute of Chemistry CAS: Institute of Chemistry Chinese Academy of Sciences Organic Solids CHINA
| | - Fengting Lv
- Institute of Chemistry Chinese Academy of Sciences Zhongguancun North First Street 2 CHINA
| | - Yiming Huang
- Institute of Chemistry CAS: Institute of Chemistry Chinese Academy of Sciences Organic Solids CHINA
| | - Haotian Bai
- Institute of Chemistry CAS: Institute of Chemistry Chinese Academy of Sciences Organic Solids CHINA
| | - Shu Wang
- Institute of Chemistry CAS: Institute of Chemistry Chinese Academy of Sciences Organic Solids CHINA
| |
Collapse
|
44
|
Defining Endocytic Pathways of Fucoidan-Coated PIBCA Nanoparticles from the Design of their Surface Architecture. Pharm Res 2022; 39:1135-1150. [PMID: 35233729 PMCID: PMC8887940 DOI: 10.1007/s11095-022-03202-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Accepted: 02/13/2022] [Indexed: 12/02/2022]
Abstract
Purpose This work investigated the endocytic pathways taken by poly(isobutylcyanoacrylate) (PIBCA) nanoparticles differing in their surface composition and architecture, assuming that this might determine their efficiency of intracellular drug delivery. Methods Nanoparticles (A0, A25, A100, R0, R25 ) were prepared by anionic or redox radical emulsion polymerization using mixtures of dextran and fucoidan (0, 25, 100 % in fucoidan). Cell uptake was evaluated by incubating J774A.1 macrophages with nanoparticles. Endocytic pathways were studied by incubating cells with endocytic pathway inhibitors (chlorpromazine, genistein, cytochalasin D, methyl-ß-cyclodextrin and nocodazole) and nanoparticle uptake was evaluated by flow cytometry and confocal microscopy. Results The fucoidan-coated PIBCA nanoparticles A25 were internalized 3-fold more efficiently than R25 due to the different architecture of the fucoidan chains presented on the surface. Different fucoidan density and architecture led to different internalization pathway preferred by the cells. Large A100 nanoparticles with surface was covered with fucoidan chains in a loop and train configuration were internalized the most efficiently, 47-fold compared with A0, and 3-fold compared with R0 and R25 through non-endocytic energy-independent pathways and reached the cell cytoplasm. Conclusion Internalization pathways of PIBCA nanoparticles by J774A.1 macrophages could be determined by nanoparticle fucoidan surface composition and architecture. In turn, this influenced the extent of internalization and localization of accumulated nanoparticles within cells. The results are of interest for rationalizing the design of nanoparticles for potential cytoplamic drug delivery by controlling the nature of the nanoparticle surface. Graphical abstract ![]()
Supplementary Information The online version contains supplementary material available at 10.1007/s11095-022-03202-4.
Collapse
|
45
|
|
46
|
Xu C, Lei C, Wang Y, Yu C. Dendritic Mesoporous Nanoparticles: Structure, Synthesis and Properties. Angew Chem Int Ed Engl 2022. [DOI: 10.1002/ange.202112752] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Affiliation(s)
- Chun Xu
- School of Dentistry The University of Queensland Brisbane Queensland 4066 Australia
| | - Chang Lei
- 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
| | - Chengzhong Yu
- Australian Institute for Bioengineering and Nanotechnology The University of Queensland Brisbane QLD 4072 Australia
- School of Chemistry and Molecular Engineering East China Normal University Shanghai 200241 P. R. China
| |
Collapse
|
47
|
van Hees M, Slott S, Hansen AH, Kim HS, Ji HP, Astakhova K. New approaches to moderate CRISPR-Cas9 activity: Addressing issues of cellular uptake and endosomal escape. Mol Ther 2022; 30:32-46. [PMID: 34091053 PMCID: PMC8753288 DOI: 10.1016/j.ymthe.2021.06.003] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2020] [Revised: 03/15/2021] [Accepted: 05/27/2021] [Indexed: 02/09/2023] Open
Abstract
CRISPR-Cas9 is rapidly entering molecular biology and biomedicine as a promising gene-editing tool. A unique feature of CRISPR-Cas9 is a single-guide RNA directing a Cas9 nuclease toward its genomic target. Herein, we highlight new approaches for improving cellular uptake and endosomal escape of CRISPR-Cas9. As opposed to other recently published works, this review is focused on non-viral carriers as a means to facilitate the cellular uptake of CRISPR-Cas9 through endocytosis. The majority of non-viral carriers, such as gold nanoparticles, polymer nanoparticles, lipid nanoparticles, and nanoscale zeolitic imidazole frameworks, is developed with a focus toward optimizing the endosomal escape of CRISPR-Cas9 by taking advantage of the acidic environment in the late endosomes. Among the most broadly used methods for in vitro and ex vivo ribonucleotide protein transfection are electroporation and microinjection. Thus, other delivery formats are warranted for in vivo delivery of CRISPR-Cas9. Herein, we specifically revise the use of peptide and nanoparticle-based systems as platforms for CRISPR-Cas9 delivery in vivo. Finally, we highlight future perspectives of the CRISPR-Cas9 gene-editing tool and the prospects of using non-viral vectors to improve its bioavailability and therapeutic potential.
Collapse
Affiliation(s)
- Maja van Hees
- Department of Chemistry, Technical University of Denmark, 2800 Lyngby, Denmark
| | - Sofie Slott
- Department of Chemistry, Technical University of Denmark, 2800 Lyngby, Denmark
| | | | - Heon Seok Kim
- School of Medicine, Stanford University, Stanford, CA 94350, USA
| | - Hanlee P. Ji
- School of Medicine, Stanford University, Stanford, CA 94350, USA
| | - Kira Astakhova
- Department of Chemistry, Technical University of Denmark, 2800 Lyngby, Denmark,Corresponding author: Kira Astakhova, Department of Chemistry, Technical University of Denmark, 2800 Lyngby, Denmark.
| |
Collapse
|
48
|
Fan Z, Zhuang C, Wang S, Zhang Y. Photodynamic and Photothermal Therapy of Hepatocellular Carcinoma. Front Oncol 2021; 11:787780. [PMID: 34950591 PMCID: PMC8688153 DOI: 10.3389/fonc.2021.787780] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Accepted: 11/22/2021] [Indexed: 01/10/2023] Open
Abstract
Hepatocellular carcinoma (HCC) is the most common primary liver tumor. It is ranked the sixth most common neoplasm and the third most common cause of cancer mortality. At present, the most common treatment for HCC is surgery, but the 5-year recurrence rates are still high. Patients with early stage HCC with few nodules can be treated with resection or radiofrequency ablation (RFA); while for multinodular HCC, transarterial chemoembolization (TACE) has been the first-line treatment. In recent years, based on medical engineering cooperation, nanotechnology has been increasingly applied to the treatment of cancer. Photodynamic therapy and photothermal therapy are effective for cancer. This paper summarizes the latest progress of photodynamic therapy and photothermal therapy for HCC, with the aim of providing new ideas for the treatment of HCC.
Collapse
Affiliation(s)
- Zhe Fan
- Department of General Surgery, the Third People’s Hospital of Dalian, Dalian Medical University, Dalian, China
- Department of Central Laboratory, the Third People’s Hospital of Dalian, Dalian Medical University, Dalian, China
| | - Chengjun Zhuang
- Department of Critical Care Medicine, The Second Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Shuang Wang
- Department of Endocrinology, The Second Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Yewei Zhang
- Department of Hepatobiliary and Pancreatic Surgery, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, China
| |
Collapse
|
49
|
Xu C, Lei C, Wang Y, Yu C. Dendritic Mesoporous Nanoparticles: Structure, Synthesis and Properties. Angew Chem Int Ed Engl 2021; 61:e202112752. [PMID: 34837444 DOI: 10.1002/anie.202112752] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2021] [Indexed: 11/10/2022]
Abstract
Recently, a new family of "dendritic" mesoporous silica nanoparticles has attracted great interest with widespread applications. Despite a large number of publications (>800), the terminology of "dendritic" is ambiguous. Understanding what possible "dendritic structures" are, their formation mechanisms and the underlying structure-property relationship is fundamentally important. With the advance of characterization techniques such as electron tomography, two types of tree branch-like and flower-like structures can be distinguished, both described as "dendritic" in literature. In this review, we start with the definition of "dendritic", then provide critical analysis of reported dendritic silica nanoparticles according to their structural classification. We also update the understandings of the formation mechanisms of two types of "dendritic" nanoparticles, with a focus on how to control different structural parameters. Various applications of dendritic mesoporous nanoparticles are also reviewed with a focus in biomedical field, providing new insights into the structure-property relationship in this family of nanomaterials.
Collapse
Affiliation(s)
- Chun Xu
- The University of Queensland, School of Dentistry, AUSTRALIA
| | - Chang Lei
- The University of Queensland - Saint Lucia Campus: The University of Queensland, AIBN, AUSTRALIA
| | - Yue Wang
- The University of Queensland, AIBN, AUSTRALIA
| | - Chengzhong Yu
- Australian Institute for Bioengineering and Nanotechnology, the University of Queensland, Australian Institute for Bioengineering and Nanotechnology, the University of Queensland, Building 75,Cnr College Rd & Cooper Rd, 4067, Brisbane, AUSTRALIA
| |
Collapse
|
50
|
Lin X, Wu W, Fu J, Yang Y, Guo B, Yu C, Song H. Asymmetric Silica Nanoparticles with Tailored Spiky Coverage Derived from Silica-Polymer Cooperative Assembly for Enhanced Hemocompatibility and Gene Delivery. ACS APPLIED MATERIALS & INTERFACES 2021; 13:50695-50704. [PMID: 34664946 DOI: 10.1021/acsami.1c13517] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Asymmetric mesoporous silica nanoparticles (AMSNs) with one side featuring a spiky nanotopography, while the other side is smooth and solid, were synthesized via an ethylenediamine (EDA)-directed silica-polymer cooperative assembly approach. By simply varying the EDA amount (x), AMSNs-x samples with adjustable spiky surface coverage were obtained. It is demonstrated that a spiky coverage higher than 50% improved the hemocompatibility of AMSN-x, possibly due to the reduced contact area of the smooth side exposed to the red blood cell (RBC) membrane. Moreover, AMSNs-175 and AMSNs-200 with high spiky coverage enhanced their plasmid DNA (pDNA) loading and binding capability, as well as cellular uptake into HEK-293T cells, thus resulting in high transfection performance. The good hemocompatibility and high performance in pDNA delivery of AMSNs-x with high spiky coverage allow them to serve as promising nonviral vectors for potential applications in gene therapies and DNA vaccines.
Collapse
Affiliation(s)
- Xiuzhen Lin
- School of Materials Science and Engineering, Dongguan University of Technology, Dongguan, Guangdong 523808, China
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Weixi Wu
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Jianye Fu
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Yannan Yang
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Bing Guo
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, Queensland 4072, Australia
- College of Chemistry and Environmental Engineering, Dongguan University of Technology, Dongguan, Guangdong 523808, China
| | - Chengzhong Yu
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Hao Song
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, Queensland 4072, Australia
| |
Collapse
|